library(tidyverse)
library(knitr)

Metabolism

L1: Intro to Metabolism


1. Appreciate how metabolism is made up of many interconnected biochemical reactions

Content Source: Slides 4, 7, 17, 18, 22, 29

Expanded Explanation:

  • Metabolism refers to the entire network of biochemical reactions in a cell, which can be broadly categorized into catabolism (breaking down molecules to extract energy) and anabolism (building complex molecules from simpler ones).

  • These reactions are tightly interconnected; for example, carbohydrates, fats, and proteins can all be funneled into central metabolic pathways like glycolysis, β-oxidation, and the Krebs cycle.

  • Fuels like glucose and fatty acids are broken into 2-carbon units (acetyl-CoA), which enter the Krebs cycle for further oxidation and ATP production.

  • The intermediates in these pathways are shared, making them crucial metabolic “crossroads.”

🔗 Interactive biochemical map: http://biochemical-pathways.com/#/map/1


2. Revise how enzymes catalyse chemical reactions

Content Source: Slides 8, 11–13

Expanded Explanation:

  • Enzymes are biological catalysts that lower the activation energy required for a reaction, thereby increasing the rate.

  • Enzyme types:

    • Kinases: catalyze phosphorylation (add phosphate, often from ATP)

    • Phosphatases: remove phosphate groups

    • Phosphorylases: uses phosphate grpup to break things apart

    • Dehydrogenases: perform oxidation-reduction reactions using NAD⁺ or FAD

      • Example:

\[{pyruvate + NADH ⇌ lactate + NAD^+}\]


NADH is the reduced form of NAD+

NADH oxidise (lose e-) → NAD+

NAD+ reduced (gain e-) → NADH

opposite for other substrate in equation:

If NADH oxidise → pyruvate reduced to lactate

If NAD+ reduce → lactate oxidise to pyruvate

  • Enzymes often work via induced fit mechanisms and may require cofactors like metals or coenzymes (e.g., NAD⁺, FAD).

📷 Diagram of enzyme action: Wikimedia Commons - Enzyme Mechanism


3. Understand some common ways in which enzymes (and metabolic pathways) are regulated

Content Source: Slides 10, 32, 33

Expanded Explanation:

  • Regulation ensures metabolic homeostasis and efficiency, adapting metabolism to energy demand.

  • Major mechanisms:

    • Allosteric regulation: Binding of regulators at non-active sites (e.g., ATP, ADP levels).

    • Covalent modification: Phosphorylation/dephosphorylation.

    • Transcriptional control: Regulation of enzyme synthesis.

  • Energy sensors like AMP-activated protein kinase (AMPK) respond to cellular energy status.

🧪 Real-world link: How AMPK regulates metabolism (Nature Reviews)


4. Appreciate the role played by the hydrogen/electron carriers in catabolism

Content Source: Slides 14, 15, 19–21

Expanded Explanation:

  • In catabolism, electrons (as hydrogen atoms, H/e⁻) are stripped from fuels and transferred to carriers:

    • NAD⁺ → NADH: oxidizes alcohol groups

    • FAD → FADH₂: oxidizes alkyl chains (C-C double bond)

  • These reduced cofactors then feed electrons into the Electron Transport Chain (ETC), driving proton pumping to generate a proton gradient for ATP production.

📘 Supplementary visual and explanation:

NAD⁺ and FAD in metabolism – YouTube (5 min)


5. Recall the basic chemistry of carbohydrates, lipids, and proteins

Content Source: Slides 24–28

Expanded Explanation:

  • Carbohydrates:

    • Basic unit: monosaccharides (e.g., glucose).

    • Hydrophilic; stored as glycogen (300g).

    • Broken down by glycolysis to pyruvate.

    • Energy yield: ~16 kJ/g (wet weight).

  • Lipids:

    • Basic unit: fatty acids.

    • Hydrophobic; stored as triglycerides (many kg).

    • Oxidized via β-oxidation to acetyl-CoA.

    • Energy-dense: ~37 kJ/g.

  • Proteins:

    • Made from amino acids.

    • Catabolized when needed, funnel into various points (pyruvate, acetyl-CoA).

    • No true “storage” – breakdown means losing function.

    • Energy: ~17 kJ/g.

📷 Image for basic biomolecule structures: Biomolecule Basics - Wikimedia


6. Understand the separate stages of fuel oxidation, electron transport, and ATP synthesis

Content Source: Slides 17–23, 30–31

Expanded Explanation:

  1. Fuel Oxidation (Stage 1)

    • Fuels → acetyl-CoA via glycolysis, β-oxidation, or amino acid breakdown.

    • H/e⁻ stripped by NAD⁺/FAD.

  2. Krebs Cycle (Stage 2)

    • Acetyl-CoA + oxaloacetate → CO₂ + NADH + FADH₂

    • 2-carbon chunks don’t just float around, they are always carried on a CoA → forming acetyl-CoA

    • No direct ATP, but reduced carriers are made.

  3. Electron Transport Chain (Stage 3)

    • NADH/FADH₂ donate electrons to ETC (inner mitochondrial membrane).

    • H⁺ pumped into intermembrane space → proton gradient.

    • movement of H/e- down the chain provides energy to pump out protons towards cytoplasm

    • ATP Synthase uses H⁺ flow to convert ADP + Pi → ATP.



THE 7 BIG CONCEPTS:

  1. The H/e- carriers are in short supply

  2. ADP is in short supply

  1. If ATP does not convert to ADP, there won’t be any available for ATP synthesis
  1. ATP is really stable
  1. so the only way it breaks down is if we do work
  1. The inner mitochondrial membrane is impermeable to protons
  1. Hence the “gate” which is the ATP synthesis core
  1. Protons only flow into the matrix if the ATP is being made
  1. Why? Proton gradient is created when H+ pumped out of matrix into intermembrane space. Protons can re-enter the matrix via a channel of ATP synthase. When the H+ re-enter the subunit rotates and spins which converts ADP + Pi to ATP. So no ATP production = No rotation = No proton flow

  2. So, if ATP synthase inhibited → proton gradient builds up → makes it harder for ETC to pump protons out to cytoplasm → ETC slows/stops

  1. The proton pumps don’t work if the proton gradient is very high

  2. No proton pumping, no H/e- movement down ETC

🌀 Animation of ATP Synthase in action: Harvard’s Molecular Biology Visual – ATP Synthase


L2: Fuel Selection During Exercise


1. Outline the effect of muscle glucose utilisation on blood glucose levels and hormonal responses

Content Source: Slides 14–16

Explanation:

  • During exercise, muscle cells increase glucose uptake via GLUT-4 transporters, reducing blood glucose levels.

  • Even gentle activity triggers these transporters to move to the cell surface.

  • Since glucose is essential for brain function, the body must maintain blood glucose at ~5 mM.

  • Hormonal response:

    • Insulin levels fall: Less stimulation of glucose uptake by liver and adipose tissue.

    • Glucagon levels rise: Stimulates glycogen breakdown in liver and lipolysis in adipose tissue to release glucose and fatty acids respectively.

    • These actions together stabilise blood glucose levels despite muscle usage.

Summary Table:

Hormone Response to Exercise / Muscle Uptake Effect
Insulin ↓ Suppressed by sympathetic activation Prevents further glucose uptake by liver/fat
Glucagon ↑ Stimulated by low glucose and catecholamines Increases hepatic glucose production
Adrenaline/Noradrenaline ↑ Sympathetic response Stimulates glycogenolysis (liver + muscle), lipolysis
Cortisol ↑ (longer term) Supports gluconeogenesis, protein catabolism Increases substrate availability

📘 More on hormonal regulation of glucose: Video: Hormonal Regulation of Blood Glucose (Armando Hasudungan)


2. Predict the effect of low insulin and high glucagon on target tissues and blood fuel levels

Content Source: Slide 16

Explanation:

First, What Are Insulin and Glucagon?

Insulin is like a storage hormone. It tells your body to put away fuel (like glucose and fat) when you’ve eaten.

Glucagon is like a release hormone. It tells your body to get fuel out of storage when you’re fasting or exercising

Now Imagine You Haven’t Eaten for a While

Your insulin levels go down, and your glucagon levels go up. Here’s what happens:

  • In the Muscle:

    • Muscles can’t take in glucose easily without insulin.

    • So they don’t get much glucose from the blood.

    *Instead, they start burning fat for energy.

  • In the Liver:

    • Glucagon tells the liver to:

      • Break down glycogen (glycogenolysis) → glucose

      • Make new glucose (gluconeogenesis)

      • Send that glucose into the blood

    So the liver saves the brain by making and releasing glucose.

  • In Fat (Adipose Tissue):

    • Low insulin + high glucagon = fat is broken down

    • This releases fatty acids into the blood

    • Muscles and other organs burn the fat instead of glucose

  • In Blood:

Fuel What Happens
Glucose Stays steady or may go up (from liver)
Fatty acids Go way up (from fat breakdown)
Ketones Go up later (especially during starvation)
  • Low insulin:

    • ↓ Glucose uptake in adipose/muscle (except contracting muscle).

    • ↓ Glycogen synthesis.

  • High glucagon:

    • ↑ Glycogenolysis in liver → ↑ blood glucose.

    • ↑ Lipolysis in adipose tissue → ↑ fatty acids.

  • Result: Blood fuel shifts from glucose to fatty acids for most tissues (glucose is conserved for brain).

🧬 Visual guide to hormone effects on metabolism: Glucagon vs Insulin infographic (Kenhub)


3. Explain why glucose needs to be recycled and describe how this is achieved

Content Source: Slide 17, 20

Explanation:

  • Glucose stores (glycogen) are limited; brain requires steady glucose supply.

  • Fatty acids cannot be converted into glucose (no net gluconeogenesis from acetyl-CoA).

  • So glucose must be:

    • Recycled via the Cori cycle: Muscle produces lactate → liver converts it to glucose via gluconeogenesis.

    • Spared: Use of fatty acids for ATP spares glucose for essential tissues.

  • This prevents rapid depletion of glucose reserves and avoids hypoglycemia.

📘 Cori Cycle Visual Explanation (Osmosis)


4. Recognise what catabolic pathways change as fatty acids become available during low intensity exercise

Content Source: Slides 12, 14, 20

Explanation:

  • In low-intensity exercise:

    • Initial energy comes from glucose (glycolysis).

    • After minutes, fatty acids (via β-oxidation) become the main fuel.

  • The increase in ADP activates oxidative phosphorylation.

  • Fatty acids are mobilised from white adipose tissue and oxidised in mitochondria to acetyl-CoA → Krebs → ETC.

🧪 Fatty Acid β-Oxidation Pathway – Video


5. Describe the general pattern of fuel oxidation in gentle exercise

Content Source: Slides 12, 14, 20

Explanation:

  • ATP demand increases slightly → ↑ ADP → stimulates ATP synthase and ETC.

  • Initial reliance on blood glucose, but transitions to:

    • Fatty acid oxidation (dominant fuel after ~10–15 min).
    • Glycolysis slows, pyruvate is converted to lactate → goes to liver.
  • Energy is generated primarily aerobically.

📘 Fuel use over time – graph example (OER Commons)


6. Predict the effect of increasing the pace of gentle exercise

Content Source: Slides 21–24

Explanation:

  • As pace increases:

    • Fatty acid oxidation cannot keep up with rising ATP demand.
    • Glucose oxidation resumes, as PDH inhibition is lifted.
    • Muscle uses glucose from liver glycogen and blood glucose.
    • Fuel shift from fat → mixed fuel use (fat + glucose).

🧬 Comparison of fuel usage by intensity (ResearchGate figure)


7. Compare the pattern of fuel utilisation in moderate exercise with that in gentle exercise

Content Source: Slide 24

Explanation:

  • Gentle: Mostly fatty acid oxidation, some glucose → lactate.

  • Moderate:

    • Fatty acid oxidation reaches maximum enzyme capacity.
    • Glucose oxidation increases to meet higher demand.
    • Glucose from liver glycogen and less recycling (less lactate).
  • Net effect: Mixed oxidation, increased reliance on glucose, faster glycogen depletion.

📘 Carbohydrate and fat usage in exercise – Lecture image


8. Describe the changes to fatty acid oxidation that occur when an athlete changes from gentle to strenuous exercise

Content Source: Slides 24–26

Explanation:

  • Fatty acid oxidation is maxed out during moderate intensity.
  • In strenuous exercise, demand outpaces mitochondrial oxidative capacity.
  • Glucose oxidation increases significantly; fatty acid contribution plateaus.
  • Eventually, reliance shifts to muscle glycogen for rapid ATP supply.

🧪 Study: Fat and carbohydrate metabolism during exercise (J Appl Physiol)


9. Explain the circumstances under which muscle glycogen is mobilised

Content Source: Slides 25–27, 29

Explanation:

  • Glycogen is mobilised when:

    • Blood glucose and fatty acids can’t meet demand.
    • Exercise intensity increases.
    • Sprinting requires rapid ATP, which oxidative phosphorylation can’t supply fast enough.
  • Glycogen → G6P → glycolysis → ATP/lactate.

  • Glycogen is stored within the muscle and is readily accessible.

📘 Why glycogen is critical to athletes (Examine.com)


10. Outline the pattern of fuel utilisation in strenuous exercise

Content Source: Slides 25–27

Explanation:

  • Fatty acids continue at max capacity.
  • Liver glycogen and blood glucose help but become limiting.
  • Muscle glycogen is mobilised and becomes primary source.
  • High glycolytic flux → ↑ lactate.
  • Anaerobic glycolysis contributes to ATP → quick but inefficient (2 ATP/glucose).

🎥 Anaerobic vs aerobic metabolism (Armando)


11. State the importance of glycogen to the competitive athlete

Content Source: Slide 29, 33

Explanation:

  • Glycogen is the only fuel fast enough for high-intensity efforts like sprinting.

  • Once depleted:

    • Rely solely on fatty acids → lower ATP rate.
    • Athlete may “hit the wall” due to fatigue.
  • Adequate glycogen stores:

    • Delay fatigue.
    • Allow sustained high performance.
    • Are replenished by carbohydrate loading pre-event.

📘 Why do athletes carbo-load? (Scientific American)


🔥 Fuel Oxidation Strategy – Lecture 2 (Week 1)


🧠 PART 1: Understanding the Principles (Strategy & Logic)

Think of the body like a hybrid car that runs on different fuels (glucose, fatty acids, amino acids), depending on what’s available and what you’re doing (resting, exercising, fasting, etc.).


⚙️ Principles of Fuel Oxidation

  1. Goal = Make ATP

    • ATP is your body’s “battery”.

    • All fuel oxidation pathways exist to make ATP to power cells.

  2. Fuel Types:

    Fuel When Used Features
    Glucose After eating, fast bursts of exercise Fast, anaerobic possible
    Fatty acids Fasting, rest, prolonged exercise Slow, high yield
    Amino acids Starvation, muscle breakdown Backup fuel
  3. Oxidation Happens in Stages:

    1. Glycolysis / β-oxidation: Break fuel into 2-carbon units (acetyl-CoA)

    2. Krebs Cycle: Burn acetyl-CoA → release electrons

    3. ETC + Oxidative Phosphorylation: Use electrons to make ATP

  4. Flexibility is key:

    • The body switches fuels based on energy needs and hormone signals (insulin vs glucagon).
  5. Insulin promotes storage and glucose use.

    Glucagon promotes release and fat use.


💡 Real-World Analogy:

Glucose is like petrol — easy to use, burns fast.

Fat is like diesel — lasts longer but burns slowly.

Protein is like burning the car’s seats — you only use it when you’re desperate.


📘 PART 2: Rote Learning (Pathways to Memorise)

Even though understanding is most important, there are certain steps and enzymes you just have to memorise.


🔹 Glycolysis (Glucose → Pyruvate)

  • Happens in cytoplasm

  • No oxygen needed

  • Key enzymes:

    • Hexokinase / Glucokinase

    • Phosphofructokinase (PFK-1) → rate-limiting step

    • Pyruvate kinase

🧪 ATP Yield:

2 ATP net per glucose (plus 2 NADH)


🔹 β-Oxidation (Fatty acids → Acetyl-CoA)

  • Happens in mitochondria

  • Requires carnitine shuttle for entry

  • Cycles remove 2 carbon units as acetyl-CoA

  • Generates lots of NADH, FADH₂

🧪 ATP Yield:

~106 ATP from one palmitate (16C fatty acid)


🔹 Krebs Cycle (Acetyl-CoA → CO₂ + energy)

  • Happens in mitochondrial matrix

  • Each turn:

    • 3 NADH

    • 1 FADH₂

    • 1 GTP (≈ ATP)

Enzymes to learn:

  • Citrate synthase

  • Isocitrate dehydrogenase

  • α-ketoglutarate dehydrogenase


🔹 Electron Transport Chain (ETC) + Oxidative Phosphorylation

  • Happens in inner mitochondrial membrane

  • NADH/FADH₂ donate electrons → pump protons

  • ATP Synthase uses proton gradient to make ATP

🧪 ATP Yield:

  • 2.5 ATP per NADH

  • 1.5 ATP per FADH₂


📊 Fuel Oxidation Flow Chart (Simplified)

Glucose → Glycolysis → Pyruvate → Acetyl-CoA → Krebs → ETC → ATP  

Fatty Acid → β-Oxidation → Acetyl-CoA → Krebs → ETC → ATP  

Amino Acid → Transamination → Keto Acids → Acetyl-CoA or TCA Intermediates → ATP  

🔁 Summary Strategy

Condition Preferred Fuel Hormone Notes
Fed Glucose ↑ Insulin Store fat, make ATP
Fasting Fatty acids ↑ Glucagon Gluconeogenesis for brain
Exercise (short, intense) Glucose (muscle glycogen) ↓ Insulin Anaerobic possible
Exercise (prolonged) Fatty acids ↓ Insulin, ↑ Glucagon Conserves glucose

🔗 Rules of Coupling – Matching Fuel Oxidation to ATP Demand

🧠 What is “Coupling”?

Coupling means that two things are linked — if one happens, the other happens too.

In the mitochondria:

Fuel oxidation (burning glucose or fat) is coupled to ATP synthesis.

So:

👉 If the cell needs more ATP, it will also burn more fuel.

👉 If no ATP is needed, fuel oxidation slows down or stops.


🧬 How is This Coupling Controlled?

It’s all about ADP levels and the proton gradient in the mitochondria:


⚙️ The Mechanism:

  1. ATP Synthase makes ATP by letting protons (H⁺) flow back into the mitochondrial matrix.

  2. This proton flow only happens when there is ADP + Pi to turn into ATP.

  3. If there’s no ADP, protons stop flowing, and the proton gradient builds up.

  4. This stops the electron transport chain, which stops fuel oxidation.


🔁 So it works like this:

  • Lots of ADP = lots of ATP being made = fuel gets burned

  • Little or no ADP = no ATP being made = fuel oxidation slows or stops

This is chemiosmotic coupling — ATP synthesis is linked to fuel oxidation through the proton gradient.


🎢 Analogy for Kids (But with Correct Terms)

Imagine a hydroelectric dam (the mitochondria):

  • The water behind the dam = proton gradient
  • The turbine = ATP synthase
  • Electricity = ATP

The water (H⁺) only flows through the turbine if you’re using electricity (need ATP).

If you’re not using electricity (no ADP), the water stops flowing, and the generators (ETC) shut down.


🧪 Biochemical Players:

Component Function
Fuel (glucose, fat) Oxidised to provide electrons (NADH/FADH₂)
ETC (Complex I–IV) Pumps protons into intermembrane space
Proton Gradient (PMF) Stores energy like a battery
ATP Synthase (Complex V) Uses proton flow to make ATP
ADP availability Acts like a switch – turns ATP synthesis (and fuel use) on or off

📊 Summary Table

Condition ADP Level Proton Flow ATP Made Fuel Oxidised
Rest Low Slow Little Little
Exercise High Fast Lots Lots
ATP synthase blocked (e.g. oligomycin) Irrelevant Blocked None None
Uncoupled (e.g. UCP-1) Normal Flow allowed Less Lots (but wasted as heat)

🧠 Why This Matters:

Coupling prevents waste.

The body only uses fuel when ATP is actually needed.

This keeps metabolism efficient, like a smart energy-saving system.


L3: Glycolysis, Fatty Acid Oxidation, and Krebs Cycle


🔷 Beta-Oxidation


1. Identify the carbon atoms in fatty acids based on different codes of nomenclature

Content Source: Slide 5, presenter notes

Explanation:

  • Fatty acids are long-chain hydrocarbons with a carboxyl group (-COOH) at one end.

  • Numbering systems:

    • IUPAC: Starts at the carboxyl carbon (C1), counts down the chain.
    • Omega (ω): Starts at the methyl end (opposite the COOH).
    • Greek letters: α = C2, β = C3. β-carbon is the site of oxidation, hence “β-oxidation”.

📘 Fatty acid nomenclature tutorial (Lumen Learning)


2. Explain how fatty acids are transported into the cytoplasm, trapped, and then transferred into the mitochondrial matrix

Content Source: Slides 7–10, 11, presenter notes

Explanation:

  • Transport: In blood, fatty acids are bound to albumin due to their hydrophobicity.

  • Trapping: In the cytoplasm, fatty acids are activated to fatty acyl-CoA by acyl-CoA synthetase, which:

    • Requires ATP → AMP + PPi (equivalent to 2 ATP).

    • Makes the reaction irreversible.

  • Mitochondrial import:

    • FA-CoA cannot cross the inner mitochondrial membrane.

    • It is transferred to carnitine by CAT-1, forming FA-carnitine.

    • Translocase shuttles it across.

    • Inside matrix, CAT-2 converts it back to FA-CoA.

📺 Beta-oxidation animation (YouTube)


3. Explain the biochemical features of CoA that enable it to do its job

Content Source: Slide 11

Explanation:

  • Coenzyme A (CoA) has a reactive thiol group (-SH) that forms high-energy thioester bonds with acyl groups (e.g., acetyl-CoA).

  • It’s a large, charged molecule, preventing free diffusion across membranes—this helps “trap” metabolites inside cells.

  • Acts as a carrier, transferring acyl groups in metabolism (e.g., fatty acid oxidation, Krebs).

  • It costs 2 ATP molecules to join CoA to FA

📘 Coenzyme A structure and role (Khan Academy)


4. Appreciate the role played by FAD and NAD in FA-CoA oxidation

Content Source: Slides 12–14

Explanation:

  • In β-oxidation, two key redox steps:

    • FAD oxidises the β–α bond, forming a trans double bond (→ FADH₂).

    • NAD⁺ oxidises the resulting β-hydroxy group to a β-keto group (→ NADH).

  • Both feed into the ETC to produce ATP:

    • FADH₂ → 1.5 ATP

    • NADH → 2.5 ATP

📘 FAD/NAD redox roles explained (LibreTexts)


5. Understand the steps within the process of beta-oxidation

Content Source: Slides 13–16

Explanation:

Each cycle of β-oxidation removes 2 carbons as acetyl-CoA and generates 1 NADH and 1 FADH₂:

  1. Oxidation by FAD: FA-CoA → enoyl-CoA (trans double bond formed).

  2. Hydration: Water adds across the double bond → hydroxyacyl-CoA.

  3. Oxidation by NAD⁺: → ketoacyl-CoA.

  4. Thiolysis: CoA cleaves acetyl-CoA off → shortened FA-CoA.

Example: Palmitate (C16) → 7 cycles → 8 acetyl-CoA, 7 FADH₂, 7 NADH

📘 Full β-oxidation pathway – Slide + summary


🔷 Glycolysis


6. Give a strategic overview of glycolysis

Content Source: Slide 20, 21, 29

Explanation:

  • Glycolysis is a cytosolic pathway that breaks down glucose (6C) into 2 pyruvate (3C) molecules.

  • Two main phases:

    • Investment: 2 ATP consumed.

    • Payoff: 4 ATP + 2 NADH generated.

  • Anaerobic: No oxygen required.

📘 Glycolysis Overview – Harvard Animation


7. Discuss the process of glucose transport into the cytoplasm and trapping within the cell

Content Source: Slide 21

Explanation:

  • Glucose enters via GLUT transporters:

    • GLUT-1 (all cells), GLUT-2 (liver, pancreas), GLUT-4 (muscle/adipose, insulin-sensitive).
  • Once inside, glucose is phosphorylated by hexokinase/glucokinase → glucose-6-phosphate (G6P).

  • This traps glucose in the cell, as G6P cannot exit through GLUTs.

📘 GLUT transporters illustrated (NCBI)


8. Outline the energy investment and return stages of glycolysis

Content Source: Slides 22–25

Explanation:

  • Investment Phase:

    • 2 ATP used:

      • Glucose → G6P (via hexokinase)

      • F6P → F-1,6-BP (via PFK) - this adds another phosphate to glucose

        • fructose 6-phosphate shape is more symmetrical because at the end of investment it will be cut down the middle to end up with 2 3-Carbon sugar phosphates
  • Payoff Phase:

    • 4 ATP formed (net gain = 2 ATP).

    • 2 NAD⁺ → 2 NADH.

    • End products: 2 pyruvate + 2 NADH + 2 ATP (net)


🍬 Glycolysis: Breaking Down Sugar for Energy

🟥 1. Energy Investment Phase (Steps 1–5)

👉 You spend ATP to get things going

  • This is like putting money in to set up a lemonade stand.

  • The cell uses 2 ATP to “activate” glucose and turn it into something ready to break apart.

Step Enzyme What Happens
Step 1 Hexokinase Glucose → Glucose-6-phosphate (uses 1 ATP)
Step 3 Phosphofructokinase-1 (PFK-1) Fructose-6-phosphate → Fructose-1,6-bisphosphate (uses 1 ATP)

🧪 Total ATP used: 2


🟩 2. Energy Payoff Phase (Steps 6–10)

👉 You get ATP and NADH back

Now the cell breaks the sugar in half and makes energy and electrons:

  • For each glucose, you get:

    • 4 ATP
    • 2 NADH
    • 2 Pyruvate
Step Enzyme What’s Made
Step 6 Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) Makes NADH
Step 7 Phosphoglycerate kinase Makes ATP
Step 10 Pyruvate kinase Makes ATP and pyruvate

🧪 Total made: 4 ATP, 2 NADH


⚖️ Net Energy Summary

Molecule Total Made Total Used Net Gain
ATP 4 2 +2 ATP
NADH 2 0 +2 NADH
Pyruvate 2 2 Pyruvate

🎯 Why This Matters

  • The cell spends energy first, then makes more back.

  • This is “priming the pump” — like pushing a swing before it comes back higher.

  • Glycolysis is fast, anaerobic, and the first step in all metabolism, whether or not oxygen is available.


🧬 Diagram (Student-friendly):

🔵 Glucose
   ↓ [–1 ATP: Hexokinase]
🔵 Glucose-6-P
   ↓
🔵 Fructose-6-P
   ↓ [–1 ATP: PFK-1]
🔵 Fructose-1,6-BP
   ↓
🔵 2 G3P
   ↓ [GAPDH → +2 NADH]
   ↓ [PGK, PK → +4 ATP]
🔵 2 Pyruvate

9. Identify the different fates of pyruvate and understand when each is required or desirable

Content Source: Slides 27, 29

Explanation:

  • Aerobic conditions:

    • Pyruvate enters mitochondria.

    • Converted to acetyl-CoA by PDH → enters Krebs cycle.

  • Anaerobic conditions:

    • Converted to lactate (via lactate dehydrogenase) to regenerate NAD⁺.
  • Alternative fates: In yeast → ethanol; in liver → gluconeogenesis.

🧪 What Happens to Pyruvate After Glycolysis?

When glucose is broken down in glycolysis, the final product is pyruvate.

Now the cell has to decide:

👉 What do I do with this pyruvate?

Think of pyruvate like a train at a station with multiple tracks.

Which track it takes depends on oxygen, energy needs, and cell type.


🚂 Pyruvate’s Possible Fates (Train Tracks)


1️⃣ Pyruvate → Acetyl-CoA

🔹 Aerobic conditions (with oxygen)

🔹 Happens in mitochondria

Enzyme Pyruvate dehydrogenase (PDH)

This is the main pathway in cells when oxygen is available.

🧬 Acetyl-CoA goes into the Krebs cycle → then to ETClots of ATP!

Used when:

  • Oxygen is present

  • Cell wants to make lots of ATP

  • Tissues like muscle, brain


2️⃣ Pyruvate → Lactate

🔹 Anaerobic conditions (no oxygen)

🔹 Cytoplasm

Enzyme Lactate dehydrogenase (LDH)

This happens in:

  • Working muscles during intense exercise

  • Red blood cells (no mitochondria)

🧪 Why? To regenerate NAD⁺ so glycolysis can keep going!

Used when:

  • Oxygen is low

  • You need ATP fast

  • For short-term emergency energy

🧪 Example: Sprinting → burning legs → lactate buildup


3️⃣ Pyruvate → Alanine

🔹 Amino acid metabolism

| Enzyme | Alanine aminotransferase (ALT) |

This happens in muscle when amino groups are transferred.

Alanine then travels to the liver, gets turned back into pyruvate, and used for gluconeogenesis.

Used when:

  • Muscle is breaking down protein

  • You need to send fuel to the liver

  • Part of the glucose-alanine cycle


4️⃣ Pyruvate → Oxaloacetate

🔹 For gluconeogenesis (making new glucose)

🔹 Also replenishes Krebs cycle intermediates

| Enzyme | Pyruvate carboxylase (needs biotin + ATP) |

🧬 Oxaloacetate is the entry point into gluconeogenesis → helps maintain blood glucose during fasting.

Used when:

  • You’re fasting

  • The body needs to make glucose (e.g. for brain)

  • Or to recharge the Krebs cycle (anaplerosis)


🧾 Summary Table: Pyruvate Fates

Pathway Product Enzyme When/Why
Oxidation Acetyl-CoA Pyruvate dehydrogenase With oxygen, for ATP
Anaerobic Lactate Lactate dehydrogenase No oxygen, fast ATP
Transamination Alanine ALT Transport nitrogen + fuel
Carboxylation Oxaloacetate Pyruvate carboxylase Gluconeogenesis or anaplerosis

🎯 Easy Analogy:

Pyruvate is like a 3-way fork in the road:

🛣️ Go to power plant (mitochondria) → Acetyl-CoA

🛻 Shortcut to keep running (lactate) if there’s no oxygen

🚚 Delivery route to liver (alanine or oxaloacetate)


🔷 Krebs Cycle


10. Understand the position of the Krebs cycle in catabolism

Content Source: Slides 31–33

Explanation:

  • Central hub of metabolism.

  • Acetyl-CoA from glycolysis, β-oxidation, or amino acids enters cycle.

  • Occurs in mitochondrial matrix.

  • Oxidises acetyl groups to 2 CO₂, while generating NADH, FADH₂, and GTP.

📘 Krebs Cycle as metabolic hub (Nature Education)


11. Outline the overall process and strategy of the Krebs cycle

Content Source: Slide 33

Explanation:

  • Acetyl-CoA (2C) + Oxaloacetate (4C) → Citrate (6C)

  • Two decarboxylations → 2 CO₂

  • Yields: 3 NADH, 1 FADH₂, 1 GTP per cycle

  • Oxaloacetate regenerated = cycle.

  • NADH/FADH₂ go to ETC for ATP production.

📘 Krebs cycle summary (Sackville HS)


12. Describe the major intermediates and regulatory steps in the Krebs cycle

Content Source: Slides 34–35 Explanation:

  • Key intermediates: Citrate, α-ketoglutarate, succinate, malate, oxaloacetate

  • Regulated enzymes:

    • Citrate synthase (entry point)
    • Isocitrate dehydrogenase (rate-limiting step)
    • α-Ketoglutarate dehydrogenase
  • Regulation by:

    • Substrates (ADP, NAD⁺) = activators
    • Products (ATP, NADH) = inhibitors

📘 Regulation summary (Biology LibreTexts)


L4: Electron Transport Chain (ETC) and ATP Synthesis


ETC

1. Describe how measurement of oxygen consumption or carbon dioxide production gives an estimate of whole-body energy expenditure

Explanation:

  • The ETC consumes O₂ and produces CO₂ as part of fuel oxidation.
  • This is the basis for indirect calorimetry: more O₂ used = more fuel oxidised = higher energy expenditure.
  • Sprinting disrupts this coupling temporarily, so post-exercise O₂ consumption remains high (EPOC).
Measuring VO₂ and VCO₂ to Estimate Energy Use
🧠 What’s the Big Idea?

Your body burns fuel (like glucose and fat) to make ATP, just like a fire burns wood to make heat.

But to burn fuel, your body needs oxygen — and when it burns it, it releases carbon dioxide.

So:

🧪 The more oxygen you use, the more energy you’re burning.

💨 The more carbon dioxide you produce, the more fuel you’ve used.


🫁 How We Measure It

Scientists use a machine called a metabolic cart to measure:

  • VO₂ = Oxygen consumption (how much O₂ you breathe in and use)

  • VCO₂ = Carbon dioxide production (how much CO₂ you breathe out)

These numbers tell us how much energy your body is using.


🔢 How It Estimates Energy

When your body uses oxygen to break down fuel:

  • Glucose uses O₂ and makes CO₂ (1:1 ratio)

  • Fat uses more O₂ and makes less CO₂

Each litre of oxygen consumed = about 20.1 kJ of energy burned (on average)


📘 Real-World Analogy

Imagine you’re watching a campfire 🔥

  • The more wood (fuel) you burn, the more oxygen is sucked in, and more smoke (CO₂) comes out.
  • So by measuring the air going in and out, you can tell how big the fire is — that’s what your body is doing with food!

🏃 When Is This Used?
  • In exercise physiology labs
  • In clinical settings to monitor metabolism
  • To calculate Resting Metabolic Rate (RMR) or Total Energy Expenditure (TEE)

🧪 Key Terms
Term What It Means
VO₂ Oxygen consumption (used to oxidise fuel)
VCO₂ CO₂ produced when fuel is broken down
Respiratory Quotient (RQ) VCO₂ / VO₂ → tells which fuel you’re using
Energy Expenditure How much energy (kJ or kcal) you’re burning

🧮 Example:

If someone uses 2 L of oxygen per minute, they are burning:

\[ 2 \, \text{L/min} \times 20.1 \, \text{kJ/L} = 40.2 \, \text{kJ/min} \]

That’s how scientists calculate how much energy your whole body is using — just from the air you breathe.


2. Explain the principles of uncoupling and how DNP and UCP-1 can lead to uncontrolled or regulated uncoupling respectively

Explanation:

  • Coupling: ETC activity is limited by ATP demand. Without ATP synthesis, the proton gradient becomes too steep, halting ETC.

  • Uncoupling = Protons return to the matrix without passing through ATP synthase, dissipating the gradient:

    • DNP (2,4-dinitrophenol): synthetic uncoupler that carries H⁺ across membrane → uncontrolled, dangerous.

    • UCP-1 (thermogenin): a regulated proton channel in brown adipose tissue, activated by noradrenaline → generates heat (thermogenesis).

  • Uncoupling increases O₂ use and fuel oxidation but prevents ATP synthesis.

📘 Image: Mechanism of DNP & UCP-1 – Biology LibreTexts


3. Explain how the chemical properties of NAD, FAD, UQ and the complexes allow them to fulfil their roles in electron transport

Explanation:

  • NAD⁺/NADH: Hydrophilic, mobile electron carrier; donates e⁻ to Complex I.

  • FAD/FADH₂: Bound tightly to Complex II, accepts 2H⁺ and 2e⁻ from β-oxidation and Krebs.

  • Ubiquinone (UQ):

    • Hydrophobic, mobile within the inner membrane.

    • Accepts electrons from Complex I, II, β-oxidation, and G3P shuttle → transfers them to Complex III.

  • Complexes I–IV contain iron-sulphur clusters and cytochromes that pass e⁻ down the chain.

🎥 NADH, FADH₂, and UQ roles – YouTube Animation


4. Outline how the exchange of electrons between different types of carriers can lead to proton pumping

Explanation:

  • Carriers that accept both e⁻ and H⁺ (like UQH₂) can transfer only the e⁻ to the next carrier.

  • The H⁺ is released into the intermembrane space → this is proton pumping.

  • Complexes I, III, IV pump protons:

    • I: ~4 H⁺

    • III: ~4 H⁺

    • IV: ~2 H⁺

  • FADH₂ → Complex II → UQ → bypasses proton pumping at Complex I.

📘 Mechanism: Proton pumping – Nature Education


5. Appreciate the electrical and concentration components of the proton motive force

Explanation:

  • The proton-motive force (PMF) is made of:

    • ΔpH (concentration gradient): [H⁺] higher in intermembrane space.
    • Δψ (membrane potential): matrix is more negative than intermembrane space.
  • This electrochemical gradient drives H⁺ back into the matrix via ATP synthase or other transporters.

📘 Diagram: Proton Motive Force


6. Appreciate the reasons why cytosolic NAD⁺ regeneration presents an important challenge and how the glycerol 3-phosphate shuttle and the malate-aspartate shuttle can help

Explanation:

  • Glycolysis produces NADH in the cytosol, but the mitochondrial inner membrane is impermeable to NADH.

  • Two shuttles transfer H⁺/e⁻ into mitochondria:

    • G3P Shuttle: uses G3P and FAD → feeds into UQ, bypassing Complex I (less ATP yield).
    • Malate-Aspartate Shuttle: uses malate/OAA cycle → preserves proton pumping via Complex I.

📘 Shuttle Comparison Chart – Lumen Learning


7. Organise the four separate routes that feed into UQ (Complex I, Complex II, G3P shuttle and beta-oxidation)

Explanation:

  • Ubiquinone (Q) receives electrons from:

    1. Complex I (via NADH)

    2. Complex II (succinate dehydrogenase, via FADH₂)

    3. G3P shuttle (via G3P → FADH₂)

    4. β-oxidation (via electron-transferring flavoprotein → ETF:UQ oxidoreductase)

📘 Figure: 4 routes to Q – Lehninger Fig. 19.8


8. Outline the mechanisms involved in the generation and destruction of free radicals

Explanation:

  • At UQ, if e⁻ builds up and leaks to O₂ prematurely → superoxide radical (O₂⁻·) forms.
  • These can damage proteins, lipids, DNA.
  • Complex III is a major site of this leakage.
  • Antioxidant systems (e.g., glutathione, SOD) neutralise free radicals.

📘 ROS and mitochondrial antioxidant systems – ScienceDirect


ATP Syntehsis

9. List the components and functions of the ATP synthase and explain the mechanism by which it produces ATP

Explanation:

  • ATP synthase (F₀F₁-ATPase):

    • F₀: membrane-embedded proton channel; protons cause rotation of γ-subunit.

    • F₁: catalytic headpiece; β-subunits cycle through 3 conformations:

      1. Bind ADP + Pi

      2. Form ATP

      3. Release ATP

  • 3 H⁺ = 1 ATP (approx.)

  • Rotation-driven conformational changes result in ATP formation without direct chemical input.

ATP Synthase Animation I – Harvard

ATP Synthase Animation II – Harvard


10. Recognise the contribution of the proton gradient to processes other than the ATP synthase

Explanation:

  • Besides ATP production, the proton gradient powers:

    • ATP/ADP antiport: exports ATP, imports ADP
    • Pi symport: imports phosphate along with H⁺
  • Also used in:

    • Calcium import/export
    • Thermogenesis (in brown adipose)
  • Gradient is essential for multiple mitochondrial functions.

📘 Transport roles of PMF – Molecular Cell Biology


11. Understand the assumptions made in tables which claim to calculate the yield of ATP from different metabolic pathways and fuels

Explanation:

  • Assumptions include:

    • 2.5 ATP per NADH
    • 1.5 ATP per FADH₂
    • Full coupling and no proton leak
  • These values may vary based on:

    • Use of shuttles (G3P = lower yield)
    • Membrane leakiness
    • ATP used for transport
  • Therefore, these are theoretical estimates, not absolute.

📘 ATP yield breakdown – BioNinja


12. Using the fundamental rules of coupling, extrapolate to predict the effects of various interventions on the rates of fuel oxidation, maintenance of the proton gradient and ATP generation

Explanation:

  • If ATP demand falls → ADP falls → ATP synthase slows → proton gradient builds → ETC stalls → oxidation slows.

  • If ATP synthase is blocked (e.g. oligomycin):

    • Gradient becomes too steep

    • ETC halts

    • NADH accumulates → oxidation stops

  • Uncoupling (e.g. DNP):

    • Gradient collapses

    • ETC speeds up (unrestrained)

    • But no ATP produced

  • Inhibitors:

    • Rotenone: blocks Complex I → NADH can’t be oxidised

    • Cyanide: blocks Complex IV → ETC backup

    • Methylene blue: bypasses block, acts as e⁻ acceptor

📘 Coupling logic – LearnBiochem


L5: Glycogenolysis, Lipolysis and Gluconeogenesis (Early Starvation)


1. Outline the fuel stores in the human body

Explanation:

  • Major energy stores:

    • Fat (white adipose tissue): ~12–15 kg, high energy density (37 kJ/g).

    • Liver glycogen: ~100 g, maintains blood glucose.

    • Muscle glycogen: ~400 g, used locally in muscle.

    • Protein: 5–10 kg, not stored for fuel; catabolised only under extreme conditions (e.g., late starvation).

  • Fat is the dominant long-term energy store, but the brain cannot use it directly due to the blood-brain barrier.

📘 Fuel store comparison – Lumen Learning


2. Describe the general principles of whole-body glucose homeostasis

Explanation:

  • Euglycemia = maintaining blood glucose ~5 mM.

  • Vital for tissues like brain, kidney medulla, RBCs.

  • Achieved by:

    1. Glycogenolysis in liver (rapid).

    2. Gluconeogenesis from lactate, glycerol, and amino acids.

    3. Inhibition of glucose oxidation in peripheral tissues.

  • Hormonal control:

    • ↓ Insulin, ↑ Glucagon → promote glucose conservation.

📘 Glucose homeostasis overview – NCBI


3. Predict the changes in blood glucose during the first few hours of a fast

Explanation:

  • Glucose use continues while dietary sources drop → blood glucose starts falling.

  • Liver rapidly releases glucose via glycogenolysis.

  • Glucagon secretion increases, insulin drops.

  • Blood glucose stabilises at ~4 mM due to hormonal compensation.

🧠 Diagram – Early fasting glucose control


8. Understand the way in which glucagon and insulin signal tissue-specific metabolic changes

Explanation:

  • Glucagon:

    • ↑ in liver and adipose tissue (not muscle).

    • Activates cAMP-PKA pathways → ↑ glycogenolysis and lipolysis .

  • Insulin:

    • Acts on muscle, liver, and fat.

    • ↓ during fasting → inhibits glycogenesis, lipogenesis, and glucose uptake.

  • Muscle relies on GLUT1 for baseline glucose uptake (GLUT4 is insulin-dependent).

📘 Infographic: Glucagon vs Insulin effects – Kenhub


11. Summarise the patterns of fuel selection and mobilisation in early starvation

Explanation:

  • 0–6 hours post-absorptive:

    • Glucose from meal still available
  • 6–24 hours:

    • Blood glucose falls → ↑ glucagon
    • Liver glycogenolysis starts
    • Muscle shifts to fatty acids
  • >24 hours:

    • Glycogen depleted
    • Gluconeogenesis + fat oxidation dominate
    • Glucose use restricted to brain, RBCs, kidney

📘 Timeline – Starvation phases chart (UpToDate)


Glycogenolysis

4. Recall how glycogen is mobilised from the liver and how the process is regulated

Source: L6.2 pg 6, 8

Explanation:

  • Glycogenolysis:

    • Glycogen → G1P → G6P → Glucose (via G6Pase)

      • G6P itself can’t get back out to blood so ONLY in the liver there is G6Pase that takes phosphate off and turn it back into glucose to be carried back out into blood

      • G6Pase is hidden away in endoplasmic reticulum

    • Exported via GLUT2

  • Regulated by glucagon:

    • Glucagon binding receptor on liver cell causes

    –> Activates adenylate cyclase → ↑ cAMP

    –> cAMP activates PKA, which:

    –> Phosphorylates phosphorylase kinase to make it active

    –> Phosphorylate glycogen phosphorylase to make it active

    –> active & phosphorylated glycogen phosphorylase will break off glucose-1-phosphate off the end of glycogen chain

  • Debranching enzyme releases ~10% of glucose directly (as free glucose) rather than glucose-1-phosphate

  • complex cascade of phosphorylation from 1 glucagon can amplify the amount of glucose that’s released into blood glucose rather than binding directly

Flowchart

Glucagon binds to receptor on liver cell surface ↓ Activates adenylate cyclase ↓ Increases cAMP levels ↓ cAMP activates Protein Kinase A (PKA) ↓ PKA phosphorylates phosphorylase kinase → activates it ↓ Active phosphorylase kinase phosphorylates glycogen phosphorylase ↓ Glycogen phosphorylase becomes active ↓ Active glycogen phosphorylase breaks glucose-1-phosphate off the ends of glycogen chains

📘 Pathway image – Glycogenolysis


5. Understand why muscle cannot contribute to blood glucose homeostasis

Explanation:

  • Muscle lacks glucagon receptors → no hormonal activation of glycogenolysis.

  • Muscle lacks G6Pase → G6P cannot be converted to free glucose.

  • Thus, glucose from muscle glycogen is used locally, not released into blood.

  • ~10% glucose can be released via hydrolysis at branch points.

📘 Explanation – Muscle glycogen is local only


6. Assess the dynamics of glycogen depletion in starvation

Explanation:

  • Liver glycogen (~100 g) provides glucose during early fasting.
  • Depleted within 24 hours due to brain demand (~120 g/day).
  • Therefore, body must switch to fat use and gluconeogenesis before depletion.
  • If not, hypoglycemia risks arise quickly.

🧠 Graph – Liver glycogen over fasting


Lipolysis

7. Describe how fat is mobilised from the adipose tissue and how the process is regulated

Explanation:

  • Low insulin and high glucagon promote fat mobilisation.

  • In WAT:

    • Glucagon → ↑ cAMP → ↑ PKA activity

    • PKA phosphorylates Hormone Sensitive Lipase (HSL) and perilipin

    • HSL hydrolyses triglycerides → 3 FA + 1 glycerol

      • Because cytoplasm is hydrophilic and lipid droplet is hydrophobic –> hard to work between these two

      • So, PERILIPIN unpicks the edges of th lipid vacuole which creates an opening for HSL to come in and start breaking down the fat.

  • FAs bind albumin in blood; glycerol goes to liver for gluconeogenesis.

    • glycerol can diffuse into the blood stream and be used as an alternative fuel to glucose

    • can diffuse into muscle cell as alternative

📘 Animation – Hormone-sensitive lipase activation


9. Predict the effect of fatty acid oxidation on glucose oxidation

Explanation:

  • Fatty acid oxidation → lots of acetyl-CoA.

  • Acetyl-CoA activates PDH kinase, which inhibits PDH (pyruvate dehydrogenase).

  • Inhibiting PDH prevents pyruvate → acetyl-CoA → blocks glucose oxidation.

  • Pyruvate is instead converted to lactate, sparing glucose for the liver.

  • Ac-CoA does not inhibit PDH directly

    • PDH works opposite to other enzymes –> phosphorylation actually INACTIVATES PDH and dephosphorylation ACTIVATES PDH

    • high levels of Ac-CoA will activate PDH kinase –> phosphorylate PDH –> when PDH phosphorylated it will become inactive

  • PDH phosphotase will activate it again

    • gets activated by insulin –> so when meal consumed and insulin releases –> switch on PDH again –> burn glucose fuel to CO2

📘 Mechanism – Glucose-fatty acid cycle (Randle cycle)


Gluconeogenesis

10. Describe the central features of the glucose-fatty acid cycle and the Cori Cycle

Explanation:

Glucose-Fatty Acid Cycle (Randle Cycle):

  • FA oxidation → ↑ acetyl-CoA → inhibits PDH → ↓ glucose oxidation.

  • Muscle switches to fat → glucose spared.

Cori Cycle:

  • Muscle: glucose → pyruvate → lactate

  • Lactate → liver → converted back to glucose via gluconeogenesis

  • Recycled glucose returns to blood → reused by muscle/RBCs.

📘 Cori cycle diagram – Osmosis


L6: Fuel Selection in Late Starvation


Gluconeogenesis

1. Summarise the substrates available for de novo gluconeogenesis

Explanation:

  • Only three substrates contribute to new (de novo) glucose:

    1. Glycerol: from lipolysis; enters as dehydroxyacetone phosphate (DHAP) (30 g/day).

    2. Amino acids: from proteolysis; carbon skeletons become glucose intermediates.

    3. Lactate: from anaerobic glycolysis; however, this is recycling, not de novo.

📘 Overview – Gluconeogenic substrates (Khan Academy)


2. Explain why using lactate as a substrate does not increase the circulating glucose pool via gluconeogenesis

Explanation:

  • Lactate → glucose via Cori Cycle is a closed loop:

    • Glucose → pyruvate → lactate in muscle

    • Lactate → pyruvate → glucose in liver

  • Net result: no new glucose added to the pool, only recycled.

  • Requires ATP in liver but doesn’t expand total glucose availability.

📘 Cori Cycle visual – Osmosis


3. Describe the overall structure and strategy of gluconeogenesis

Explanation:

  • Gluconeogenesis = almost reverse of glycolysis, but:

    • Bypasses 3 irreversible steps:

      • Pyruvate kinase → uses pyruvate carboxylase + PEPCK

      • Phosphofructokinase → bypassed by fructose-1,6-bisphosphatase

      • Hexokinase → bypassed by G6Pase

  • Occurs mainly in the liver, partly mitochondrial (pyruvate carboxylase).

  • Uses ATP and NADH.

📘 Pathway map – Lumen Learning


Proteolysis

4. Frame the overarching principles of amino acid processing

Explanation:

  • After proteolysis, amino acids are:

    • Deaminated using PLP (vitamin B6)

    • Amine groups, using amino-transferase enzyme, transferred to:

      • Pyruvate → alanine

      • α-ketoglutarate → glutamate

      • OAA → aspartate

  • Carbon skeletons → enter gluconeogenesis.

  • Amine groups → sent to urea cycle for excretion.

  • If an AA backbone can only be made into acetyl-CoA, it CANNOT be made into glucose

  • If it can be made into pyruvate or Krebs cycle intermediate, it CAN be made into glucose.

📘 Amino group transfer and urea cycle – LibreTexts


5. Outline the general flow of nitrogenous compounds in starvation

Explanation:

  • Most nitrogen comes from muscle protein degradation.

  • NH₃ is toxic → converted into urea in the liver:

    • Inputs: glutamate + aspartate

    • Urea excreted in urine.

  • Urea cycle is energy-intensive.

📘 Urea cycle simplified – Osmosis


Ketone Body Synthesis

6. Understand how ketone bodies are formed and how they help to address the shortfall on glucose demanded by the brain during long-term starvation

Explanation:

  • After 2–3 days: liver oxidises fatty acids → acetyl-CoA accumulates.

  • Every step of beta-oxidation needs FAD, NAD+ and CoA

    • CoA in limited supply. So if it runs out, 2-carbon chunking can’t perform in beta oxidation
  • So, Acetyl-CoA converted into ketone bodies:

    1. Acetoacetate

    2. β-hydroxybutyrate

  • Turning Ac-CoA into something that has no CoA on it so we can get our CoAs back to continue to burn fats in the liver despite no demand for ATP

  • Taken up by brain, converted to acetyl-CoA → Krebs cycle fuel.

  • Reduces brain glucose demand from 120g → ~30g/day.

📘 Ketone body formation – Nature


7. Predict the source of inefficiencies in energy metabolism induced by the ketotic state

Explanation:

  • Ketone body oxidation itself is efficient, but:

    • Some ketones are lost in urine or sweat (e.g., acetone).

    • Spontaneous decarboxylation → metabolic waste.

    • Net loss of carbon-containing energy substrates.

📘 Inefficiencies in ketone metabolism – PMC


Summary

8. Summarise the patterns of fuel selection and mobilisation in late starvation

Explanation:

Tissue Primary Fuel
Brain Ketone bodies, some glucose
Muscle Fatty acids
Liver Fatty acids (for ATP), amino acids and glycerol for gluconeogenesis
Kidney Ketones, some gluconeogenesis
RBCs Glucose → lactate (Cori cycle)
  • By day 5:

    • Proteolysis drops (from >100g to ~50g/day)

    • Ketones take over as primary brain fuel

    • Fat and glycerol support gluconeogenesis

📘 Metabolic shift in starvation – Cahill 2006


9. Construct flow diagrams to encapsulate the movement, source and fate of fuels during extended starvation

Diagram Suggestion: L6 pg 18, 21

Adipose Tissue → FA & Glycerol
                  ↓       ↓
             (Liver)  (Liver) 
           β-oxidation → ATP
               ↓          ↓
     Ketogenesis     Gluconeogenesis ← Amino Acids (from muscle)

                       ↓
         Blood glucose + ketones → Brain, RBCs, kidney

📘 [Starvation metabolism overview – BioRender or Lehninger Textbook Fig 27-3]


10. Explain the role played by glucagon in extended starvation

Explanation:

  • Glucagon dominates in prolonged fasting:

    • Stimulates lipolysis in adipose tissue
    • Activates gluconeogenesis in liver
    • Promotes ketogenesis
    • Inhibits glycolysis and lipogenesis
  • Maintains blood glucose and ketone levels despite low insulin.

📘 Glucagon signalling – Khan Academy

TRY THIS:

A person has been starving for 12 hours. They are then treated with a compound. Which IMMEDIATE outcome (i.e. in the next hour) matches each the treatment. (NB. Each outcome could result from more than one treatment):

  1. Injection of an inhibitor of hormone sensitive lipase

  2. Injection of an inhibitor of carnitine acyl transferase

  3. Injection of an inhibitor of muscle pyruvate dehydrogenase


Blood [Glycerol] Muscle Fatty Acid Oxidation Brain Glucose Oxidation Liver Gluconeogenesis
A DECREASES DECREASES SAME DECREASES
B SAME SAME SAME SAME
C SAME DECREASES SAME DECREASES
D DECREASES INCREASES INCREASES SAME
E SAME SAME DECREASES INCREASES




ANSWER

1. Inhibitor of Hormone Sensitive Lipase (HSL)

HSL function: HSL breaks down triglycerides in adipose tissue, releasing free fatty acids (FFA) and glycerol into the blood. Inhibition effect:

  • ↓ Lipolysis → ↓ glycerol in blood
  • ↓ FFA availability → ↓ fatty acid oxidation in muscle
  • ↓ Glycerol to liver → ↓ substrate for gluconeogenesis Answer: A | Blood [Glycerol] ↓ | Muscle FA oxidation ↓ | Brain glucose SAME | Liver gluconeogenesis ↓ | ✅ Matches all consequences of reduced lipolysis.

2. Inhibitor of Carnitine Acyl Transferase (CAT)

CAT (specifically CAT I) is essential for transporting long-chain fatty acids into mitochondria for β-oxidation in muscle. Inhibition effect:

  • ↓ Fatty acid oxidation in muscle
  • Brain glucose oxidation remains SAME (brain depends on glucose and ketones, not muscle FA oxidation)
  • ↓ Acetyl-CoA from FA oxidation → ↓ allosteric activation of pyruvate carboxylase → ↓ gluconeogenesis Answer: C | Blood [Glycerol] SAME | Muscle FA oxidation ↓ | Brain glucose SAME | Liver gluconeogenesis ↓ | ✅ All outcomes are logical and match.

3. Inhibitor of Muscle Pyruvate Dehydrogenase (PDH)

PDH function: Converts pyruvate to acetyl-CoA in muscle. Inhibition effect:

  • Muscle can’t oxidize glucose efficiently → ↑ glucose in blood → ↓ glucose uptake by muscle → more available to brain
  • Brain ↑ glucose oxidation due to more available glucose
  • May increase muscle reliance on fatty acids → possibly ↑ FA oxidation
  • Liver gluconeogenesis unchanged (PDH inhibition is in muscle, not liver) Answer: D | Blood [Glycerol] ↓ | Muscle FA oxidation ↑ | Brain glucose oxidation ↑ | Liver gluconeogenesis SAME | ✅ Matches: PDH inhibition increases glucose availability → brain uses more. Also, muscle shifts to FA usage.

Final Matching Table:

Treatment Matched Outcome Explanation Summary
1. Inhibitor of Hormone Sensitive Lipase A ↓ Lipolysis → ↓ glycerol, ↓ muscle FA use, ↓ gluconeogenesis (↓ glycerol substrate)
2. Inhibitor of Carnitine Acyl Transferase C ↓ FA oxidation in muscle, ↓ gluconeogenesis (↓ acetyl-CoA activation of PC)
3. Inhibitor of Muscle Pyruvate Dehydrogenase D Muscle uses more fat, brain uses more glucose (more available), gluconeogenesis unaffected

L7: Regulation, Enzymes and Rate Limiting Steps


1. Describe the chemical features of ATP which make it ideal for use as an energy currency

Explanation:

  • ATP contains three phosphate groups, each with high-energy phosphoanhydride bonds.

  • Hydrolysis of ATP → ADP or AMP releases substantial free energy (~-30.5 kJ/mol).

  • Phosphoryl group transfer is kinetically stable but thermodynamically favourable.

  • ATP is soluble, abundant (~5 mM), and universally recognised by enzymes.

📘 ATP structure and energy – Khan Academy


2. Explain the concept of energy charge with reference to the concentration of adenine nucleotides

Explanation:

  • Energy charge reflects cellular energy status:

    \(\text{Energy Charge} = \frac{[ATP] + 0.5[ADP]}{[ATP] + [ADP] + [AMP]}\)

  • Range: 0 (all AMP) → 1 (all ATP).

  • Cells maintain charge ~0.9; slight ATP changes → large AMP response.

  • Acts as a metabolic gauge, especially through AMP-activated protein kinase (AMPK).

📘 Energy charge – Biochemistry LibreTexts


3. Review how a small change in ATP concentration is translated into a large relative change in AMP concentration

Explanation:

  • Via the action of adenylate kinase:

    \(2 ADP ⇌ ATP + AMP\)

  • Minor ATP depletion → large increase in AMP, which potently activates AMPK and other enzymes.

  • AMP is the most sensitive indicator of energy stress.

📘 Adenylate kinase role – PubMed


4. Identify the most likely control points in metabolic pathways

Explanation:

  • Control is focused on irreversible steps (far from equilibrium).

  • Often the first committed step in a pathway.

  • Enzymes working at saturation (low Km, high [S]) are ideal control points.

📘 Control points in glycolysis – Wikipedia


5. Interpret enzyme kinetic parameters to identify potential rate-limiting steps

Explanation:

  • RLS enzymes typically:

    • Have low Km (high affinity)

      • don’t have high reaction velocity

      • Enzymes (that we’re trying to control flux of) are generally working at substrate concentration that are much higher than their Km value.

      • so, changes in substrate concentration on high end (where plateau is) don’t change the rate of reaction –> already plenty of substrate

      • Enzyme working at max velocity trying to turn substrates into products

      • substrates basically just waiting there —> giving more substrate or taking some away doesn’t change rate of reaction

      • only time that substrate concentration does affect an enzyme to catalyse a reaction is when concentration gets close to Km value.

      • Km is the substrate concentration at which the enzyme is catalysing the reaction at half of the max (half of max reaction velocity).

    • Operate near Vmax

    • Show strong saturation

    • Are regulated allosterically or by covalent modification

📘 Vmax and Km visualisation – Lumen Learning


6. Describe the properties of rate limiting steps

Explanation:

  • Irreversible under physiological conditions.

  • Control flux through the pathway.

  • Sites of feedback and feedforward regulation.

  • Often committed steps unique to the pathway.

📘 RLS in metabolism – ScienceDirect


7. Review the major ways in which enzyme activity can be changed

Source: L3.1 pg 11

Explanation:

  • Allosteric regulation: ligand binding at non-active site → conformational change (e.g., AMP, citrate).

  • Covalent modification: phosphorylation/dephosphorylation alters activity (e.g., PDH).

  • Isoenzyme expression: tissue-specific variants.

  • Transcriptional control: change enzyme levels.

From lecture:

  1. Change intrinsic activity of the step: make rate limiting enzyme go faster/slower

  2. Switch on/off: turn the rate limiting enzyme on/off or make it work the other way around

  3. Make and destroy gates according to need: increase the rate of transcription/translation of the RLS, or change its rate of degradation

📘 Mechanisms of enzyme regulation – NCERT


8. List the key rate limiting steps in the major pathways of catabolism

Source: L3.1 pg 12

Examples:

Pathway RLS Enzyme
Glycolysis Phosphofructokinase-1 (PFK-1)
β-Oxidation Carnitine acyltransferase I
Krebs Cycle Isocitrate dehydrogenase
Glycogenolysis Glycogen phosphorylase
Pyruvate Oxidation Pyruvate dehydrogenase (PDH)

Possible rate limiting steps:

Fatty acid oxidation - need the availability of CoA in cytoplasm to trap FA in the cell

Beta oxidation - need availability of carnitine to transport FA-CoA from cytoplasm into the mitochondria martrix

Beta oxidation - dehydrogenases working above Km (working at max rate to break FA into 2-carbon chunks)

Beta oxidation - availability of CoA, NAD+, or FAD

Glycolysis - transport of glucose into the cell using GLUT 2 or 4 (especially GLUT-4 which is insulin-sensitive)

Glycolysis - Hexokinase will stop trapping more glucose if we don’t use the phosphorylated glucose, allowing it to flow back out again

glycolysis - Phosphofructokinase (PFK) phosphorylating G6P to Glucose 1,6-bisphosphate

glycolysis - pyruvate kinase

glycolysis - NAD+ for glycolysis to continue. Unlike Fatty acid oxidation, glycolysis does not need FAD

Pyruvate oxidation - PDH

Pyruvate oxidation - CoA and NAD+, for PDH to convert into Ac-CoA it will need a CoA and this step also converts one NAD+ to NADH

glycogenolysis - glycogen phosphorylase which takes glucose off glycogen

📘 [Table – Lehninger Principles of Biochemistry]


9. Provide an overview of the regulation of phosphofructokinase (PFK)

Source: L3.1 pg 13 - graph on this page

Explanation:

Allosteric means site away from active site

  • Allosterically inhibited by:

    • ATP (substrate and inhibitor) –> i.e. it doesn’t really like ATP

      • at physiological [ATP], PFK rx rate very low

      • but if AMP is added (even in tiny amounts) now PFK activity greatly activated

    • Citrate (signal of Krebs cycle activity)

  • How inhibition works:

    • If there’s plenty of glycolysis –> plenty pyruvate from glycolysis –> pyruvate turning to Ac-CoA to enter Krebs –> gets converted to citrate –> lots of citrate = lots of Ac-CoA comiing into Krebs –> so citrate makes its way out into cytoplasm and tells PFK to switch glycolysis off
  • Activated by:

    • AMP (signals low energy)

    • Fructose-2,6-bisphosphate (potent liver activator)

  • How it works:

    • changes the shape of the active site so that the active site would have a higher affinity to the substrate
  • Classic control point of glycolysis.


10. Provide an overview of the regulation of hexokinase

Source: L3.1 pg 14

Explanation:

  • Inhibited by product G6P (negative feedback).

  • Prevents excess glucose trapping.

  • Allows glucose to exit cell via GLUT2 if not needed.

  • Liver enzyme glucokinase is not inhibited by G6P, enabling storage.

📘 Hexokinase vs glucokinase – Osmosis


11. Using an example, illustrate how control motifs act synergistically to regulate pathways

Source: L3.1 pg 15 - diagram here

Example:

At rest:

  • glucose converted to G6P by hexokinase but since we’re restinf not much glycolysis is performed

  • causes build up of G6P which gives negative feedback loop to inhibit hexokinase

  • we stop trapping glucose as G6P

  • energy charge is high when resting and lots of ATP but no AMP

  • energy charge will act on PFK

  • PFK gets switched off since there’s not much AMP around and it doesn’t like ATP

  • PK also gets switched off

During exercise, falling ATP and rising AMP:

  • Using up trapped G6P so inhibition on Hexokinase is relieved

  • energy charge switched from high ATP to high AMP

  • Activate PFK-1 allosterically to switch on glycolysis

  • Inhibit PDH kinase → activates PDH

  • feedforward fructose 1,6-bisphosphate onto last enzyme in glycolysis Pyruvate kinase (i.e. its telling PK to hurry up)

  • All work together to upregulate glycolysis and fuel oxidation.

📘 AMPK and exercise – Cell Metabolism

Also pg 16:

The Role of Mitochondrial Fat Oxidation in Cancer Cell Proliferation and Survival - Scientific Figure on ResearchGate. Available from: https://www.researchgate.net/figure/The-key-enzymes-determining-the-competition-of-glycolysis-versus-mitochondrial-fatty-acid_fig1_347695455 [accessed 5 May, 2022]

Figure 1. The key enzymes determining the competition of glycolysis versus mitochondrial fatty acid oxidation (FAO) defined by the Randle cycle. (A) Increased extracellular free fatty acids availability (FFA) can increase mitochondrial fatty acid oxidation (FAO), resulting in higher levels of acetyl-CoA, NADH, and ATP inside the mitochondria.

As a result, citrate production by citrate synthase (CS) is increased, but the activity of PDH and TCA cycle dehydrogenases is decreased by the elevation in acetyl CoA/CoA, NADH/NAD+, and ATP/ADP ratios. Thus, FAO leads to an accumulation of mitochondrial citrate, which is exported to the cytosol via the CIC/SLC25A1 exporter.

High citrate in the cytosol can inhibit phosphofructokinase-1 (PFK-1) and pyruvate kinase (PK) activities, decreasing glycolysis, pyruvate synthesis and oxidation. (B) When extracellular glucose increases, the concomitant upregulation in glycolysis provides more pyruvate to the mitochondria. Pyruvate oxidation by PDH and pyruvate carboxylation by pyruvate carboxylase (PC) generate acetyl-CoA and OAA respectively, increasing citrate synthase (CS) activity.

The amount of citrate produced is higher than needed to sustain the carbon pool of TCA cycle intermediates, which causes its export to the cytosol. Under high glucose, cytosolic citrate is hydrolyzed by ATP-citrate lyase (ACLY) into acetylCoA and OAA. Acetyl-CoA is carboxylated by two acetyl-CoA carboxylases, ACC1 and ACC2, to generate malonyl-CoA. ACC2 is localized in the outer mitochondrial membrane, causing malonylCoA to be more accessible to carnitine-palmitoyl transferase 1 (CPT1). Malonyl-CoA inhibits CPT1


12. Using an example, show how enzymes are controlled by reversible phosphorylation

Source: L3.1 pg 17

Example: PDH Regulation

  • PDH kinase phosphorylates PDH → inactivates it.

    • PDH kinase itself is activated by high levels Ac-CoA
  • PDH phosphatase dephosphorylates → reactivates PDH.

    • PDH phosphotase signalled by insulin
  • Controlled by energy status (acetyl-CoA, NADH levels).

📘 PDH regulation – KEGG Pathway


13. Recognise that rate limiting steps can change with circumstances

Explanation:

  • RLS are context-dependent:

    • Fasted vs fed state

    • Rest vs exercise

    • Oxygen availability

  • Example: In fasting, carnitine shuttle becomes limiting for β-oxidation.

  • generally not ‘availability of substrates’ because we are demand driven not supply driven

    • demand = want/ don’t want, supply = enough/ not enough

    • normally every cell gets some glucose through GLUT-1 which is on every cell in body. Therefore, will always have SOME supply to a degree so it comes down to whether or not we NEED it

  • can be ‘transport & trapping’ –> maybe need CoA to trap glucose inside cell. maybe hexokinase need ATP to trap glucose inside cell

  • can be ‘transport into mitochondria’ –> so to get FA into matrix we need carnitine (which is the RLS)

  • can be ‘having enough gates (enzymes) at each step’ –> to get all the way through pathway

    • in ETC: oxygen is terminal electron acceptor. So if cells doing lots and lots of work, supply of oxygen can be RLS

IN GENERAL: usually dictated by AMP level and availbility of NAD+, FAD, CoA, or whatever cofactors required by these enzymes. NAD+ and FAD are controlled by the cellular demand for ATP. If there’s no demand for ATP –> NADH and FADH2 stuck holding H/e- –> can’t drop off at ETC until some ATP is used.

📘 Dynamic RLS in metabolism – PMC


14. Explain the principles of reciprocal regulation of pathways

Explanation:

  • Opposing pathways are regulated in opposite ways:

    • E.g., glycolysis vs gluconeogenesis

    • β-oxidation vs fatty acid synthesis

  • Prevents futile cycling.

  • Often mediated by shared molecules (e.g., citrate, fructose 2,6-bisphosphate).

LOOK AT L3.1 pg20 for example with citrate

The regulatory role of citrate in the metabolism. Citrate is synthesized inside the mitochondria by citrate synthase from acetyl-CoA and OAA. It is exported outside the mitochondria by CIC. Citrate inhibits PFK1, PK, PDH, and SDH. Citrate inhibits also PFK2, which produces F2,6P, an allosteric activator of PFK1 in cancer cells. Activation effect is exerted on lipid biosynthesis through ACC, which produces malonyl-CoA, the first product of lipid biosynthesis, which, in turn, inhibits the CPT-1, the first enzyme of β -oxidation process. Through F1,6BPase, citrate stimulates gluconeogenesis. ACC, acetyl-CoA carboxylase; CS, citrate synthase; F1,6BPase, fruc- tose 1,6 bisphosphatase; G6P, glucose-6-phosphate; F6P, fructose- 6-phosphate; HK, hexokinase; PEP, phosphoenolpyruvate. Symbols + and – indicate stimulation and inhibition, respectively.

Citrate - new functions for an old metabolite - Scientific Figure on ResearchGate. Available from: https://www.researchgate.net/figure/The-regulatory-role-of-citrate-in-the-metabolism-Citrate-is-synthesized-inside-the_fig1_259825846 [accessed 5 May, 2022]


L8: Gluconeogenesis in Detail


1. Define the key steps involved in the conversion of 3-carbon intermediates to 6-carbon intermediates in gluconeogenesis

Source: L3.2 pg 2, 3

Explanation:

Gluconeogenesis: From 3-Carbon to 6-Carbon Intermediates

Gluconeogenesis synthesizes glucose (6-carbons) from smaller carbon units, especially 3-carbon intermediates like pyruvate and lactate.

Here’s how 3-carbon intermediates are converted to 6-carbon sugars:

  • 2 × pyruvate → 2 × oxaloacetate (OAA) via pyruvate carboxylase (mitochondrial)

    • OAA → 2 × phosphoenolpyruvate (PEP) via PEPCK

    • After several steps: 2 × triose phosphates → fructose 1,6-bisphosphate → F6P → G6P

  • The process bypasses irreversible glycolysis steps (hexokinase, PFK-1, pyruvate kinase).


Stepwise Process (with opposing glycolytic enzymes shown in pink)

Step 1: Pyruvate to Phosphoenolpyruvate (PEP)

  • Enzymes:

    1. Pyruvate carboxylase (in mitochondria):

      • Pyruvate + CO₂ + ATP → Oxaloacetate (OAA) + ADP
    2. PEP carboxykinase (in cytosol):

      • OAA + GTP → PEP + CO₂ + GDP
  • Opposes: Pyruvate kinase (glycolysis, irreversible)


Step 2: PEP to 1,3-Bisphosphoglycerate → Glyceraldehyde-3-phosphate (G3P)

  • Multiple reversible steps occur here (opposite of glycolysis), involving:

    • Conversion of PEP → 2-phosphoglycerate → 3-phosphoglycerate → 1,3-bisphosphoglycerate → G3P

    • NADH + H⁺ used to reduce 1,3-BPG to G3P (reversal of glycolysis)


Step 3: Two 3-carbon G3P molecules → Fructose-1,6-bisphosphate (F1,6BP)

  • Via aldolase, two G3Ps are combined (via DHAP intermediate) to form:

    \[ \text{G3P} + \text{DHAP} \rightarrow \text{Fructose-1,6-bisphosphate} \]

  • Now we have a 6-carbon intermediate


Step 4: Fructose-1,6-bisphosphate → Fructose-6-phosphate

  • Enzyme: Fructose-1,6-bisphosphatase

  • Opposes: Phosphofructokinase-1 (PFK-1) in glycolysis


Step 5: Fructose-6-phosphate → Glucose-6-phosphate → Glucose

  • Final steps:

    • Fructose-6-P → Glucose-6-P (reversible isomerization)

    • Glucose-6-P → Glucose via Glucose-6-phosphatase

  • Opposes: Hexokinase in glycolysis


Summary Table: Gluconeogenesis vs Glycolysis Key Steps

Step Gluconeogenesis (Blue) Glycolysis (Pink) Notes
1 Pyruvate → OAA → PEP (pyruvate carboxylase, PEPCK) PEP → Pyruvate (pyruvate kinase) Bypasses irreversible step
2 1,3-BPG → G3P (uses NADH) G3P → 1,3-BPG (makes NADH) Reversible
3 G3P + DHAP → F1,6BP (aldolase) Same Reversible
4 F1,6BP → F6P (F1,6-bisphosphatase) F6P → F1,6BP (PFK-1) Bypasses irreversible step
5 G6P → Glucose (G6-phosphatase) Glucose → G6P (hexokinase) Final bypass step

Key Point:

The conversion of two 3-carbon molecules (G3P/DHAP) into a single 6-carbon intermediate (F1,6BP) is a central milestone in gluconeogenesis, opposing the breakdown of glucose in glycolysis.


2. Review the steps involved in the release of glucose from cytosolic glucose 6-phosphate

Source: L3.2 pg 4 –> ins diagram

Explanation:

  • To get G6P converted back to glucose –>

  • G6P enters the ER lumen via a transporter (T1).

  • Hydrolysed by glucose-6-phosphatase → glucose + Pi.

  • Glucose exits via T3; released into blood via GLUT2.

  • Only occurs in liver and pancreas.

📘 [G6Pase system – Lehninger Figure 15-28]


3. Analyse the effects of the absence of the 2-OH group in glucose and generate rationales for the use of 2-deoxy derivatives in clinical diagnostics

Source: L3.2 pg 7

Explanation:

  • 2-Deoxyglucose (2-DG) lacks a 2’-OH → cannot be isomerised to F6P.

  • Trapped in cells as 2-DG-6P.

  • Used in FDG-PET imaging to track glucose uptake (e.g. in cancers).

  • Accumulation inhibits glycolysis (Warburg effect exploited).

📘 FDG-PET application – Radiopaedia


4. Predict the flow of glycolysis and gluconeogenesis based on the relative activities of phosphofructokinase and fructose 1,6 bisphosphatase

Explanation:

  • PFK-1 activity → glycolysis.

  • F1,6-BPase activity → gluconeogenesis.

  • Fructose 2,6-bisphosphate regulates both:

    • ↑ F26BP → activates PFK-1, inhibits F1,6-BPase → glycolysis.
    • ↓ F26BP → opposite → gluconeogenesis.

📘 Diagram – F26BP control loop (Biochem Text)


5. Compare the regulators of phosphofructokinase and fructose 1,6 bisphosphatase

Explanation:

Regulator Effect on PFK-1 Effect on F1,6-BPase
ATP Inhibits
AMP Activates Inhibits
Citrate Inhibits
Fructose 2,6-BP Strong activator Strong inhibitor

📘 [Comparison – Stryer Biochemistry, Ch. 16]


6. Outline the reasons to produce fructose 2,6 bisphosphate

Explanation:

  • F26BP is a key switch:

    • Activates glycolysis (PFK-1)

    • Inhibits gluconeogenesis (F1,6-BPase)

  • Synthesised by PFK-2, degraded by F2,6-BPase

  • Allows rapid hormonal control (glucagon vs insulin).

📘 [Visual Summary – Figure 15-17, Lehninger]


7. Apply knowledge of the effects of fructose 2,6 bisphosphate to the regulation of PFK and F16BPase

Source: L3.2 pg 10-16

Explanation:

  • ↑ F26BP:

    • Lowers Km of PFK-1 for F6P → ↑ glycolysis

    • Inhibits F1,6-BPase → ↓ gluconeogenesis

  • ↓ F26BP: reverse occurs

  • Rapidly adjusts metabolism based on fed vs fasting state.

Fructose 2,6-Bisphosphate (F2,6BP) – The Key Regulator

F2,6BP is a powerful regulatory molecule that controls the balance between glycolysis and gluconeogenesis by affecting two opposing enzymes:

Glycolysis Enzyme: Phosphofructokinase-1 (PFK-1)
  • PFK-1 function: Converts fructose-6-phosphate → fructose-1,6-bisphosphate

  • F2,6BP effect: Activates PFK-1

    This speeds up glycolysis, helping the cell break down glucose for energy.

Gluconeogenesis Enzyme: Fructose-1,6-bisphosphatase (F1,6BPase)
  • F1,6BPase function: Converts fructose-1,6-bisphosphate → fructose-6-phosphate
  • F2,6BP effect: Inhibits F1,6BPase 🚫 This slows down gluconeogenesis, preventing the cell from making new glucose.
Simple Summary:
Molecule Target Enzyme Effect Pathway Affected
F2,6BP PFK-1 ✅ Activates ↑ Glycolysis
F2,6BP F1,6BPase 🚫 Inhibits ↓ Gluconeogenesis

💡 Why it matters:

F2,6BP acts like a switch:

  • When energy is needed, it turns glycolysis ON and gluconeogenesis OFF.

  • This helps the cell avoid doing both pathways at once, which would waste energy.


8. Illustrate the dual identity of PFK/F26BPase through phosphorylation changes

Source: L3.2 pg 10-16

Explanation:

  • PFK-2 and F26BPase are parts of the same bifunctional enzyme.

  • Phosphorylation by PKA (activated by cAMP/glucagon) → activates F26BPase.

  • Dephosphorylation (insulin) → activates PFK-2.

  • A classic example of reciprocal regulation.

📘 [Bifunctional enzyme mechanism – Nature Fig. 16-31]


9. Outline the consequences of changes in PFK/F26BPase activity

Source: L3.2 pg 10-16

Explanation:

  • Fed state (insulin):

    • Dephosphorylation → ↑ F26BP → glycolysis ↑
  • Fasting (glucagon):

    • Phosphorylation → ↓ F26BP → gluconeogenesis ↑
  • Allows the liver to switch rapidly between pathways.


10. Compare the conversion of pyruvate to phosphoenolpyruvate in gluconeogenesis to the opposite process in glycolysis

Explanation:

Process Enzymes Involved Regulation
Glycolysis Pyruvate kinase Inhibited by ATP, alanine
Gluconeogenesis Pyruvate carboxylase (PC) + PEPCK PC activated by acetyl-CoA
  • Involves 2 steps and ATP/GTP expenditure.

  • Pyruvate –> phosphoenolpyruvate: require 2 enzymes to get back

    • pyruvate carboxylase - stimulated by Ac-CoA

    • PEP pyruvate carboxykinase - quickly turns the 4-carbon oxaloacetate (OAA) into a 3-carbon and also adds phosphate group as well

Summary: getting pyruvate back to PEP - going from 3-carbon pyruvate –> pyruvate carboxylase uses CO2 (from bicarbonate) to join onto pyruvate –> taking CO2 back off again (using different enzymes (using GTP) and adds a phosphate.


11. Predict the effect of fatty acid oxidation on glucose fluxes as a consequence of its effects on pyruvate carboxylase

Explanation:

  • FA oxidation → ↑ acetyl-CoA → activates pyruvate carboxylase

    • which is the first enzyme in gluconeogenesis

    • and forms more OAA which can potentially increase the number of Krebs cycles performed

  • Also inhibits PDH, limiting glucose oxidation

  • Promotes gluconeogenesis during fasting


12. Evaluate the importance of anaplerosis of Krebs Cycle intermediates

Explanation:

  • Anaplerosis = replenishment of Krebs cycle intermediates.

  • OAA pulled out for gluconeogenesis must be replaced.

  • Pyruvate → OAA (via PC) serves both gluconeogenesis and anaplerosis.

  • Critical during exercise, starvation, or rapid biosynthesis.

📘 Anaplerosis and metabolic flux – PMC


L9: Glycemic Responses and Glycogenesis (Glucose Disposal Part 1)


1. Dynamics of post-prandial glucose disposal and the key hormones involved

Explanation:

  • After a meal (~50g glucose), blood glucose spikes and must be cleared rapidly to prevent toxicity.

  • The key players:

    • Insulin (from pancreatic β-cells): promotes glucose uptake and storage.

    • Liver acts as a “glucose sponge” via GLUT2.

    • Muscle and WAT take up glucose via insulin-responsive GLUT4 transporters.

  • Target: return glucose to ~5 mM within 2 hours.

  • Post-prandial glucose disposal is biphasic: initial hepatic uptake followed by peripheral tissue clearance.

📘 Video: Post-prandial glucose regulation


2. Understand the post-prandial glucose responses for subjects with glucose intolerance, insulin resistance and diabetes

Explanation:

Condition Fasting Glucose Insulin Response Clearance Speed
Tolerant ~5 mM Normal, dynamic Fast (returns in ~2 h)
Intolerant Normal High (ineffective) Slow
Insulin resistant Normal or high Very high Sluggish
Type 2 Diabetes High Impaired Severely delayed
  • Higher and prolonged glucose exposure → more glycation → more damage (e.g., HbA1c increase).

📘 HbA1c and glucose tolerance – CDC


3. Understand what glycemic index is, how it is measured and how it is clinically useful

Explanation:

  • Glycemic Index (GI) = (AUC test food / AUC glucose) × 100

  • Test: 50g available carbohydrate → measure blood glucose over 2 hours.

  • High GI: rapid rise/fall (e.g., white bread, amylopectin-rich).

  • Low GI: slower rise/fall (e.g., legumes, amylose-rich foods).

  • Clinically useful in managing diabetes, appetite, and long-term risk of metabolic disease.

📘 Glycemic index explained – Harvard School of Public Health


4. Appreciate the role of different glucose transporters in different tissues and how these function

Explanation:

Transporter Location Features
GLUT1 All cells Basal glucose uptake
GLUT2 Liver, β-cells High capacity, low affinity; bidirectional
GLUT4 Muscle, WAT Insulin-dependent; translocates to membrane
  • Insulin → GLUT4 translocation → increased uptake in muscle and fat.

  • Liver uses GLUT2, independent of insulin.

📘 GLUT transporters visual – Osmosis


5. Review the chemical structure of glycogen and the chemical strategy for its synthesis

Source L3.3 pg 15, 16 - diagram here

Explanation:

  • Glycogen = branched polymer of glucose, α-1,4 chains with α-1,6 branches.

  • we’d love to build up muscle glycogen stores

    • so you’ve got 2 choices: Glucose comes in and phosphorylated by hexokinase, trapped as G6P, then:

      • go down glycolysis to give ATP

      • get converted to glucose 1-phosphate through isomerisation reaction. it then needs to be activated if we want it to turn into glycogen storage. This activation requires UTP

  • Synthesised from UDP-glucose, which is formed using UTP + G1P.

  • Glycogen synthase adds glucose units (C1→C4).

  • Branching enzyme creates new α-1,6 linkages.

This is the story:

  • got UTP coming in. UTP carries 3 phosphates and we’re working on G1P which already has 1 phosphate. So, together there’s 4 phosphates involved.

  • Now 2 phosphates are released as PPi (Pyrophosphate). So now we have the 1 phosphate on G1P and also the remaining one that’s now on UMP (becomes monophosphate since it lost 2 phosphates)

  • Then these join together to give UDP glucose (diphosphate joined to glucose)

  • So we’re adding UDP to glucose to prepare it to join onto the glycogen chains. And it specifically joins on the ends of the branches

  • So now, when UDP glucose gets joined onto the glycogen chain it just takes the glucose part and now we’re left with the UDP

  • The UDP left behind can now go through that process again. But we need some UTP so we use ATP to convert the UDP back into UTP. So it gets converted by taking the phosphate from ATP, so now our ATP becomes ADP.

    • So we have UDP and use ATP –> UTP and ADP
  • glycogen needs branching otherwise it will end up as one long linear chain. So the enzyme ‘glycogen-branching enzyme’ will cut the glycogen chain and join it back as a branch

📘 Glycogen structure & synthesis – LibreTexts


6. Understand how increased glycogen synthesis stimulates glycolysis

Explanation:

  • Glycogenesis is anabolic: uses ATP → reduces energy charge → ↑ AMP/ADP

  • This drop activates phosphofructokinase (PFK-1).

  • Therefore, insulin indirectly stimulates glycolysis by stimulating storage processes that lower ATP.

📘 Coupling of glycogenesis and glycolysis – Nature Education

🔁 How Increased Glycogen Synthesis Stimulates Glycolysis

📌 Key Idea:

Glycogenesis (glycogen synthesis) is an energy-consuming anabolic process, and this energy demand indirectly stimulates glycolysis, a catabolic pathway that provides ATP.


✅ Step-by-Step Explanation:

1. Insulin activates glycogen synthase (GS):
  • Insulin → activates Protein Phosphatase 1 (PPI)

  • PPI dephosphorylates glycogen synthaseactivates GS

  • Result: ↑ Glycogen synthesis


2. Glycogen synthesis consumes energy:
  • Forming UDP-glucose and linking glucose to glycogen requires ATP

  • This lowers ATP, raises ADP and AMP levels

  • ↓ ATP / ↑ AMP = low energy charge


3. Low energy charge activates PFK-1 (rate-limiting enzyme of glycolysis):
  • PFK-1 is not directly activated by insulin

  • But it’s allosterically activated by AMP, which rises due to ATP usage

  • Result: ↑ Glycolytic flux → more ATP produced


🔄 Coupling Explained Simply:

When insulin tells the cell to store glucose as glycogen (an energy-expensive task), the drop in ATP signals the cell to burn some glucose for energy — stimulating glycolysis.

This is a beautiful example of metabolic balance:

  • Anabolism (storage) pulls on catabolism (energy production)

  • Insulin, though anabolic, causes some glucose to be burned to fuel the storage process


🧠 Final Summary:

Pathway Triggered by Effect
Glycogen synthesis Insulin → PPI → Active GS Uses ATP
Glycolysis AMP ↑ → Activates PFK-1 Makes ATP
Coupling effect Energy demand from glycogen synthesis indirectly stimulates glycolysis

7. Explain the consequences of the different activities of hexokinase and glucokinase

Explanation:

Feature Hexokinase Glucokinase
Tissue All cells Liver and β-cells
Substrate range Any 6C sugar Only glucose
Km for glucose Low (~0.1 mM) → high affinity High (~10 mM) → low affinity
Inhibition Inhibited by G6P Not inhibited by G6P
Saturation Easily saturated Active only at high [glucose]
  • Glucokinase allows liver to act as a glucose buffer.

📘 Glucokinase vs Hexokinase – Osmosis


8. Summarise the similarities and differences in glycogenesis in different tissues (liver vs muscle)

Explanation:

Feature Liver Muscle
Uptake transporter GLUT2 (insulin-independent) GLUT4 (insulin-dependent)
Hexokinase isoform Glucokinase (high Km, not inhibited by G6P) Hexokinase (low Km, G6P-inhibited)
Regulation style Push – high [glucose] → passive uptake Pull – insulin stimulates GLUT4 + GS
GS regulation Stimulated by glucose and G6P Stimulated by insulin-dependent GS action
Function Maintains blood glucose Fuel for contraction

📘 [Glycogenesis in liver vs muscle – Lehninger Ch. 15]


L10: Lipogenesis & Pentose Phosphate Pathway


1. Recall the overall scheme for the synthesis of fatty acids from glucose

Explanation:

  • Glucose → G6P → pyruvate via glycolysis

  • Pyruvate enters mitochondria → converted to acetyl-CoA by PDH

  • Acetyl-CoA condenses with OAA → citrate

  • Citrate exported → cleaved by ATP-citrate lyase (ACL) → cytosolic acetyl-CoA

  • Acetyl-CoA → malonyl-CoA (via ACC)fatty acid synthesis (FAS)

  • Requires: NADPH (from PPP), ATP, glycerol-3-phosphate (from glycolysis)

📘 Video: Overview of Lipogenesis


2. Understand the reaction catalysed by and the regulation of acetyl CoA carboxylase (ACC)

Reaction:

\[ \text{Acetyl-CoA} + \text{HCO₃⁻} + \text{ATP} \xrightarrow{\text{ACC}} \text{Malonyl-CoA} + \text{ADP} + \text{Pi} \] - taking Ac-CoA and adding CO2 (from bicarbonate) to form 3-carbon chain called malonyl-CoA

  • conversion step requires vitamin called biotin and uses some ATP

  • conversion also requires cofactor magnesium

  • basically an activated/primed ‘Ac-CoA’ ready for lipogenesis

Regulation:

  • Activated by insulin (via dephosphorylation)

    • insulin activates the phosphotase to perform dephosphorylation on ACC which activates ACC

    • so insulin is telling cells to convert Ac-CoA to malonyl-CoA for fatty acid synthesis.

  • Allosterically activated by citrate (forms polymeric filaments)

    • If lots of citrate enter cytoplasm where FA synthesis occurs –> ACC activated to convert Ac-CoA to malynol-CoA
  • Inhibited by fatty acyl-CoA (feedback inhibition)

    • If we’ve made lots of fatty acid chains –> we should allow those long fatty acid chains to feed back and tell the first enzyme in the process to stop converting Ac-CoA into malynol-CoA because we’ve got enough FA right now.

📘 Biochemical pathway – ACC regulation


3. Describe the reaction sequence of fatty acyl synthase (FAS)

Source: L4.1 pg 6

Explanation:

Lipogenesis is basically reduction then dehydration then reduction again (to add on 2-carbon chains at a time)

  • Large multifunctional enzyme complex

  • Key steps (per 2C addition cycle):

    1. Loading: Acetyl group + Malonyl group attached to ACP (FAS binds to its substrates)

    2. Condensation - malynol will be decarboxylated so the CO2 comes off

    3. Reduction (NADPH) - negative charge will attack at the cabonyk carbon and join itself onto the chain, maing it 4-carbon long

    4. Dehydration

    5. Second reduction (NADPH)

    6. Chain transfer and repeat

  • Final product: Palmitate (C16) → released when chain reaches 14–18 carbons

📘 Diagram – Fatty Acid Synthase Cycle


4. Apply knowledge of the regulators of ACC to different physiological situations

Fed state (insulin present):

  • ACC activated → ↑ malonyl-CoAfat synthesis ON, fat oxidation OFF

Fasted/exercise (AMPK active):

  • AMPK phosphorylates ACC → inactive → ↓ fat synthesis, ↑ β-oxidation

📘 AMPK and ACC regulation – ScienceDirect

The Fatty Acid Synthase (FAS) complex catalyzes the de novo synthesis of palmitate (C16:0) from acetyl-CoA and malonyl-CoA, using NADPH as a reducing agent. This process occurs in the cytosol and consists of a cyclic reaction sequence repeated 7 times to build the 16-carbon saturated fatty acid.


🧬 Overview of FAS Reaction Sequence:

  1. Loading phase

  2. Condensation

  3. Reduction

  4. Dehydration

  5. Reduction

  6. Chain transfer

  7. Repeat cycle

  8. Termination


🔁 Step-by-Step Reaction Sequence of Fatty Acid Synthase:

Step Enzyme Domain Substrate / Action Product / Outcome
1. Loading - Acetyl transferase (AT) loads acetyl-CoA onto the KS domain (β-ketoacyl synthase)
- Malonyl transferase (MT) loads malonyl-CoA onto ACP (acyl carrier protein)
Acetyl-KS and Malonyl-ACP formed
2. Condensation β-Ketoacyl Synthase (KS) Acetyl group + Malonyl group → 4-carbon β-ketoacyl-ACP + CO₂ (decarboxylation drives the reaction) β-ketobutyryl-ACP
3. Reduction β-Ketoacyl reductase (KR) Reduces β-keto group to hydroxyl using NADPH β-Hydroxyacyl-ACP
4. Dehydration Dehydratase (DH) Removes H₂O → forms a double bond (trans-Δ²-enoyl-ACP) Enoyl-ACP
5. Reduction Enoyl reductase (ER) Reduces double bond using NADPH → saturated acyl-ACP Butyryl-ACP (4-carbon chain)
6. Chain Transfer Transfer the growing acyl chain from ACP to KS, freeing ACP for the next malonyl-CoA Butyryl-KS formed
7. Repeat Another malonyl-CoA is loaded to ACP, and steps 2–6 repeat 6 more times Adds 2C per cycle until 16C palmitoyl-ACP
8. Termination Thioesterase (TE) cleaves palmitate from ACP Final product: Palmitate (C16:0)

🧠 Key Inputs & Outputs:

  • Inputs per cycle: 1 acetyl-CoA (start), 7 malonyl-CoA, 14 NADPH

  • Final product: Palmitate (C16:0)

  • Byproducts: CO₂, H₂O, NADP⁺


🧾 Summary Statement:

Fatty Acid Synthase builds a 16-carbon saturated fatty acid through 7 cycles of condensation, reduction, dehydration, and reduction, using malonyl-CoA as the 2-carbon donor and NADPH as the reducing power, terminating in palmitate release.


5. Describe the process of esterification

Explanation:

  • 3 fatty acyl-CoA + glycerol-3-phosphate → triacylglycerol (TAG)

  • Requires FA-CoA, not free FA

  • G3P from:

    • Liver: via glycerol kinase

    • Adipose: from glycolysis (DHAP → G3P)

  • Esterification enzymes and FAS are upregulated by insulin

📘 TAG synthesis pathway – LibreTexts

🔬 Biological Esterification of Fatty Acids: Formation of Triacylglycerol (TAG)

The esterification process you’re referring to is the biosynthesis of triacylglycerol (TAG), where three fatty acyl-CoA molecules are esterified to a glycerol backbone (specifically glycerol-3-phosphate) to form a TAG (also called a triglyceride).


🧬 Overall Reaction:

\[ \text{Glycerol-3-phosphate} + 3\ \text{Fatty acyl-CoA} \longrightarrow \text{Triacylglycerol (TAG)} + 3\ \text{CoA-SH} + \text{Pi} \]


🔄 Step-by-Step Process:

Step 1: Activation of fatty acids
  • Fatty acids are first “activated” by linking to Coenzyme A (CoA):

    \[ \text{Fatty acid} + \text{CoA} + \text{ATP} \rightarrow \text{Fatty acyl-CoA} + AMP + PPi \]

  • This forms a high-energy thioester bond, making the fatty acid reactive for esterification.


Step 2: Formation of lysophosphatidic acid
  • The first fatty acyl-CoA is esterified to the sn-1 hydroxyl of glycerol-3-phosphate:

    \[ \text{Glycerol-3-P} + \text{Fatty acyl-CoA} \rightarrow \text{Lysophosphatidic acid} + \text{CoA-SH} \]


Step 3: Formation of phosphatidic acid (diacylglycerol-3-phosphate)
  • A second fatty acyl-CoA is esterified to the sn-2 hydroxyl:

    \[ \text{Lysophosphatidic acid} + \text{Fatty acyl-CoA} \rightarrow \text{Phosphatidic acid} + \text{CoA-SH} \]


Step 4: Dephosphorylation to diacylglycerol (DAG)
  • The phosphate group is removed from phosphatidic acid:

    \[ \text{Phosphatidic acid} \rightarrow \text{Diacylglycerol (DAG)} + \text{Pi} \]


Step 5: Final esterification to form TAG
  • A third fatty acyl-CoA is added to the sn-3 hydroxyl:

    \[ \text{DAG} + \text{Fatty acyl-CoA} \rightarrow \text{Triacylglycerol (TAG)} + \text{CoA-SH} \]


🔧 Enzymes Involved:

Step Enzyme
2 Glycerol-3-phosphate acyltransferase (GPAT)
3 1-Acylglycerol-3-phosphate acyltransferase (AGPAT)
4 Phosphatidic acid phosphatase (lipin)
5 Diacylglycerol acyltransferase (DGAT)

🧠 Summary:

TAG synthesis involves the sequential esterification of three fatty acyl-CoAs to a glycerol-3-phosphate backbone, forming a highly energy-dense molecule used for long-term energy storage in adipose tissue. This process occurs primarily in the liver and adipose tissue.


6. Integrate the pathways and processes that need to come together to support and supply lipogenesis

Pathways involved:

  • Glycolysis: glucose → pyruvate + G3P

  • PDH: pyruvate → acetyl-CoA

  • Krebs cycle: citrate formed → exported

  • ATP-citrate lyase: citrate → acetyl-CoA + OAA

  • ACC: acetyl-CoA → malonyl-CoA

  • PPP: provides NADPH

  • Gluconeogenesis (glyceroneogenesis): G3P production

📘 Integrated map – Stryer Biochemistry


7. Summarise the effect of insulin on the activity of pyruvate dehydrogenase

Explanation:

  • Insulin activates PDH phosphatase → dephosphorylates PDH → active

  • PDH converts pyruvate to acetyl-CoA, promoting substrate availability for lipogenesis

    • enters the Krebs cycle and allow it to convert to citrate which we can then transport out and get that CoA back

📘 PDH regulation – KEGG Pathway


8. Evaluate the probability of the different fates of acetyl CoA in lipogenic tissue

Since PDH activated, we will have lots of Ac-CoA forming

Two fates:

  1. Krebs cycle: oxidised to CO₂ + NADH (ATP production)
  • we require it to enter Krebs because we need it to fully oxidise and make lots of NADH to generate more ATP

  • we also need ATP for Malynol-CoA

  1. Lipogenesis: converted to fatty acids
  • firstly, need Ac-CoA to make its way into cytoplasm –> so we need to allow it to enter Krebs to join onto OAA to form citrate

  • this allows us to get the CoA back so that PDH can continue to convert pyruvate to Ac-CoA and it needs a CoA to do that

  • we also allow the formed citrate to flow back out into cytoplasm for lipogenesis

  • Decided by:

    • Energy status (ATP/AMP): High ATP → citrate exported

    • Hormonal state (insulin vs glucagon)

    • Availability of OAA for citrate formation

📘 Metabolic fate of acetyl-CoA – PMC


9. Explain the role of citrate in controlling the major anabolic and catabolic pathways

Source L4.1 pg 18 - diagram here

Cytosolic citrate:

  • Activates ACC (↑ fat synthesis)

  • Inhibits PFK-1 (↓ glycolysis)

  • Converted to acetyl-CoA + OAA (via ACL)

  • Converted to malonyl-CoA → inhibits CPT-1 → ↓ β-oxidation

From Lecture:

  • we want some ac-CoA out in the cytoplasm so we can use it as a 2-carbon building block

  • the CoA can’t cross membranes so we’ll allow it to enter Krebs cycle

  • it gets converted to citrate

  • citrate is transported out to cytoplasm

  • break the ac-CoA back off it again so now it becomes a 4-carbon OAA again

  • OAA gets reduced by NADH to give malate

  • malate gets converted to pyruvate to re-enter the mitochondria and in this process it gives us some NADPH

  • NADPH is the reductant needed to reduce FA in our cytoplasm

see diagram in lecture


10. Outline the mechanisms for the return of oxaloacetate to the mitochondria after citrate cleavage

Explanation:

  • OAA can’t cross the inner mitochondrial membrane

  • Converted to:

    • Malate, then back to OAA in mitochondria

    • Or pyruvate via malic enzyme (generates NADPH)

  • Pyruvate re-enters mitochondria → carboxylated to OAA by pyruvate carboxylase

📘 OAA shuttle diagram – Nature


11. Describe the purpose of the pentose phosphate pathway and how it fits into lipogenesis

Purpose:

  • Generates NADPH for reductive biosynthesis (e.g., lipogenesis)

  • Provides ribose-5-phosphate for nucleotide synthesis

Fit with lipogenesis:

  • Demand for NADPH ↑ → G6PDH (key PPP enzyme) activated

  • PPP enzymes return 3C and 6C sugars back to glycolysis

📘 PPP animation – YouTube


12. Illustrate the flow of substrates and key enzyme control points for lipogenesis

Uncategorised: Malonyl-CoA inhibits beta oxidation:

  • Malynol CoA inhibits CAT-1.

  • We’re inhibiting transport of FA into the mitochondria where they would undergo beta oxidation

  • we don’t want to be building FA while we’re oxidising FA inside the mitochondria.

  • so when ac-CoA activated by turning it into malynol-coA (activated by insulin), the malynol-CoA can inhibit CAT1

  • this stops us from transporting fatty acyl coA back into mitochondria as acyl carnitines

Key substrates and checkpoints:

Substrate Enzyme/Control Role
Glucose GLUT4/HK Entry/trapping
Pyruvate PDH Mitochondrial conversion
Citrate ACL Exported carbon source
Acetyl-CoA ACC Malonyl-CoA production (RLS)
NADPH G6PDH/PPP Reductant for FAS
G3P Glyceroneogenesis Backbone for esterification
FA-CoA + G3P Esterification enzyme Forms triacylglycerol

📘 [Integrated regulatory map – Figure in lecture slide 28]

ANNOTATE DIAGRAM ON PAGE 25 L4.1:

Um, this, that citrate, what remains the oxalo acetate needs to get back into the mitochondria to, to stay in the, the reb cycle. Going back in, that will give us some more NADPH. You could annotate even more things along the way, that acetylchoA gets converted to Malanol CA. Um, the fatty acids are going to get userified together to form a triase or glycerol. So you’ve got lots and lots of things feeding, all just so that this fatty acid synthesis can occur.


L11: Lipoprotein and Cholesterol Metabolism (Fat Disposal)


Fat Digestion & Absorption

1. Illustrate the problems involved in the digestion of fat and the role of bile salts in digestion

Explanation:

  • Dietary fats are hydrophobic, forming large lipid droplets.

  • Digestion problem: enzymes (like lipase) are water-soluble and can’t access fat interior.

  • Bile salts (amphipathic molecules made from cholesterol in the liver and stored in gall bladder):

    • Provide surface area for enzyme action

    • Release: When a meal containing fat is consumed, the gall bladder contracts and releases bile salts into small intestine.

    • Emulsify fats into micelles: bile salts mix with dietary fats in small intestine

    • cholesterol excretion via loss of bile salts in faeces is the only way body eliminates cholesterol

    • Recycled via enterohepatic circulation: bile salts are efficiently reabsorbed (98-99%) in the small intestine and recycled back into the body

    • cholesterol is either lost as bile salts or retained in the body, cannot be oxidised.


2. Understand the way in which fat is digested by pancreatic lipase

Explanation:

Pancreatic lipases are enzymes secreted into small intestine that act on fat emulsion created by bile salts. –> enzymes break down triglycerides into smaller absorbable components (monoacylglycerol, diacylglycerol, fatty acids, and glycerol).

  • Pancreatic lipase is secreted into the duodenum.

  • Acts on triglycerides inside micelles → releases:

    • 3 Free fatty acids

      • gets absorbed across mucosa into cells lining small intestine
    • Monoacylglycerols

    • Glycerol

  • Products diffuse into enterocytes → re-esterified into TAG → packaged into chylomicrons


3. Predict the consequences of problems in fat digestion, using fat substitutes or lipase inhibitors

Examples:

  • Gallstones block bile flow → impaired micelle formation (prevents release of bile salts into small intestine) → poor fat digestion → malabsorption of dietary fats

  • Orlistat: lipase inhibitor (inhibits digestion and absorption of dietary fats from small intestine) → undigested fat → steatorrhea (oily stools)

  • Olestra: artificial fat (FA + sucrose) not digestible (allows consumption of fried foods without adding calories)→ passes through gut; can deplete fat-soluble vitamins

📘 Clinical case: Orlistat side effects


Lipoprotein Function

4. Describe the general features of lipoproteins

source: L4.2 pg 14 - diagram here

Explanation:

  • Lipoproteins = spherical particles that carry fats in blood

  • Core: TAG and cholesterol esters

  • Surface: phospholipids + apoproteins

    • Apoproteins serve as labels, enzyme activators, and docking molecules for cellular receptors
  • Differ by:

    • Density (chylomicrons → HDL)

    • Size

    • Apoprotein content (e.g., ApoB-48, ApoC-II, ApoE)


graph LR A[Phospholipid Monolayer] --> B(Hydrophobic Tails Inward) A --> C(Apoproteins on Surface) B --> D{Core: Triglycerides & Cholesterol} C --> E[Receptor Recognition] C --> F[Enzyme Activation] C --> G[Lipoprotein Labeling] D --> H[Fat Transport] style A fill:#ff99ff,stroke:#333,stroke-width:2px style B fill:#ccccff,stroke:#333,stroke-width:2px style C fill:#ccccff,stroke:#333,stroke-width:2px style D fill:#ccccff,stroke:#333,stroke-width:2px style E fill:#ff99ff,stroke:#333,stroke-width:2px style F fill:#ff99ff,stroke:#333,stroke-width:2px style G fill:#ff99ff,stroke:#333,stroke-width:2px style H fill:#ff99ff,stroke:#333,stroke-width:2px

5. Understand the role of chylomicrons, VLDL, LDL, HDL and cholesterol in fat digestion and distribution of fatty acids and cholesterol to tissues

Lipoprotein Function Origin Fate
Chylomicrons Transport dietary TAG & cholesterol Intestine Lipolysis by LPL → remnants to liver
VLDL Distributes liver-made TAG Liver Becomes IDL → LDL
LDL Delivers cholesterol to cells VLDL remnant Endocytosed via LDL receptor
HDL Removes cholesterol from tissues Liver Returns to liver (reverse transport)

Clinical Significance: Measuring blood cholesterol involves assessing the levels and ratios of different lipoproteins (VLDL, LDL, HDL) rather than just free-floating cholesterol

Chylomicrons
  • Characteristic: the largest and least dense lipoprotein

  • Formation: formed in the intestinal muscosa to transport dietary fats and cholesterol from the small intestine to the rest of the body

  • Composition: packed with dietary fats (triglycerides) and cholesterol.

    • To effectively transport cholesterol within lipoproteins, it must be rendered completely hydrophobic.

    • This involves modifying cholesterol by attaching a fatty acid to it, forming a cholesterol ester (this makes it fully hydrophobic)

    • Acyl-CoA Cholesterol Acyltransferase (ACAT): The enzyme that catalyzes the esterification of cholesterol. Transfers a fatty acyl group from acyl-CoA to cholesterol

  • Entry into circulation: enter the lymph system instead of directly to the bloodstream

  • Destination: Transport fats (dietary triglycerides and cholesterol) to various tissues throughout body

  • Plasma Appearance: Blood plasma appears turbid or milky after a fatty meal due to chylomicrons

  • Metabolism through Lipoprotein lipase (LPL):

    • LPL is an enzyme located on the surface of blood vessels cappilary walls that hydrolyses triglycerides in lipoproteins –> releasing fatty acids and glycerol for uptake by cells

    • re-esterification requires a Glycerol 3-phosphate (to store fat away). liver converts glycerol to G3P using glycerol kinase (enzyme only present in liver). any other cells that want to store that feet needs to steal from glycolysis.

  • Chylomicron remnants: after LPL renoves trigylcerides from chylomicrons, remaining particle is called chylomicron remnant –> gets returned to liver

    • chylomicron remnant low in fat and higher in cholesterol

    -gets taken up by liver via endocytosis –> liver repackages with newly synthesised fats and glycerol

    • liver exports repackaged lipids as very-low-density lipoproteins (VLDL)
VLDL
  • Characteristic: pretty much does same thing. Goes around the body and any cells that want the fat of the VLDL are going to express LPL at capillary wall

    • cells digest some of the fat out of it and send it back to liver again.

    • if enough fat is taken out of the VLDL it becomes LDL, if a little bit taken it becomes IDL

LDL
  • Characteristic: rich in cholesterol and delivers it to cells and tissues throughout body

  • Delivery: Cells take up LDLs via LDL receptors on cell surface. Cells internalise LDL via endocytosis

  • Destination: cells break down LDLs to use the cholesterol and fat inside

HDL
  • Characteristic:

  • Formation:

  • Composition:

  • Entry into circulation:

  • Destination:

Summary Table:

Lipoprotein Size Density Primary Cargo Origin Destination
Chylomicrons Largest Lowest Dietary Triglycerides Intestinal Mucosa Lymph, then tissues
VLDL Large Low Endogenous Triglycerides Liver Tissues
IDL Medium Intermediate Triglycerides, Cholesterol VLDL Metabolism LDL or Liver
LDL Small High Cholesterol VLDL/IDL Metabolism Tissues, can lead to plaque formation
HDL Smallest Highest Cholesterol Liver, Intestine Liver (Reverse Cholesterol Transport)

Summary Story

Source: L4.2 pg 22 - diagram here

  1. Dietary fat intake: Fat coming in the diet

  2. small intestine: fat is emulsified with bile salts and broken down by pancreatic lipase

  3. absorption: fatty acids and glycerol are absorbed across intestinal walls

  4. chylomicron formation: intestinal cells repackage fats into chylomicron lipoproteins

  5. lymphatic system: chylomicrons enter the lymph and then the blood

  6. LPL action: any cells that want fats would use LPL in the lining of capillaries to digests tryglycerides from chylomicrons

  7. chylomicron remnant uptake: chylomicron remnants return to the liver

  8. liver processing: the liver uses cholesterol to make bile salts or packages fats into VLDLs

  9. VLDL metabolism: VLDLs deliver fat to cells via LPL, eventually becoming LDL

  10. LDL delivery: LDLs deliver cholesterol to cells via LDL receptors. any remaining will return to the liver again for repackaging

graph LR DF[Dietary Fats] --> SI[Small Intestine] SI --> CM[Chylomicrons] CM --> LS[Lymphatic System] LS --> BS[Bloodstream] BS --> LPL[Lipoprotein Lipase (LPL)] LPL --> FA[Fatty Acids + Glycerol] FA --> Cells[Peripheral Cells] BS --> CR[Chylomicron Remnants] CR --> Liver[Liver] Liver --> VLDL[VLDL] VLDL --> BS2[Bloodstream Again] VLDL --> IDL[IDL] IDL --> LDL[LDL] LDL --> Receptor[Cells via LDL Receptor] Receptor --> Chol[Cholesterol]

Cholesterol Metabolism

6. Summarise the endogenous formation of cholesterol and identify the rate limiting step

Most cells can synthesize their own cholesterol, often in amounts exceeding dietary intake. The body tightly regulates cholesterol synthesis, and statins, one of the most prescribed drugs, target the initial enzyme in this pathway.

Source: L4.2 pg 23

Pathway:

  • Acetyl-CoA → HMG-CoA → Mevalonate → Cholesterol

  • Rate-limiting enzyme: HMG-CoA Reductase

    • Regulated by:

      • Insulin (↑)

      • Cholesterol (↓ via feedback)

      • Statins (competitive inhibitors)

      • Circadian rhythm

  • By inhibiting cholesterol synthesis, statins force cells to uptake LDLs from the blood, lowering LDL concentration


7. Generate arguments for why LDL is bad and HDL is good

LDL:

  • Oxidised LDL (oxLDL) taken up by macrophages → foam cells → contributing to plaque formation in arteries by trigerring immune response (atherosclerosis)

  • Pro-inflammatory, plaque forming

    • Plaque formation in coronary arteries can lead to heart attacks, while in brain arteries, it can cause strokes.

HDL:

  • Removes excess cholesterol from tissues → delivered to liver for processing

  • Anti-inflammatory, protective against CVD

  • HDL Synthesis: HDLs are produced in the liver.

  • Reverse Cholesterol Transport: HDLs scavenge excess cholesterol from tissues and transport it back to the liver.

  • Liver Processing: The liver can use the cholesterol from HDLs to synthesize bile salts or repackage it into VLDLs.

  • Desirable Levels: High HDL levels are generally considered beneficial.

📘 LDL vs HDL summary – AHA


8. Critically analyse the different methods for reducing blood cholesterol levels

Strategy Mechanism
Statins Inhibit HMG-CoA reductase → ↑ LDL receptor expression
Phytosterols Compete with cholesterol for gut absorption
Resins Bind bile salts → ↑ conversion of cholesterol to bile acids
Diet Reduce saturated fat → improves HDL/LDL ratio
CETP inhibitors (experimental) Block cholesterol transfer from HDL → VLDL

📘 Cholesterol treatment guidelines – AHA


9. Describe the importance of cholesterol in membrane integrity

Explanation:

  • Cholesterol modulates membrane fluidity:

    • Prevents crystallisation of saturated FAs

    • Adds stability in fluid unsaturated membranes

  • Embedded between phospholipids

  • Essential for lipid raft formation, receptor function, membrane proteins

📘 Membrane dynamics – Nature Education

🧬 Importance of Chesterol in Membrane Integrity

Cholesterol is a vital structural component of animal cell membranes. It helps maintain the balance between fluidity and rigidity, ensuring that membranes stay functional and stable under different conditions.


🧱 Key Roles of Cholesterol in Membranes:

1. Maintains Membrane Fluidity

  • At low temperatures: Cholesterol prevents phospholipids from packing too tightly, keeping the membrane flexible.
  • At high temperatures: Cholesterol restrains phospholipid movement, preventing the membrane from becoming too fluid or leaky.

✅ Acts like a buffer — stabilizing membrane behavior across temperature changes.


2. Strengthens the Membrane

  • Fits snugly between phospholipid tails, making the membrane less permeable to small water-soluble molecules (like ions).
  • Increases mechanical stability, reducing membrane rupture or distortion.

3. Creates Membrane Microdomains (Lipid Rafts)

  • Cholesterol helps form lipid rafts — small, ordered regions within the membrane.
  • These are platforms for cell signaling, protein sorting, and endocytosis.

🧠 Summary:

Cholesterol is essential for maintaining the structure, fluidity, and function of cell membranes. It acts as a fluidity buffer, barrier, and organizer, allowing the membrane to be both flexible and strong — critical for cell survival and communication.


Unsaturated Fatty Acids

10. Provide an overview of the types and sources of dietary unsaturated fatty acids

Types:

  • Monounsaturated (MUFA): olive oil (oleic acid)

  • Polyunsaturated (PUFA):

    • Omega-6 (linoleic acid): plant oils

    • Omega-3 (alpha-linolenic acid): flaxseed, fish oil

Function:

  • Increase membrane fluidity

  • Cardioprotective effects

  • Some essential (must be obtained from diet)

📘 FA types and sources – Harvard

🥑 Overview of Unsaturated Fatty Acids: Types & Dietary Sources

Unsaturated fatty acids are fats that contain one or more double bonds in their carbon chains. These are generally considered heart-healthy and are liquid at room temperature.


🧬 Types of Unsaturated Fatty Acids

1. Monounsaturated Fatty Acids (MUFAs)
  • Have one double bond
  • More stable than polyunsaturated fats
  • May help reduce LDL (“bad”) cholesterol

Example:

  • Oleic acid (found in olive oil)

2. Polyunsaturated Fatty Acids (PUFAs)
  • Have two or more double bonds
  • Essential fatty acids (must be obtained from diet)
a. Omega-3 Fatty Acids (n-3)
  • First double bond at the 3rd carbon from the methyl end
  • Anti-inflammatory, support heart and brain health

Examples:

  • ALA (alpha-linolenic acid) – plant-based

  • EPA and DHA – marine-based

b. Omega-6 Fatty Acids (n-6)
  • First double bond at the 6th carbon from the methyl end

  • Can be pro-inflammatory in excess but are essential

Examples:

  • Linoleic acid

  • Arachidonic acid


🍽️ Dietary Sources
Type Fatty Acid Sources
MUFAs Oleic acid Olive oil, avocados, almonds, peanuts, canola oil
PUFAs (Omega-3) ALA Flaxseeds, chia seeds, walnuts, soybean oil
EPA, DHA Fatty fish (salmon, mackerel, sardines), fish oil
PUFAs (Omega-6) Linoleic acid Sunflower oil, corn oil, soybean oil, walnuts
Arachidonic acid Meats, eggs, poultry (small amounts)

🧠 Summary:

Unsaturated fatty acids include monounsaturated (MUFAs) and polyunsaturated (PUFAs) fats. They are found in plant oils, fish, nuts, and seeds, and are crucial for cell membranes, inflammation control, and cardiovascular health.


11. Review the key steps in the oxidation of unsaturated fatty acids

Explanation:

  • β-oxidation can proceed normally after cis→trans conversion

  • Requires enoyl-CoA isomerase

  • Oxidation restarts from the trans-intermediate

  • Trans fats (industrial) are not handled efficiently → raise LDL:HDL ratio

📘 [FA oxidation pathway – Stryer Biochem Fig. 17-9]

🔥 Oxidation of Unsaturated Fatty Acids – Key Steps

Unsaturated fatty acids undergo β-oxidation, just like saturated fatty acids, but with extra steps to handle their double bonds, which can interfere with standard oxidation enzymes.


🧬 Overview:
  • Standard β-oxidation breaks down fatty acids 2 carbons at a time.
  • Double bonds (especially in cis form) disrupt enzyme activity and need to be rearranged or reduced before continuing.

🔄 Key Additional Steps for Unsaturated Fatty Acids:
1. 🔁 β-Oxidation Proceeds Normally Until a Double Bond Is Encountered

2. 🔧 Isomerization of Cis-Δ³ Double Bonds
  • Enzyme: Enoyl-CoA isomerase
  • Function: Converts a cis-Δ³ double bond to a trans-Δ², which is compatible with β-oxidation enzymes.

Used for: Fatty acids with odd-numbered double bonds (e.g. oleic acid)


3. 🔧 Reduction of Conjugated Double Bonds
  • Enzyme: 2,4-Dienoyl-CoA reductase
  • Function: Converts a conjugated Δ²,Δ⁴-dienoyl-CoA to a trans-Δ³-enoyl-CoA, which is then converted by enoyl-CoA isomerase to the usable trans-Δ²-enoyl-CoA.

Used for: Fatty acids with even-numbered double bonds (e.g. linoleic acid)


🧠 Summary Table:
Issue Caused by Double Bond Enzyme Needed Action
Cis-Δ³ double bond Enoyl-CoA isomerase Converts it to trans-Δ²
Conjugated Δ²,Δ⁴ system 2,4-Dienoyl-CoA reductase + Enoyl-CoA isomerase Reduces and rearranges to trans-Δ²

🧾 Final Products:
  • Same as saturated FA oxidation: Acetyl-CoA, FADH₂, NADH

  • But less ATP is produced than with saturated fatty acids of the same length because:

    • Some double bonds skip FADH₂-generating steps

📌 Summary:

Oxidation of unsaturated fatty acids requires extra enzymes to rearrange or reduce double bonds so they can enter the normal β-oxidation cycle. These steps ensure that even with double bonds, the fatty acid can still be broken down for energy.


L12: Nitrogen Metabolism and Pathway Integration

1. Quantify the Inputs and Outputs That Represent Daily Nitrogen Balance

📥 Nitrogen Inputs:

  • Protein intake: ~100 g/day ↳ Contains ~16.5 g of nitrogen

🔁 Nitrogen Turnover:

  • Total body protein pool: ~6 kg in a 70 kg adult

  • Daily turnover: ~200 g/day (~4%)

    • This is the recycling of existing proteins into new ones (not full degradation).

    • Varies by tissue:

      • Muscle: ~2% turnover (largest total pool but slow renewal)
      • Intestine: ~15% turnover (high exposure to oxidative and physical stress)

📤 Nitrogen Outputs:

Route Amount (approx.)
Urea (urine) ~14 g nitrogen/day
Faeces ~2 g nitrogen/day
Skin ~0.5 g nitrogen/day

Total nitrogen loss16.5 g/day, matching intake → neutral nitrogen balance


🔎 Important Clarifications:

  • Excess amino acids are not stored: Unlike carbohydrates (glycogen) and fats, amino acids must be used, recycled, or excreted.
  • Most nitrogen leaves the body as urea, generated by the urea cycle in the liver.

🧠 TLDR Summary:

Daily nitrogen balance reflects 100 g/day protein intake (~16.5 g nitrogen) matched by equivalent nitrogen loss, mostly as urea. Around 200 g of body protein is recycled daily, and although muscle holds most protein, it’s slow to turn over compared to rapidly renewing tissues like the intestine. Amino acids can’t be stored, so any excess must be degraded and detoxified.


2. Recognise the Physiological Periods in Which Amino Acid Processing Is Highest and Lowest

📈 Periods of High Amino Acid Processing:

  1. Starvation or fasting

    • Low insulin → stimulates proteolysis
    • Muscle releases amino acids (especially alanine and glutamine)
    • Alanine transports nitrogen to liver (via the glucose–alanine cycle)
    • Carbon skeletons used for gluconeogenesis and energy
  2. High-protein diet

    • Excess amino acids → must be detoxified (amine groups → urea)
    • Carbon skeletons → oxidised, or used for glucose/fat synthesis
  3. High-turnover tissues (e.g. intestinal epithelium, liver)

    • Constant damage/renewal → constant protein turnover and amino acid processing

Periods of Low Amino Acid Processing:

  1. Well-fed state with balanced diet

    • Dietary amino acids primarily used for protein synthesis
    • Minimal oxidation or urea production
  2. Resting muscle

    • Despite containing the bulk of body protein, only ~2% turnover/day
    • Amino acids mostly locked in contractile proteins

Key Point:

  • Processing is driven by need: We process amino acids when:

    • Protein is being broken down (starvation, turnover)
    • Protein is being consumed in excess
  • The liver handles most of the metabolic sorting:

    • Processes nitrogen (→ urea)
    • Handles carbon skeletons (→ glucose, fat, or energy)

TLDR Summary:

Amino acid processing spikes during starvation, excess protein intake, and in fast-renewing tissues like the intestine. In these states, amino acids are broken down for energy, glucose, or fat, and nitrogen is converted to urea. Processing is lowest in well-fed, non-stressed states, especially in muscle, which has low protein turnover despite being the largest protein reserve.


3. Describe the basic principles of amino acid processing

Explanation: Amino acid processing involves handling the nitrogen (amine group) and the carbon skeleton of amino acids once they are released from proteins (via turnover, dietary intake, or catabolism).

Core Principles:

  1. Two key parts of every amino acid:
  • Amine group (–NH₂) → toxic as free ammonia (NH₃) → must be detoxified via the urea cycle.

  • Carbon skeleton → used for energy, glucose synthesis, or fatty acid synthesis

  1. Amino acids cannot be stored
  • Unlike fat or glycogen, amino acids must be used, recycled, or degraded

  • Excess amino acids are broken down; their nitrogen is excreted and carbon skeletons repurposed

  1. The liver is the central processor
  • First organ to receive dietary amino acids via the hepatic portal vein

  • Performs:

    • Transamination to collect nitrogen into glutamate/alanine

    • Urea synthesis (from ammonia and aspartate)

    • Carbon skeleton processing (oxidation, gluconeogenesis, lipogenesis)

  1. Processing situations:
  • Normal turnover: proteins degraded and rebuilt; unused amino acids are oxidised or excreted

  • Starvation: proteolysis increases, amino acids used for gluconeogenesis

  • High-protein intake: excess amino acids → detoxified and used for energy or fat synthesis

  1. Branch-chain amino acids (BCAAs):
  • Leucine, isoleucine, valine

  • Not metabolised significantly by the liver

  • Released into circulation → taken up and oxidised by muscle

TLDR Summary: Amino acid processing revolves around detoxifying nitrogen (via the urea cycle) and reusing carbon skeletons (via gluconeogenesis, lipogenesis, or oxidation). The liver is the central organ, coordinating nitrogen disposal and carbon fate. Amino acids can’t be stored, so any excess must be metabolised. BCAAs are the exception, bypassing liver metabolism and serving as an energy source in muscle.

📘 AA metabolism – LibreTexts

Basic Principles of Amino Acid Processing

Amino acid processing refers to how the body handles, breaks down, and uses amino acids — especially when they’re not needed for protein synthesis. This involves separating their nitrogen (amino group) from their carbon skeleton, and using those parts for different purposes.


Key Steps in Amino Acid Processing

1. Transamination

  • The amino group is transferred from the amino acid to α-ketoglutarate, forming glutamate.

  • Catalyzed by aminotransferase enzymes (requires pyridoxal phosphate, PLP).

  • Result:

    \[ \text{Amino acid} + \alpha\text{-ketoglutarate} \rightarrow \text{α-keto acid} + \text{glutamate} \]


2. Deamination

  • Glutamate releases the amino group as ammonia (NH₃).
  • Occurs in the liver via glutamate dehydrogenase.
  • Ammonia is toxic and must be dealt with quickly.

3. Urea Cycle (in liver)

  • Ammonia + CO₂ are converted into urea.
  • Urea is non-toxic and excreted by the kidneys in urine.
  • This detoxifies nitrogen waste from amino acid breakdown.

4. Fate of the Carbon Skeleton

  • The leftover carbon skeleton (α-keto acid) is used for:

    • Energy production (via TCA cycle)
    • Gluconeogenesis (→ glucose)
    • Ketogenesis (→ ketone bodies or fat)

🧩 Glucogenic vs Ketogenic:

Type Converted Into
Glucogenic amino acids Pyruvate or TCA intermediates → glucose
Ketogenic amino acids Acetyl-CoA or acetoacetate → ketones or fat

Summary:

Amino acid processing involves removing nitrogen (via transamination and urea cycle) and reusing the carbon skeleton for energy, glucose, or fat. This process is essential during fasting, low-carb intake, or protein-rich diets.


4. Understand the significance of amino acid degradation enzymes having a high Km

🔬 What is Km?

  • Km is a measure of an enzyme’s affinity for its substrate.
  • A high Km means the enzyme has low affinity and is only active when substrate concentration is high.

📉 Amino acid degradation enzymes have high Km:

  • These enzymes only become significantly active when amino acid levels are high (e.g. after a protein-rich meal or during catabolism).
  • This ensures amino acids are not broken down unnecessarily when in low supply (preserving them for protein synthesis).

🔑 Why this matters:

  • Not rate-limiting: These enzymes don’t control the speed of degradation; they act when [amino acid] is high.

  • This makes amino acid degradation:

    • Opportunistic (responsive to surplus)
    • Unregulated by feedback inhibition or hormones (unlike glycolysis/fat metabolism)
  • Ensures that any amino acids not immediately needed are efficiently degraded and detoxified


🚮 Consequence:

If excess amino acids are present, they are rapidly degraded, regardless of metabolic state — because the enzymes are already there and waiting to act.


🧠 TLDR Summary:

The enzymes that break down amino acids have high Km, meaning they only work when amino acid concentrations are high. This ensures the body preserves amino acids during deficiency but efficiently degrades excess when present. These enzymes are not tightly regulated, making amino acid degradation dependent on availability, not on hormonal control.


5. Describe the central role of transaminases in the shuffling of amino groups during amino acid processing

Understood. Here’s the comprehensive explanation for learning objectives 5, 6, and 7, using both the transcript and lecture slides, without emojis or informal elements:


5. Describe the Central Role of Transaminases in the Shuffling of Amino Groups During Amino Acid Processing

Key Concepts:

Transaminases (aminotransferases) are enzymes that transfer amino groups from one amino acid to an α-keto acid. This process is central to:

  • Recycling nitrogen between amino acids
  • Concentrating nitrogen for safe excretion
  • Reconfiguring carbon skeletons for use in metabolism

Mechanism:

  • The amino group from an amino acid is transferred to an α-keto acid.

  • The major acceptors of amino groups are:

    • α-ketoglutarate → becomes glutamate
    • Pyruvate → becomes alanine
    • Oxaloacetate → becomes aspartate

These reactions allow the nitrogen to be safely collected and shuttled toward the liver for disposal via the urea cycle.

Role in Muscle and Liver:

  • In muscle, amino groups are transferred to pyruvate to form alanine, which enters the glucose-alanine cycle.
  • In liver, transaminases help produce aspartate and glutamate, which feed into the urea cycle.

Summary:

Transaminases enable the safe, reversible transfer of nitrogen, allowing amino acids to be broken down or re-synthesised. They also help channel nitrogen into glutamate, alanine, or aspartate for entry into the urea cycle.


6. Summarise the Key Features of the Urea Cycle

Purpose:

The urea cycle disposes of excess nitrogen (from amino acids) by converting toxic ammonia into urea, which is excreted via urine.

Key Features:

  • Occurs primarily in the liver

  • Takes place in both the mitochondrial matrix and the cytosol

  • Requires input of two nitrogen atoms:

    • One from free ammonia (NH₄⁺)
    • One from aspartate
  • Involves the carrier molecule ornithine, analogous to oxaloacetate in the TCA cycle

  • Produces urea and fumarate (which feeds back into the TCA cycle)

Energetics:

  • Energy-intensive: costs 3 ATP equivalents per urea molecule
  • Despite this, it is essential to prevent ammonia toxicity

Entry Molecules:

  • Glutamate and glutamine deliver ammonia
  • Aspartate provides the second nitrogen

Summary:

The urea cycle detoxifies ammonia by forming urea, using inputs from glutamate and aspartate. It requires energy, is compartmentalised in mitochondria and cytosol, and links to central carbon metabolism via fumarate.


7. Conceptualise the Relationship Between the Urea Cycle and Other Central Metabolic Pathways

Integration Points:

1. With the TCA (Krebs) Cycle:

  • The urea cycle produces fumarate, which feeds directly into the TCA cycle.
  • Aspartate, formed from oxaloacetate in the TCA cycle, is a nitrogen donor in the urea cycle.
  • The two cycles are tightly linked and often referred to as the Krebs–Henseleit cycle.

2. With Amino Acid Metabolism:

  • Transamination reactions produce glutamate and aspartate, key nitrogen donors for the urea cycle.
  • Glutamine carries two amino groups and can be hydrolysed to release ammonia.

3. With Gluconeogenesis:

  • The carbon skeletons of amino acids (after transamination) feed into the TCA cycle or gluconeogenesis.
  • The glucose–alanine cycle shuttles amino nitrogen from muscle to liver and pyruvate back to glucose.

4. With Energy Metabolism:

  • The urea cycle consumes ATP, meaning nitrogen disposal has an energy cost.
  • During high rates of amino acid catabolism (e.g., starvation, high-protein diet), the energy burden of the urea cycle increases.

Summary:

The urea cycle is deeply interconnected with the TCA cycle, amino acid metabolism, gluconeogenesis, and energy metabolism. It shares intermediates (e.g., fumarate, aspartate) and relies on nitrogen-carrying amino acids to dispose of excess nitrogen, integrating nitrogen disposal with carbon and energy homeostasis.

Summary Concept Map:

         Amino Acid Catabolism
             ↓         ↑
     (Transamination) (Amino acid synthesis)
             ↓
          Glutamate
             ↓
     Glutamate Dehydrogenase
             ↓
           NH₃  →→→→→→→→→→→→→→
             ↓                 ↓
         Urea Cycle      Aspartate (from OAA)
             ↓                 ↑
       Urea (excreted) ←←←← Fumarate
                             ↓
                      TCA Cycle → OAA
                             ↓
                    Gluconeogenesis

📌 Final Takeaway:

The urea cycle is a nitrogen-disposal system that is deeply interwoven with carbon metabolism, especially the TCA cycle and gluconeogenesis. It ensures that while amino acids are used for energy or glucose, their nitrogen is safely excreted.


8. Understand why some amino acids are ketogenic and some are glucogenic

Amino acids are classified based on whether their carbon skeletons can be converted into glucose (gluconeogenic) or ketone bodies/fatty acids (ketogenic).

Glucogenic: Carbon skeletons can be converted into intermediates of the Krebs cycle and used for gluconeogenesis

  • Carbon skeletons → pyruvate, α-KG, OAA, fumarate, succinyl-CoA

  • Can feed gluconeogenesis

Ketogenic: Carbon skeletons are converted into acetyl-CoA, which can be used for fatty acid synthesis or ketone body formation

  • → acetyl-CoA or acetoacetate

  • Used in ketone synthesis or energy only

Both:

  • e.g., isoleucine, phenylalanine, tryptophan

📘 AA fate chart – Lumen Learning

🧬 Why Are Some Amino Acids Ketogenic and Some Glucogenic?

The classification of amino acids as ketogenic or glucogenic depends on the metabolic fate of their carbon skeletons after the amino (–NH₂) group is removed during amino acid catabolism.


🧪 Definitions:

Type What they can be converted into Final metabolic fate
Glucogenic Pyruvate or TCA cycle intermediates Used to make glucose via gluconeogenesis
Ketogenic Acetyl-CoA or acetoacetate Used to make ketone bodies or fat
Both → Some carbon goes to glucose, some to ketones Have dual metabolic pathways

🔁 Why This Matters:

The human body needs to:

  • Maintain blood glucose (especially for the brain)
  • Produce energy during fasting or low-carb states (via ketone bodies)

So depending on where an amino acid enters the metabolic map, it will be classified based on its carbon fate.


🧭 Where the Carbon Skeleton Enters Determines the Classification:

Entry Point Type Why?
Pyruvate Glucogenic Pyruvate can → glucose
Oxaloacetate, α-ketoglutarate, fumarate, succinyl-CoA Glucogenic All are TCA cycle intermediates that can → glucose
Acetyl-CoA, acetoacetate Ketogenic Cannot → glucose (carbons lost as CO₂ in TCA cycle)

🔍 Examples:

Amino Acid Type Reason
Leucine, Lysine Ketogenic only Carbon skeleton → acetyl-CoA/acetoacetate
Alanine, Glutamate, Aspartate Glucogenic only Carbon skeleton → pyruvate or TCA intermediates
Isoleucine, Phenylalanine, Tyrosine, Tryptophan Both Yields both glucogenic and ketogenic products

🔒 Key Concept:

Ketogenic amino acids cannot be used to make glucose because acetyl-CoA cannot be converted back to pyruvate — its carbons are lost as CO₂ in the TCA cycle.


🧠 Summary:

  • Glucogenic amino acids → help maintain blood glucose
  • Ketogenic amino acids → support ketone body production or fat synthesis
  • Entry point into metabolism determines this classification

9. Appreciate the reasons why most amino acids need to be in the human diet

Explanation:

  • Humans lack enzymes to synthesise certain AA side chains

  • Essential AAs often require:

    • Multiple steps

    • Cofactors (e.g., folate, B₆)

  • 9 essential AAs (e.g., lysine, threonine, leucine)

Conditional essentiality: e.g., arginine in children

  • Protein Synthesis Requirement: All 20 amino acids are required to synthesize any protein. (00:44:06)

  • Protein Composition: Most proteins contain hundreds of amino acids, with each of the 20 amino acids represented. (00:44:17)

  • Ribosome Function: Ribosomes translate mRNA into proteins by adding amino acids to the growing polypeptide chain.

The process of protein synthesis is highly dependent on the availability of all 20 amino acids. If even one amino acid is missing, the ribosome stalls, and the incomplete polypeptide chain is released and degraded. This is unlike language learning, where understanding can still be gleaned even if a word is unknown. In protein synthesis, the absence of a required amino acid halts the entire process.

  • Key Points:

    • Ribosomal Stalling: If an aminoacyl tRNA is not available to deliver the required amino acid, the ribosome stalls.

    • Incomplete Protein Degradation: The ribosome releases the partially constructed polypeptide, which is then broken down.

    • All 20 Amino Acids Required: The absence of even one amino acid prevents the cell from synthesizing any proteins.

📘 Essential AAs – NIH Fact Sheet


10. Predict the effects of purine synthesis inhibitors on cell health

Explanation:

  • e.g., 6-mercaptopurine inhibits PRPP amidotransferase

  • Affects de novo purine synthesis

  • High-impact in rapidly dividing cells (cancer, bone marrow)

  • Used in leukemia therapy

📘 Purine inhibitors – Cancer.gov

Synthesizing nucleotides de novo is energetically expensive. Therefore, cells prioritize salvaging and recycling existing purines rather than synthesizing them from scratch every time they are needed. This is particularly important for rapidly dividing cells, such as cancer cells, which require vast amounts of nucleotides to replicate their DNA. Salvage pathways are thus a key target for certain medicines, especially in cancer treatment. (00:46:56-00:48:31)

Key Points:

  • High Energy Cost of Synthesis: De novo nucleotide synthesis is energetically demanding.

  • Purine Salvage: Cells recycle purines from dead cells to conserve energy and resources.

  • Target for Medicines: Salvage pathways are targeted by drugs, especially in cancer therapy.

🔬 Predicting the Effects of Purine Synthesis Inhibitors on Cell Health

Purine synthesis inhibitors block the production of adenine (A) and guanine (G) nucleotides, which are essential building blocks of DNA, RNA, and ATP/GTP. Because of this, these inhibitors have profound effects on rapidly dividing and metabolically active cells.


🧬 Key Roles of Purines in Cells:

  • DNA/RNA synthesis → cell replication
  • ATP/GTP → energy and signaling
  • NAD⁺/FAD → redox coenzymes
  • cAMP/cGMP → second messengers

🔄 Predicted Cellular Effects of Purine Synthesis Inhibition:

1. 🛑 Inhibition of DNA & RNA Synthesis

  • Without purines, DNA replication stallscell cycle arrest, especially in S-phase.
  • RNA transcription slows → less protein production.

📌 Effect: Strongly affects rapidly dividing cells (e.g. cancer, immune, bone marrow, gut lining).


2. ⚡ Reduced ATP and GTP Levels

  • ↓ ATP = low energy charge → impairs all energy-dependent processes
  • ↓ GTP = impairs protein synthesis (GTP needed for translation) and signal transduction

📌 Effect: Cellular fatigue, slowed metabolism, cell death (apoptosis) in severe cases


3. ❌ Compromised Cell Signaling

  • ↓ cAMP and cGMP → defective hormonal responses, neurotransmission, and vascular tone

📌 Effect: Widespread dysregulation of intracellular signaling


4. 🧪 Tissue-Specific Toxicity

  • High turnover tissues are most vulnerable:

    Tissue Effect
    Bone marrow ↓ white/red blood cells → immunosuppression, anemia
    GI tract ↓ epithelial renewal → nausea, diarrhea, ulcers
    Hair follicles ↓ cell division → hair loss
    Immune system ↓ lymphocyte proliferation → immunosuppression (used therapeutically)

💊 Clinical Use of Purine Synthesis Inhibitors

  • Methotrexate, azathioprine, mycophenolate mofetil = used as:

    • Anti-cancer drugs
    • Immunosuppressants (e.g., in transplants, autoimmune disease)

🧠 Summary:

Purine synthesis inhibitors impair cell survival and division by blocking the production of purine nucleotides needed for DNA, RNA, energy, and signaling. Their impact is most severe in rapidly dividing or highly active cells, leading to immunosuppression, cytotoxicity, and organ-specific side effects.


11. Deduce the reasons for uric acid accumulation in cells experiencing chronic energy charge crisis

Explanation:

  • AMP → IMP → uric acid via degradation

  • During energy stress, AMP ↑ due to adenylate kinase

    \[ 2ADP ⇌ ATP + AMP \]

  • Chronic stress → excess urate → crystallises as gout

  • Exacerbated by low hydration, kidney dysfunction, alcohol

📘 Gout mechanism – Mayo Clinic

🧪 Why Does Uric Acid Accumulate During a Chronic Energy Charge Crisis?

A chronic energy charge crisis means the cell has persistently low ATP and high AMP/ADP — a sign that it’s under metabolic stress, like in ischemia, hypoxia, or starvation. In these conditions, uric acid builds up, especially in tissues like muscles, kidneys, and joints. Here’s why:


🔄 Step-by-Step Reasoning:

1. ATP Depletion Increases AMP

  • In energy stress, ATP is rapidly broken down:

    \[ \text{ATP} → \text{ADP} → \text{AMP} \]

  • AMP levels rise, and the cell needs to manage this buildup.


2. AMP is Degraded to Purine Bases

  • When AMP accumulates, it’s degraded through the purine nucleotide degradation pathway:

    \[ \text{AMP} → \text{Inosine} → \text{Hypoxanthine} → \text{Xanthine} → \text{Uric Acid} \]

  • These reactions are catalyzed by:

    • AMP deaminase
    • Xanthine oxidase (critical step producing uric acid)

3. Xanthine Oxidase Activity Increases

  • Under stress, xanthine oxidase becomes more active.

  • It catalyzes:

    \[ \text{Hypoxanthine} → \text{Xanthine} → \text{Uric Acid} \]

  • Both steps generate reactive oxygen species (ROS), adding to oxidative damage.


4. No Efficient Pathway to Recycle Uric Acid

  • Humans lack uricase, the enzyme that degrades uric acid further into soluble products.
  • So uric acid accumulates in blood and tissues, especially when purine degradation increases.

🧠 Summary of Causes:

Cause Effect
Low ATP Increases AMP breakdown
AMP degradation Produces hypoxanthine, xanthine → uric acid
Xanthine oxidase Converts purine bases → uric acid + ROS
Lack of uricase Uric acid can’t be broken down further
Chronic stress (e.g. ischemia) Sustained AMP degradation → sustained uric acid buildup

🔬 Clinical Relevance:

  • Seen in gout, ischemic tissue injury, tumor lysis syndrome, and metabolic syndrome
  • Uric acid may crystallize in joints → inflammation
  • Antioxidant in low amounts, damaging in excess

📌 Final Takeaway:

In a chronic energy crisis, the cell breaks down excess AMP through the purine degradation pathway, leading to uric acid accumulation — especially because humans cannot degrade uric acid further.


L13: Integration of Metabolism (Type 1 Diabetes)

1. Describe the Essential Features of Type I Diabetes

Core Features:

  • Autoimmune destruction of pancreatic β-cells

    • Leads to absolute insulin deficiency
    • Presence of autoantibodies against β-cell components
  • Onset typically in childhood or adolescence

  • Symptoms:

    • Polyuria (excessive urination)
    • Polydipsia (excessive thirst)
    • Weight loss despite adequate nutrition
    • Hyperglycemia (high blood glucose)
    • Ketonuria (ketones in urine), sometimes sweet/organic breath due to acetone
  • Rapid progression without treatment:

    • Before insulin therapy (pre-1920s), patients would die within weeks
  • Diagnosis:

    • Measured by C-peptide levels (marker of endogenous insulin production)

      • Clinically, pt’s serum we measure C-peptide instead of insulin because insulin doesn’t hang around for long as there are receptors around the body that call for insulin to come.

TLDR:

Type I diabetes is caused by autoimmune destruction of pancreatic β-cells, leading to complete insulin deficiency. This results in severe metabolic imbalance: hyperglycemia, ketone production, and rapid weight loss. Without insulin therapy, it is fatal. Key diagnostic features include low or absent C-peptide and autoantibodies against β-cells.


2. Illustrate the Phases of Insulin Secretion

Biphasic Insulin Response:

  1. First phase (Rapid)

    • Pre-synthesised insulin released immediately
    • Driven by glucose entering β-cells via GLUT2 and triggering exocytosis
  2. Second phase (Sustained)

    • Involves transcription and translation of new insulin
    • Requires processing of proinsulin into insulin + C-peptide

C-Peptide:

  • Cleaved off proinsulin during insulin production
  • Used clinically as a stable marker of endogenous insulin secretion
  • Absent in injected insulin, making it useful to distinguish endogenous from exogenous insulin

TLDR:

Insulin secretion occurs in two phases: a rapid first phase (pre-stored insulin) and a slower second phase (new insulin synthesis). Measuring C-peptide reflects β-cell function and distinguishes endogenous from injected insulin.


3. Review the Proposed Time-Course of Diabetes Development

Development Timeline:

  • Begins with genetic predisposition (e.g. HLA associations, family history)

  • Progresses through a latent autoimmune phase:

    • Autoantibodies appear long before clinical symptoms
    • β-cell mass gradually declines
  • First defect: loss of first-phase insulin secretion

  • Pre-diabetes stage: mild hyperglycemia; insulin secretion no longer sufficient to control glucose

  • Clinical onset:

    • Significant β-cell destruction
    • C-peptide levels fall
    • Full insulin dependence develops

Disease progression varies:

  • May take months to years
  • Immunosuppression in early stages may delay progression

TLDR:

Type I diabetes develops over time, starting with autoantibodies, progressing to partial β-cell loss, and culminating in insulin dependence. Early intervention in pre-diabetes (e.g. immunosuppression) may delay onset. Diagnosis occurs when endogenous insulin secretion becomes inadequate.


4. Review the Role of Insulin in Glucose Uptake into Tissues

Mechanism of Glucose Uptake:

  • Insulin promotes glucose uptake into certain tissues by triggering the translocation of GLUT-4 transporters to the cell membrane.

  • This is essential in:

    • Skeletal muscle
    • Adipose tissue (white fat)
    • So insulin switches on protein synthesis, lipogenesis, glycogenesis

GLUT Transporter Specificity:

  • GLUT-4: insulin-dependent (muscle, fat)
  • GLUT-1, GLUT-2: insulin-independent (brain, liver, basal glucose transport)

Tissue Differences:

  • Liver: uses GLUT-2; does not require insulin for glucose entry.
  • Brain: uses GLUT-1; glucose uptake unaffected by insulin status.
  • Muscle/fat: require insulin for significant glucose uptake and use.

Without Insulin:

  • GLUT-4 remains intracellular
  • Glucose uptake in muscle and fat is impaired, contributing to hyperglycemia

TLDR:

Insulin enables glucose uptake in muscle and fat by promoting GLUT-4 translocation to the membrane. The liver and brain remain insulin-independent. In Type I diabetes, GLUT-4 remains inactive, reducing tissue glucose uptake and causing hyperglycemia.


5. Quantify the Role of Insulin in Glucose Disposal

Post-uptake Fate of Glucose:

  • Insulin doesn’t just help glucose enter the cell — it also activates metabolic pathways to trap and use glucose:

    • Hexokinase traps glucose as glucose-6-phosphate (G6P)
    • Glycogenesis: insulin activates glycogen synthase
    • Glycolysis: insulin indirectly stimulates key enzymes like PFK-1
    • Lipogenesis: insulin promotes fatty acid synthesis from glucose

Consequences of Insulin Deficiency:

  • Hexokinase activity is inhibited due to G6P accumulation
  • Glucose uptake becomes futile — glucose enters, but without being used, it diffuses back out
  • Result: reduced glucose disposal, elevated blood glucose

Quantitative Contribution:

  • Muscle and fat together are responsible for disposing majority (~80%) of postprandial glucose — all insulin-dependent

TLDR:

Insulin is essential not only for glucose uptake into cells but also for promoting glucose usage via glycolysis, glycogen synthesis, and fat synthesis. Without insulin, glucose disposal halts, leading to G6P accumulation, feedback inhibition of hexokinase, and continued hyperglycemia.


6. Assess the Relative Importance of Insulin’s Anti-Lipolytic Role

Insulin’s Anti-Lipolytic Action:

  • Insulin inhibits lipolysis in adipose tissue by:

    • Lowering cAMP levels (via activation of phosphodiesterase)
    • Inhibiting hormone-sensitive lipase (HSL)

Without Insulin:

  • Uncontrolled lipolysis:

    • Massive release of fatty acids and glycerol into circulation
    • Fatty acids used by tissues → inhibit glucose oxidation
    • Glycerol used by liver for gluconeogenesis, exacerbating hyperglycemia

Consequences:

  • Contributes to ketogenesis in liver:

    • Excess acetyl-CoA (from fatty acids) + depletion of TCA intermediates → ketone body formation
  • Leads to metabolic acidosis (ketotic acidosis)

  • Lipid breakdown drives:

    • Fuel shift away from glucose
    • Further reduction in glucose usage

Comparative Significance:

  • Among insulin’s metabolic roles, anti-lipolysis is critically important in preventing:

    • Excess substrate for gluconeogenesis
    • Dangerous acidosis from ketone body overproduction

TLDR:

Insulin strongly inhibits fat breakdown. Without it, lipolysis becomes uncontrolled, releasing glycerol and fatty acids that drive gluconeogenesis, suppress glucose oxidation, and fuel ketone production. This contributes to both hyperglycemia and ketoacidosis, making insulin’s anti-lipolytic role central to metabolic stability.


7. Derive the Relationship Between Hepatic Glucose Production and Hypoinsulinemia

Normal Hepatic Regulation:

  • Insulin inhibits gluconeogenic enzymes in the liver.

  • Suppresses:

    • PEP carboxykinase
    • Fructose-1,6-bisphosphatase
    • Glucose-6-phosphatase
  • Insulin also reduces substrate availability for gluconeogenesis.

In Hypoinsulinemia (Type I Diabetes):

  • Loss of inhibition leads to uncontrolled gluconeogenesis.

  • Additionally:

    • Lipolysis releases glycerol → substrate for gluconeogenesis.
    • Proteolysis releases amino acids → especially alanine.
    • Fatty acid oxidation increases acetyl-CoA → inhibits PDH, increasing lactate production.
  • Thus, liver continues to make glucose despite hyperglycemia.

Net Effect:

  • Hepatic glucose production increases via:

    • Unchecked gluconeogenesis
    • Glycogenolysis (if glycogen still available)
  • Contributes directly to sustained hyperglycemia

TLDR:

Without insulin, the liver fails to suppress gluconeogenesis. Elevated glycerol, amino acids, and lactate supply abundant substrates. Hepatic glucose output rises even when blood glucose is already high, worsening hyperglycemia in Type I diabetes.


8. Predict the Effects of Ketosis on Fuel Oxidation

What Triggers Ketosis:

  • In hypoinsulinemia:

    • Excess fatty acids flood the liver due to uncontrolled lipolysis.
    • TCA cycle intermediates (e.g., oxaloacetate) are diverted to gluconeogenesis.
    • Acetyl-CoA from β-oxidation accumulates → diverted into ketogenesis.

Fuel Oxidation Consequences:

  • Ketone bodies (acetoacetate, β-hydroxybutyrate) produced in liver and exported.

  • Brain and peripheral tissues:

    • Begin oxidising ketones instead of glucose
    • Reduce glucose uptake and oxidation
  • PDH is inhibited by high acetyl-CoA and low insulin, so:

    • Pyruvate is converted to lactate, not acetyl-CoA
    • Glucose oxidation is further reduced

Net Metabolic Shift:

  • From glucosefatty acids and ketones as primary fuels
  • Promotes further glucose sparing, compounding hyperglycemia

TLDR:

Ketosis causes tissues (including the brain) to switch from glucose to ketone oxidation. This suppresses glucose usage, reinforces hyperglycemia, and drives the shift toward fat metabolism. Ketones become the dominant energy source in uncontrolled diabetes.


9. Outline the Main Metabolic Consequences of Hypoinsulinemia

Global Metabolic Effects:

System Affected Metabolic Effect
Carbohydrate metabolism ↓ Glucose uptake (GLUT-4 inactive)
↓ Glycolysis
↓ Glycogenesis
↑ Hepatic glucose production
Lipid metabolism ↑ Lipolysis
↑ Fatty acid oxidation
↑ Ketone body production
Protein metabolism ↑ Proteolysis
↑ Amino acid release
↑ Substrate supply for gluconeogenesis
Energy metabolism Tissues rely on fat and ketones, not glucose

Clinical Consequences:

  • Hyperglycemia: due to decreased uptake and increased hepatic output
  • Ketoacidosis: from excess ketone production
  • Weight loss: due to proteolysis and lipolysis
  • Polyuria and polydipsia: osmotic diuresis from glucose in urine
  • Fatigue and muscle wasting

TLDR:

Hypoinsulinemia unleashes unregulated catabolism: glucose uptake drops, fat and protein breakdown accelerate, and hepatic glucose production surges. The result is profound hyperglycemia, ketone body overproduction, metabolic acidosis, and rapid tissue wasting characteristic of uncontrolled Type I diabetes.


10. Describe the Main Components of Ketotic Acidosis

Biochemical Basis:

  • In hypoinsulinemia, excess fatty acids are released and oxidised to acetyl-CoA.

  • With TCA intermediates depleted (e.g. oxaloacetate used for gluconeogenesis), acetyl-CoA is diverted to ketogenesis.

  • Results in overproduction of ketone bodies:

    • Acetoacetate
    • β-hydroxybutyrate
    • Acetone (from spontaneous decarboxylation)

Why Acidosis Occurs:

  • All ketone bodies are acids (carboxyl groups)
  • Alongside ketones, excess lactate (from anaerobic glycolysis) and fatty acids contribute additional acidity
  • The combined acid load leads to a drop in blood pH

Clinical Consequence:

  • Metabolic acidosis
  • Potentially fatal if not treated
  • Characteristic in diabetic ketoacidosis (DKA)

TLDR:

Ketotic acidosis in Type I diabetes arises from uncontrolled ketone production due to high fatty acid oxidation and low TCA cycle capacity. Alongside lactate and fatty acids, ketone accumulation causes systemic acidosis that can be fatal without intervention.


11. Summarise How Hypoinsulinemia Causes the Main Signs and Symptoms of Type I Diabetes

Symptom Underlying Cause
Polyuria (frequent urination) Glucose exceeds renal threshold → osmotic diuresis
Polydipsia (excessive thirst) Water loss from tissues → dehydration triggers thirst
Weight loss Uncontrolled lipolysis and proteolysis
Fatigue, weakness Muscle wasting, loss of energy substrates
Ketotic breath Acetone from spontaneous decarboxylation of acetoacetate
Hyperglycemia ↓ Glucose uptake (GLUT-4), ↑ Hepatic gluconeogenesis
Glycosuria Glucose appears in urine once plasma glucose > 10 mM

This clinical picture is described as “starvation in the face of plenty”—cells cannot use glucose despite high blood levels.

TLDR:

Hypoinsulinemia causes osmotic diuresis, tissue dehydration, muscle and fat breakdown, hyperglycemia, and ketone accumulation. The result is rapid weight loss, thirst, ketotic breath, and fatigue—all hallmark symptoms of untreated Type I diabetes.


12. Judge the Main Aims of Diabetes Control

Short-Term Aims:

  • Avoid acute complications:

    • Hyperglycemia
    • Ketosis
    • Ketoacidosis

Long-Term Aims:

  • Prevent chronic complications caused by sustained hyperglycemia:

    • Capillary damage (retinopathy, nephropathy)
    • Neuropathy (due to sorbitol accumulation via polyol pathway)
    • Atherosclerosis and cardiovascular disease
    • Glycation of proteins (e.g. HbA1c formation)

Glycemic Control Target:

  • Maintain HbA1c < 7–7.5%
  • Avoid hypoglycemia, which can be more dangerous than moderate hyperglycemia

TLDR:

Diabetes control aims to prevent acute crises (like DKA) and long-term complications (e.g. vascular and nerve damage). The goal is stable glycemia with minimal excursions, using HbA1c and symptom tracking as indicators.


13. Assess the Different Methods for Monitoring and Insulin Delivery

Monitoring:

  1. Finger-prick glucose testing

    • Provides point-in-time measurements
  2. Continuous glucose monitoring (CGM)

    • Real-time glucose levels and trends
    • Increasingly used for tighter control
  3. HbA1c

    • Reflects average glucose over ~100 days
    • Indicator of long-term control

Insulin Delivery:

  1. Subcutaneous injections:

    • Short-, intermediate-, and long-acting insulins
  2. Insulin pumps:

    • Programmable continuous delivery
  3. Newer formulations:

    • Rapid-acting analogs (e.g. lispro, aspart)
    • Long-acting analogs (e.g. glargine, detemir)
    • Combinations tailored to patient needs

Diagnostic Marker:

  • C-peptide used to differentiate endogenous vs injected insulin

TLDR:

Diabetes management includes glucose monitoring (finger-prick, CGM, HbA1c) and insulin delivery (injections, pumps, analogs). Monitoring guides treatment decisions. Long-term success depends on maintaining stable glucose while avoiding hypoglycemia.


L15: ELMA Design (Spectrophotometry in Ethanol Assay)


1. Use standard curves and spectrophotometry to interpolate concentrations of unknown compounds

Explanation:

  • Standard curve: plot of absorbance vs known concentrations of ethanol
  • Spectrophotometry: measures light absorbance of a coloured product—in this case, the phenyl-aminoantipyrine complex at 500 nm
  • Linear range of the curve is critical for interpolation
  • Use the curve equation (e.g., y = mx + c) to determine the concentration of unknown samples by substituting their absorbance values into the equation

📘 Spectrophotometry & Standard Curves – Khan Academy


2. Explain why it is important to consider the working range when determining the concentration of a compound

Explanation:

  • The working range is the absorbance range where the standard curve remains linear

  • Readings outside this range:

    • Too low: signal may be indistinguishable from background
    • Too high: risk of saturation or non-linear response
  • Ensuring your unknowns fall within this range prevents invalid interpolation

  • Dilutions may be necessary to bring unknowns into the valid range

📘 Understanding the Beer-Lambert Law – LibreTexts


3. Apply the principles of spectrophotometry to biological contexts

Explanation:

  • In this assay, ethanol is oxidised by ethanol oxidase to acetaldehyde, producing H₂O₂

  • H₂O₂ is used by peroxidase to convert 4-AAP and phenol into a red chromogen

  • Reaction:

    \[ \text{Ethanol} + O₂ → \text{Acetaldehyde} + H₂O₂ \\ \text{4-AAP} + \text{Phenol} + 2H₂O₂ → \text{Red complex} + H₂O \]

  • Absorbance of red product measured at 500 nm (in glucose ELMA it was 540 nm)

  • The assay is adapted to test blood alcohol concentration (BAC) using red samples—so blanks are critical to correct for colour interference

📘 Spectrophotometry animation – YouTube


🔍 Additional Applied Concepts from Slides


🔬 Scenario Design – Experimental Overview

  • Aim: Measure blood alcohol concentration (BAC) from 4 drivers
  • BAC range: 0.00–0.30% w/v = 0–65 mM
  • Legal BAC threshold: 0.05% (w/v) = ~10.9 mM
  • You receive: 25 mM ethanol stock, enzyme mix, PPA buffer, H₂O, and blood-coloured driver samples

⚙️ Design Considerations

  • Assay volume limit: 200 µL (microplate format)

  • Dilutions needed to bring test samples into linear absorbance range

  • Must determine reaction completion time (~40 min for glucose ELMA)

  • Blank solution: all reagents except analyte (ethanol) → used to subtract background absorbance

  • Manually blank your data:

    \[ A_{\text{corrected}} = A_{\text{sample}} - A_{\text{blank}} \]

📘 Using spectrophotometric blanks – ThermoFisher Guide


📊 Practical Tips for Analysis

  • Prepare standard curve using known ethanol concentrations (e.g., 0–25 mM)
  • Measure absorbance at 500 nm at end-point
  • Apply standard curve to unknown absorbances
  • Use technical replicates (triplicates) for reliability
  • Ensure blanking is done against each sample matrix (i.e., red blood effect)

Molecular Biology

L18: Introduction to Molecular Biology

1. Describe the Flow of Genetic Information

a) Between Generations

  • Genetic information is passed down via DNA replication during cell division and reproduction.
  • Inherited DNA encodes all the genetic instructions for the organism.
  • Experiments (e.g. Griffith, Avery-MacLeod-McCarty, Hershey-Chase) confirmed that DNA, not protein, carries heritable information.
  • In special cases (e.g. viruses), RNA can act as the genetic material and be copied into RNA or reverse transcribed into DNA.

b) Within the Cell

  • The central dogma outlines the general information flow:

    1. Replication: DNA → DNA (during cell division)
    2. Transcription: DNA → RNA (via RNA polymerase)
    3. Translation: RNA → Protein (by ribosomes)
  • Reverse transcription (RNA → DNA) also occurs in some cells (e.g. retroviruses).

  • DNA is the long-term store, RNA is a short-term intermediate, and proteins (and some RNAs) are functional molecules.

  • Each cell has the full genome but expresses only what’s necessary based on context (e.g. skin cells don’t express insulin).

Molecular Analogy:

  • DNA = hard drive
  • RNA = RAM
  • Protein/RNA = functional tools or software This analogy illustrates the stability, selectivity, and regulation of genetic information access and usage.

TLDR:

Genetic information flows from DNA to RNA to protein within cells and is faithfully copied during cell division between generations. Most cells carry the complete genome, but only express genes as needed, ensuring efficiency and specificity. Experimental evidence firmly established DNA (not protein) as the hereditary material.


2. Appreciate the Sequence Abundance and Diversity in the Genome, Transcriptome and Proteome

Genome:

  • Defined as the entire DNA content of a cell.

  • Nearly all cells (except RBCs) have a full genome.

  • Genome includes:

    • Protein-coding genes
    • Non-coding regions (introns, regulatory sequences, repetitive elements)
  • In eukaryotes, only a small portion codes for proteins—the rest may have regulatory or unknown functions.

Transcriptome:

  • The complete set of RNA molecules transcribed from the genome at a given time.

  • Includes:

    • mRNA (~1–2% by weight)
    • rRNA (~80%) – forms ribosomes
    • tRNA (~15%) – transfers amino acids
    • snRNA, miRNA – regulatory roles
  • Transcriptome is dynamic: varies by cell type, developmental stage, and environmental conditions.

Proteome:

  • The entire set of proteins expressed in a cell or organism at a given time.

  • Proteins include:

    • Enzymes, ion channels, transcription factors, receptors, antibodies
  • Reflects not only gene expression but also post-transcriptional and post-translational modifications.

  • More complex than the genome due to alternative splicing and protein folding.

Regulation:

  • Because transcription and translation are resource-intensive, cells regulate expression tightly.
  • E.g., pancreatic β-cells produce insulin only when blood glucose is high.

TLDR:

While every cell contains the same genome, the transcriptome and proteome differ based on cell function and environmental cues. mRNA makes up only a tiny fraction of RNA; most is rRNA and tRNA. Proteins diversify cell functions and can be far more varied due to regulation, splicing, and folding. These molecular layers highlight the complexity beyond the DNA sequence itself.

🔹 Comparative Summary

Layer Contents Constant? Function
Genome All DNA Yes (in all nucleated cells) Information storage
Transcriptome All RNA expressed No (varies by cell type/time) Regulates protein synthesis
Proteome All proteins present No (cell-specific, dynamic) Executes cellular functions

📌 Conceptual Analogy Used in Lecture

Component Analogy Function
DNA Hard drive (permanent storage) Long-term, stable
mRNA RAM (temporary access) Transient access
Protein Application or device Functional unit

📘 Suggested Visual Resources


L19: Nucleic Acid Structure

🔹 1. Identify the main components of DNA and RNA with reference to Chargaff’s rules

DNA and RNA components:

  • Sugar: DNA = deoxyribose (no 2′-OH), RNA = ribose (2′-OH present)

  • Bases:

    • Purines: adenine (A), guanine (G)
    • Pyrimidines: cytosine (C), thymine (T) [DNA], uracil (U) [RNA]
  • Phosphate backbone: negatively charged, links 5′-phosphate of one nucleotide to 3′-OH of the next

Chargaff’s Rules (for double-stranded DNA):

  • %A = %T
  • %G = %C
  • Thus, purines = pyrimidines
  • Not applicable to single-stranded DNA or RNA

📘 Detailed: https://doi.org/10.1016/S0021-9258(19)50884-5


🔹 2. Describe the key covalent bonds in DNA and RNA: phosphodiester, N-glycosidic

Phosphodiester bond:

  • Connects 3′-OH of one sugar to 5′-phosphate of the next nucleotide
  • Forms the backbone of nucleic acids

N-glycosidic bond:

  • Connects the 1′ carbon of sugar to the nitrogen of the base
  • N9 of purines (A, G) or N1 of pyrimidines (C, T/U)

These are covalent bonds, making the backbone stable.


🔹 3. Explain how weak forces maintain the double helix

✅ Base stacking (hydrophobic & electronic interactions)

  • Aromatic rings are hydrophobic → stack via van der Waals forces
  • Provides structural stability

✅ Base pairing (hydrogen bonding)

  • A-T = 2 H-bonds, G-C = 3 H-bonds (more stable)
  • H-bonds are non-covalent, allow helix to unzip easily

✅ Ionic interactions

  • DNA’s backbone is negatively charged (phosphates)
  • Stabilized by Mg²⁺ or Na⁺ which shield repulsion

✅ Van der Waals interactions

  • Arise from optimally spaced stacked bases
  • Contribute to base stacking

📘 Video on forces: https://www.youtube.com/watch?v=UY8jXo1KwUY


4. Describe the Structural Features That Make DNA a Very Good Store of Genetic Information

Key Structural Features:

a) Double-Stranded Complementarity

  • DNA consists of two antiparallel strands held by complementary base pairing (A–T, G–C).
  • This allows redundancy—if one strand is damaged, the other can be used as a template for repair.
  • Ensures faithful replication and error correction.

b) Use of Deoxyribose Sugar

  • Deoxyribose (lacking a 2′-OH group) makes DNA more chemically stable than RNA.
  • RNA is more prone to hydrolysis due to the 2′-OH → DNA is better suited for long-term information storage.

c) Thymine Instead of Uracil

  • DNA contains thymine (T) instead of uracil (U), which is found in RNA.
  • Cytosine can spontaneously deaminate to uracil; the presence of thymine allows cells to detect and correct this type of mutation.
  • Helps maintain genomic integrity.

d) Supercoiling and Compaction

  • DNA can be compacted into chromatin, enabling long sequences to be stored in small spaces.
  • DNA is also topologically constrained (e.g. via supercoiling), enhancing packaging and regulation.

TLDR:

DNA’s double-stranded, complementary structure allows accurate replication and repair. The use of deoxyribose and thymine enhances its chemical stability and fidelity, making it an ideal long-term information storage molecule. Its ability to be compacted further supports its role as the cell’s genetic archive.


5. Explain the Importance of the Major and Minor Grooves to Gene Expression

What Are Grooves?

  • The DNA double helix has two grooves:

    • Major groove: wider and deeper
    • Minor groove: narrower and shallower
  • Arise due to the asymmetrical positioning of the sugar-phosphate backbone around the helical axis.

Functional Importance:

  • Major groove:

    • Exposes base-pair edges in a readable format

    • Allows sequence-specific binding by proteins such as:

      • Transcription factors
      • Restriction enzymes
      • Polymerases
  • Proteins can “read” the DNA sequence via hydrogen bond donors/acceptors and shape

  • Enables precise gene regulation

  • Minor groove:

    • Less chemically distinct but still used by some proteins and drugs
    • Important for non-specific DNA interactions (e.g. histones, structural proteins)

Regulatory Implication:

  • Access to grooves is essential for transcriptional activation or repression
  • Mutations or changes in groove accessibility can disrupt gene expression

TLDR:

The major and minor grooves in DNA expose specific chemical patterns that proteins use to recognize and bind to target sequences. The major groove, in particular, allows sequence-specific regulation, making it critical for gene expression control.


6. Identify the Structural Differences Between DNA and RNA

Feature DNA RNA
Sugar Deoxyribose (no 2′-OH) Ribose (has 2′-OH)
Bases A, T, G, C A, U, G, C
Strandedness Double-stranded (dsDNA) Typically single-stranded
Stability More stable Less stable (prone to hydrolysis)
Function Long-term genetic storage Temporary message, catalysis, regulation
Helical Form B-form (right-handed helix) Many forms, often complex secondary structure (e.g. loops, stems)

Structural Consequences:

  • RNA’s 2′-OH allows complex folding, but also makes it chemically unstable in alkaline conditions.
  • DNA is more uniform and stable, enabling long-term genetic storage.

TLDR:

DNA is a stable, double-stranded molecule with deoxyribose and thymine, optimised for information storage. RNA is typically single-stranded, contains ribose and uracil, and functions dynamically in gene expression and regulation. Their differences reflect their distinct biological roles.


L20: Prokaryotic DNA Replication (E. coli Focus)

1: Describe the general mechanism for nucleic acid synthesis (RNA and DNA), considering: substrate and products; orientation; supply of energy; key enzymes and proteins and their roles


1. Substrates and Products

Category Description
Substrates Deoxyribonucleoside triphosphates (dNTPs) for DNA synthesis (e.g., dATP, dGTP, dCTP, dTTP); NTPs for RNA synthesis
Products Newly synthesized nucleic acid strands: DNA strands with complementary base pairing to the template strand
  • Nucleotides are added to the 3’ hydroxyl (OH) end of the growing strand.
  • DNA polymerases require a primer with a 3’ OH to initiate synthesis (RNA primers made by primase).

2. Orientation

Strand Direction
Template strand Read 3’ to 5’
New strand Synthesized 5’ to 3’
  • This orientation results in the leading strand being synthesized continuously and the lagging strand in fragments (Okazaki fragments).

3. Supply of Energy

  • DNA and RNA synthesis is thermodynamically unfavourable, but energy is supplied by hydrolysis of dNTPs/NTPs:

    • When a nucleotide is added, pyrophosphate (PPi) is released.
    • PPi is further hydrolyzed into two inorganic phosphates (Pi), driving the reaction forward.

4. Key Enzymes and Proteins

Enzyme/Protein Function
Helicase (DnaB) Unwinds DNA at the replication fork
Single-Stranded Binding Proteins (SSBs) Stabilize single strands after separation
Primase (DnaG) Synthesizes short RNA primers to provide 3’ OH group
DNA Polymerase III Main replicative polymerase; adds nucleotides and proofreads (3’→5’ exonuclease)
Sliding Clamp (β-clamp) Holds DNA Pol III in place on DNA
Clamp Loader Loads sliding clamp onto DNA
DNA Polymerase I Replaces RNA primers with DNA; has both 5’→3’ and 3’→5’ exonuclease activities
DNA Ligase Seals nicks between adjacent nucleotides after RNA replacement
Topoisomerase II (DNA Gyrase) Relieves supercoiling ahead of replication fork
Topoisomerase IV Separates circular chromosomes after replication
DnaA Recognizes oriC (origin of replication) and initiates DNA unwinding

5. Replication Overview Flowchart

1. Initiation
   └─ DnaA binds oriC and unwinds DNA
   └─ Helicase (DnaB) unwinds DNA → replication bubble
   └─ SSBs stabilize single strands

2. Primer Synthesis
   └─ Primase (DnaG) synthesizes RNA primers on both strands

3. Elongation
   └─ DNA Pol III adds nucleotides 5'→3' from RNA primers
      └─ Leading strand: continuous
      └─ Lagging strand: discontinuous (Okazaki fragments)
   └─ Sliding clamp ensures processivity
   └─ Clamp loader assists clamp attachment

4. Primer Removal & Replacement
   └─ DNA Pol I removes RNA (5’→3’ exonuclease) and fills gap with DNA

5. Ligation
   └─ DNA ligase forms final phosphodiester bond

6. Termination
   └─ Tus-Ter complex halts replication forks
   └─ Topoisomerase IV separates circular chromosomes

TLDR:

  • DNA synthesis is 5’→3’, using dNTPs as substrates and requiring RNA primers.
  • It proceeds continuously on the leading strand and discontinuously on the lagging strand (Okazaki fragments).
  • Energy is supplied by pyrophosphate hydrolysis.
  • Key players include helicase, SSBs, primase, DNA polymerases (III for synthesis, I for primer replacement), ligase, and topoisomerases.
  • The entire mechanism ensures fast, accurate replication suitable for rapid prokaryotic division, especially in organisms like E. coli.

2. Describe the Problems Associated with Replication and How the Cell Overcomes Them

Replication in prokaryotes presents several biochemical and structural challenges. Here’s how the cell solves each one:


Problem 1: DNA Must Be Unwound

  • Issue: DNA is a stable double helix that must be separated into single strands for copying.

  • Solution:

    • Helicase (DnaB) unwinds DNA at the replication fork.
    • SSBs (single-stranded binding proteins) stabilise the unwound regions and prevent re-annealing.

Problem 2: DNA Supercoiling Ahead of the Fork

  • Issue: As helicase unwinds DNA, positive supercoils form ahead of the replication fork, halting progression.

  • Solution:

    • Topoisomerase II (DNA gyrase) relaxes supercoiling by breaking and rejoining DNA strands.

Problem 3: DNA Polymerase Can’t Start from Scratch

  • Issue: DNA polymerase cannot initiate synthesis de novo; it requires a 3′-OH group.

  • Solution:

    • Primase (DnaG) synthesises short RNA primers to provide a starting 3′-OH for DNA Pol III.

Problem 4: Lagging Strand Must Be Synthesised in Pieces

  • Issue: DNA is antiparallel, but DNA polymerase only works 5′→3′. The lagging strand runs the “wrong way.”

  • Solution:

    • Lagging strand is synthesised discontinuously in Okazaki fragments, each with its own RNA primer.
    • DNA Pol I removes RNA primers and fills in with DNA.
    • DNA ligase seals the nicks between fragments.

Problem 5: Maintaining Processivity

  • Issue: DNA polymerase could fall off the template during replication.

  • Solution:

    • The sliding clamp (β-clamp) encircles DNA and holds DNA Pol III in place.
    • The clamp loader places and repositions the clamp as needed.

Problem 6: Circular Chromosomes Must Be Resolved

  • Issue: Prokaryotic chromosomes are circular and become interlinked after replication.

  • Solution:

    • Topoisomerase IV separates the two daughter chromosomes after termination.

TLDR:

Replication requires overcoming structural barriers like supercoiling, antiparallel strand orientation, and polymerase limitations. Helicase, topoisomerases, primase, DNA polymerases, clamps, and ligase coordinate to unwind, copy, and rejoin the genome with high efficiency and structural integrity.


3. Relate the Properties of DNA Polymerases to the Need for Fidelity in the Copying Process

Replication must be extremely accurate, far more so than transcription, for the following reasons:


Why Replication Requires High Fidelity:

Reason Explanation
Mutations are permanent Errors in DNA replication become fixed in the genome and passed on to daughter cells.
Protein errors are transient Mistakes in transcription or translation affect only that molecule, not future generations.
Genome is only copied once per cycle There’s little opportunity to “average out” errors like there is in mRNA/protein production.

How DNA Polymerases Ensure Fidelity:

Property Function
Base selection DNA Pol III selects the correct dNTP based on base-pairing geometry.
Proofreading activity 3′→5′ exonuclease activity removes mismatched bases as they are added.
Post-replication repair Additional mechanisms fix occasional mismatches (e.g., methyl-directed mismatch repair).
  • Error rate with proofreading: ~1 in 10⁷
  • With mismatch repair: ~1 in 10⁹–10¹⁰

Comparison with RNA Polymerase:

Feature DNA Polymerase III RNA Polymerase
Proofreading Yes (3′→5′ exonuclease) No proofreading
Error rate 10⁻⁷ (or better) 10⁻⁴
Consequence of error Inherited mutation Misfolded or nonfunctional protein (temporary)

TLDR:

DNA polymerases are highly accurate due to nucleotide selectivity and proofreading. This high fidelity is essential because replication errors are heritable, unlike transcription errors, which are transient. Additional post-replication repair mechanisms further reduce mutation rates, preserving genomic stability.


L21: Eukaryotic Replication

1. Describe the Controls on the Cell Cycle and How Mutations in These Can Lead to Cancer

Core Concepts

Phases of the Eukaryotic Cell Cycle

Phase Description
G₁ (Gap 1) Decision to divide or enter quiescence (G₀)
G₀ Non-dividing state; can be reversible (quiescent) or irreversible (senescent)
S (Synthesis) DNA replication occurs
G₂ (Gap 2) Final preparations for mitosis
M (Mitosis) Cell division into two daughter cells

Key Regulatory Elements

  • Cyclins + CDKs: Cyclin-dependent kinases regulate progression by phosphorylating target proteins. Cyclin levels fluctuate; CDK levels stay relatively constant.

  • Checkpoints:

    • G₁ → S: DNA damage check
    • S → G₂: Ensure complete, error-free DNA replication
    • M: Chromatid attachment to spindle apparatus

Cancer and Mutation

Class Function Cancer Relevance
Proto-oncogenes Stimulate cell growth Mutated → Oncogenes → gain-of-function → unregulated growth
Tumour Suppressors Inhibit cell division or promote apoptosis Loss-of-function in both alleles required for dysfunction

Example: Retinoblastoma Protein (pRb)

  • Regulates G₁ checkpoint
  • Mutation in both alleles → retinoblastoma + risk of other cancers

TLDR:

The cell cycle is tightly regulated by cyclins, CDKs, and checkpoints to ensure cells only divide when safe. Mutations in growth-promoting or inhibitory genes (proto-oncogenes or tumour suppressors) can disrupt this balance, leading to unregulated division and cancer.


2. Appreciate the Similarities and Differences Between Eukaryotic and Prokaryotic DNA Replication

Comparison Table

Feature Prokaryotic (e.g., E. coli) Eukaryotic (e.g., Human)
Chromosome Structure Single, circular Multiple, linear
Genome Size ~4.6 million bp ~6 billion bp (diploid)
Origins of Replication 1 per genome 30,000–50,000 per genome
Replication Rate Fast (~1000 nt/s) Slower (~50 nt/s)
Polymerases DNA Pol III (main) DNA Pol δ (main)
Proofreading Yes (3’→5’ exonuclease) Yes (DNA Pol δ)
Packaging Supercoiling Histone wrapping, chromatin structure
Re-initiation Control oriC control via DnaA CDKs prevent reactivation of origins

Commonalities

  • Bidirectional replication
  • Primase lays down RNA primers
  • Okazaki fragments on lagging strand
  • 5′ → 3′ synthesis direction
  • Replication forks and helicases

TLDR:

While the core mechanism of DNA replication is conserved across life—bidirectional synthesis, priming, and proofreading—eukaryotes face added complexity due to larger, linear genomes and the need for coordinated cell cycle regulation. Multiple origins and slower polymerases compensate for size, and histone packaging adds a layer of regulation.


3. Explain the Unique Challenges of Eukaryotic Replication at Initiation and the Ends and How the End Problem Is Solved in the Cell

Initiation Challenges

  • Multiple origins of replication must be selected and activated precisely.

  • Pre-replicative complex (pre-RC) assembled in G₁:

    • ORC (origin recognition complex), CDC6, CDT1, MCM helicase
  • Activation in S phase:

    • CDKs phosphorylate and recruit replication machinery
  • Regulation:

    • CDKs inhibit re-initiation → ensures DNA is copied only once

Chromatin Repackaging

  • DNA must be re-wrapped around histones after replication.

  • Histone genes:

    • Present in multiple copies (rapid transcription)
    • No introns or polyA tail → fast degradation
    • Transcriptionally and post-transcriptionally regulated

The End Replication Problem

Problem:

  • Lagging strand cannot be fully replicated at 3′ ends → telomere shortening

Solution:

  • Telomerase:

    • Ribonucleoprotein with:

      • RNA template (~1.5 repeats of 5′-TTAGGG-3′)
      • Reverse transcriptase activity
    • Extends 3′ end → allows primer addition and lagging strand fill-in

TLDR:

Eukaryotic replication begins from multiple regulated origins, requiring orchestration via cyclin-dependent kinases. Replication also demands rapid histone production for DNA packaging. Linear chromosomes pose a unique challenge: end regions cannot be replicated conventionally, which is solved by telomerase extending telomeres—a mechanism vital for cell immortality and highly active in cancer cells.


4. Predict the Implications of Telomerase Activity: Ageing and Immortality

Telomerase Overview

  • Telomerase is a ribonucleoprotein reverse transcriptase.
  • Carries its own RNA template, which it uses to extend the 3′ end of the DNA strand at telomeres.
  • Compensates for the end replication problem on the lagging strand.

Telomerase Expression Varies by Cell Type

Cell Type Telomerase Activity
Germ cells High
Stem cells Moderate
Somatic cells Very low or absent
Cancer cells Reactivated and high

Implications for Ageing

  • In somatic cells, telomerase is inactive → telomeres shorten with each division.
  • Eventually, cells reach a replicative limit (the Hayflick limit) and enter senescence.
  • This acts as a biological clock, limiting tissue renewal and contributing to ageing.

Implications for Immortality

  • Cells that reactivate telomerase escape senescence and can divide indefinitely.
  • Cancer cells often show high telomerase activity → essential for tumour immortality.
  • ~85–90% of human cancers have detectable telomerase activity.

TLDR:

Telomerase extends telomeres, allowing cells to bypass the end replication problem. In most somatic cells, its inactivity leads to gradual telomere shortening and cellular ageing. In contrast, germline and cancer cells maintain telomerase activity, granting them the ability to divide indefinitely—contributing to tissue renewal or tumour immortality, respectively.


5. Explain Why Telomerase Is an Attractive Target for Cancer Treatments

Why Target Telomerase in Cancer?

  • Key rationale: Most tumours depend on telomerase to sustain unlimited proliferation.
  • Targeting telomerase could limit tumour cell lifespan, pushing them toward senescence or apoptosis.

Therapeutic Strategies

Strategy Mechanism
Direct enzyme inhibitors Block telomerase’s reverse transcriptase activity
Antisense oligonucleotides Target the RNA template to prevent extension
Immunotherapy Target telomerase-expressing cells (e.g., vaccines, T-cells)
Small molecules Stabilise telomeric structures (e.g., G-quadruplexes) to inhibit access

Challenges of Telomerase-Based Therapy

  • Delayed effect: Cancer cells with long telomeres won’t die immediately.
  • Potential toxicity: May affect stem or germline cells with natural telomerase expression.
  • Escape pathways: Some cancers use ALT (Alternative Lengthening of Telomeres), a telomerase-independent mechanism.

TLDR:

Telomerase is a prime target in cancer because it’s active in most tumours but largely absent in healthy somatic cells. Inhibiting telomerase could limit cancer cell division. However, challenges include delayed therapeutic effects, potential off-target impacts, and alternative telomere maintenance pathways in some cancers.


L22: DNA Synthesis in the Lab

1. Explain the Relevance of Base Stacking/Base Pairing to Molecular Biology Techniques

Base Pairing

  • Complementary base pairing (A–T, G–C) via hydrogen bonds allows:

    • Primer-template binding in PCR
    • Specificity in Sanger sequencing, RT-PCR
    • Re-annealing of DNA strands when cooled

Example: In PCR, primers bind to their complementary DNA sequence through hydrogen bonding. Correct annealing is required for successful amplification.


Base Stacking

  • π–π interactions between adjacent planar bases stabilise the double helix.

  • Important for:

    • Melting temperature (Tm) of DNA
    • UV absorbance at 260 nm (used in spectrophotometry to estimate DNA concentration)

Example: ssDNA absorbs more UV light than dsDNA because base stacking is disrupted when strands are separated.


Impact on Techniques

Technique Role of Base Pairing/Stacking
PCR Primer annealing depends on base pairing; melting point influenced by GC content and stacking
Gel electrophoresis DNA denaturation is affected by GC content (more GC = higher Tm)
Spectrophotometry Based on absorbance change when dsDNA melts into ssDNA

TLDR:

Base pairing allows sequence-specific primer binding in molecular biology methods, while base stacking contributes to DNA stability and UV absorbance properties. Together, these weak interactions underpin key lab techniques like PCR, electrophoresis, and quantification by spectrophotometry.


2. Explain the Relevance of Backbone Charge to Molecular Biology Techniques

Structure of the DNA Backbone

  • Repeating sugar-phosphate backbone
  • Phosphate groups carry negative charges

Importance of Negative Charge

Technique Relevance
Gel electrophoresis DNA migrates toward the positive electrode because of its negatively charged phosphate backbone
Primer specificity Repulsion between negatively charged strands promotes specificity; magnesium ions (Mg²⁺) shield these repulsions and affect binding
DNA stability High salt concentrations (e.g., Mg²⁺) shield repulsive forces, increase Tm, but can reduce specificity in PCR if excessive

Visual Summary: Mg²⁺ in PCR

DNA Strand (-)   Mg²⁺   DNA Strand (-)
   ↓              ⇄        ↓
Repulsion       Shielding → Improved annealing
                            ↓
                 May reduce specificity

TLDR:

The phosphate backbone’s negative charge drives DNA migration in electrophoresis and influences hybridisation. Controlling ionic conditions (especially Mg²⁺) is essential for tuning strand annealing and primer specificity in techniques like PCR.


3. Describe How DNA Is Synthesised In Vitro in PCR

Overview of PCR (Polymerase Chain Reaction)

A technique to amplify specific DNA regions using thermal cycling.


Essential Components

Component Role
Template DNA Contains the target region to be amplified
DNA polymerase Adds dNTPs to extend from primers (e.g., Taq polymerase)
Primers (forward & reverse) Short DNA sequences that define the region of interest
dNTPs Building blocks for new DNA (dATP, dTTP, dCTP, dGTP)
Buffer + Mg²⁺ Provides optimal pH and ionic conditions

PCR Cycle

Step 1: Denaturation (~95°C)
- Heat separates dsDNA → ssDNA

Step 2: Annealing (~55–65°C)
- Primers bind to complementary sequences

Step 3: Extension (~72°C)
- Polymerase adds nucleotides to 3′ end of primer

→ Repeat 25–35 times (exponential amplification)

Polymerase Used: Taq Polymerase

  • Isolated from Thermus aquaticus
  • Thermostable: survives high temperatures
  • No proofreading: fast, but error-prone (acceptable in applications like genotyping)

PCR vs. Cellular Replication

Feature PCR Cellular DNA Replication
Strand separation Heat Helicase
Primer Synthetic DNA RNA primer by primase
Enzyme Taq polymerase DNA Pol III / δ
Proofreading No (Taq) Yes (high-fidelity polymerases)
Scope Targeted sequence Whole genome
Re-initiation Repeats per cycle Only once per S phase

TLDR:

PCR mimics natural DNA replication in vitro but uses heat to separate DNA strands and synthetic primers for targeted amplification. Taq polymerase extends DNA from primers without proofreading. Exponential DNA synthesis through repeated cycles makes PCR a rapid and powerful molecular biology tool.


4. Explain How Properties of DNA Polymerases Are Useful in PCR

Why Polymerase Choice Matters

DNA polymerases differ in fidelity, processivity, thermostability, and speed. The choice of polymerase directly impacts the success and specificity of PCR.


Key Properties Used in PCR

Property Relevance to PCR
Thermostability Essential to withstand repeated heating cycles during denaturation (95 °C). Taq polymerase remains active after >30 cycles.
5′→3′ polymerase activity Extends DNA strand from the primer. Core enzymatic function.
Lack of 3′→5′ exonuclease (in Taq) Increases speed but reduces fidelity (no proofreading).
Processivity Ability to synthesise long stretches of DNA without falling off the template.
Optimal activity at 72 °C Matches PCR extension temperature; maximises polymerase function.

Polymerase Options

Enzyme Notes
Taq polymerase Most common; high thermostability, low fidelity (error-prone)
Pfu polymerase From Pyrococcus furiosus; proofreading ability, higher fidelity
High-fidelity blends Engineered polymerases combining Taq with proofreading enzymes for both speed and accuracy

Considerations in Primer Design and Fidelity

  • Errors from low-fidelity polymerases (like Taq) can result in mutated PCR products.
  • For cloning, sequencing, or diagnostic applications, high-fidelity enzymes are preferred.

TLDR:

DNA polymerases used in PCR must be thermostable and active at high temperatures. Taq polymerase, the most common, is fast and heat-resistant but lacks proofreading, making it less accurate. For high-fidelity applications, proofreading polymerases (e.g. Pfu) are used. The choice of polymerase influences the accuracy, speed, and success of PCR.


5. Describe How DNA Is Synthesised In Vitro in Reverse Transcription for RNA Analysis

Reverse Transcription (RT)

  • Converts RNA into complementary DNA (cDNA) using a reverse transcriptase enzyme.
  • Enables RNA-based analyses using DNA-compatible methods (e.g., PCR, sequencing).

Reverse Transcriptase Enzymes

Source Notes
Retroviruses (e.g., MMLV, AMV) Naturally encode reverse transcriptase
Engineered variants Optimised for thermal stability and reduced RNase activity

Reaction Components

Component Function
RNA template Usually mRNA extracted from cells
Primer Provides a 3′-OH; may be:
– Oligo(dT) (binds poly-A tail)
– Random hexamers
– Gene-specific primers
dNTPs Building blocks for DNA
Reverse transcriptase Synthesises DNA from RNA template
RNase inhibitor Prevents degradation of RNA

Steps in RT Reaction

1. RNA + primer anneal
2. Reverse transcriptase synthesises cDNA (5′→3′)
3. RNA is degraded (optionally by RNase H)
4. Result: single-stranded cDNA

The cDNA can then be amplified by PCR using gene-specific primers (→ RT-PCR).


Applications of RT:

  • Gene expression analysis
  • Viral RNA detection (e.g., HIV, SARS-CoV-2)
  • RNA sequencing library prep

RT Reaction Overview Diagram:

mRNA (poly-A tail)
   ↓ primer (oligo-dT or random hexamer)
Reverse Transcriptase
   ↓
cDNA synthesis → cDNA + PCR = gene expression profiling

TLDR:

Reverse transcription synthesises complementary DNA from RNA templates using reverse transcriptase enzymes. This enables analysis of gene expression and RNA viruses via standard DNA-based tools like PCR. The primer choice determines whether all mRNA or specific genes are converted into cDNA.


6. Describe How DNA Is Synthesised In Vitro in DNA Sequencing

Focus: Sanger Sequencing (Chain-Termination Method)

Developed by Frederick Sanger in 1977, this method remains foundational in molecular biology for sequencing short to medium-length DNA fragments.


Core Principle

Sanger sequencing uses selective incorporation of dideoxynucleotides (ddNTPs), which terminate DNA synthesis when incorporated.

Molecule Structure Result
dNTP Has 3′-OH Allows elongation
ddNTP No 3′-OH Terminates DNA strand extension

Reaction Components

Component Function
Template DNA Sequence to be determined
Primer Anneals to template; provides 3′-OH
DNA polymerase Extends primer using dNTPs
dNTPs Normal nucleotides for elongation
Fluorescently-labelled ddNTPs Chain terminators (one for each base)
Buffer + Mg²⁺ Required for polymerase activity

Reaction and Detection

  1. Primer binds to template
  2. Polymerase extends strand
  3. Random ddNTP incorporation halts extension
  4. Fragments of different lengths are generated, each ending in a labelled ddNTP
  5. Capillary electrophoresis separates fragments by size
  6. Laser detection reads terminal ddNTP fluorescence

Flowchart: Sanger Sequencing Overview

1. DNA template + primer + polymerase
2. Add dNTPs + fluorescent ddNTPs
3. Chain termination at random positions
4. Size separation via capillary electrophoresis
5. Laser reads terminal base → sequence reconstructed

Output: Chromatogram

  • Each peak = base identity at that position
  • Colour of peak = ddNTP fluorophore
  • Sequence is read from small (5′) to large (3′) fragments

TLDR:

Sanger sequencing uses labelled ddNTPs to terminate DNA synthesis at specific bases, creating fragments of varying lengths. These are separated and detected via fluorescence, revealing the DNA sequence. The method is accurate, robust, and still widely used for short sequences.


7. Explain How Properties of DNA Polymerases Are Useful in Sanger Sequencing

Key Polymerase Requirements

Property Why It Matters in Sequencing
5′→3′ polymerase activity Adds nucleotides from primer for strand synthesis
Efficient ddNTP incorporation Must be able to use both dNTPs and ddNTPs as substrates
No 3′→5′ proofreading Proofreading would remove ddNTPs, ruining termination accuracy
High processivity Ensures full extension until termination point

Sanger sequencing polymerases are often engineered to lack exonuclease (proofreading) activity to preserve ddNTP incorporation events.


Polymerase Commonly Used:

  • Modified Taq polymerase (no proofreading, thermostable)
  • Sequenase: modified T7 DNA polymerase with improved ddNTP incorporation

Special Considerations:

  • Fidelity is less critical than in PCR or cloning — minor polymerase errors won’t affect the sequence if ddNTP incorporation is successful
  • Thermostability not essential (unlike PCR), because sequencing reactions are usually done at lower temperatures

TLDR:

DNA polymerases used in Sanger sequencing must efficiently incorporate both dNTPs and ddNTPs without proofreading activity, to allow accurate chain termination. Enzymes like Sequenase or modified Taq polymerase are preferred for their high processivity and compatibility with fluorescent ddNTPs.


L23: The Eukaryotic Genome

1. Account for the Large Amount of Non-Coding DNA in the Genomes of Complex Organisms

Key Categories of Non-Coding DNA

Type Description
Introns Non-coding regions within genes; spliced out during mRNA processing. Can comprise >90% of a gene’s length.
Repetitive DNA Includes telomeres, centromeres, and Variable Number Tandem Repeats (VNTRs); important for structural and forensic purposes.
Transposons Mobile DNA sequences; can copy and insert themselves into new genomic locations (e.g. LINEs, SINEs).
Pseudogenes Non-functional copies or remnants of once-active genes, often arising from duplication or retrotransposition.
Regulatory elements Sequences controlling gene expression, including enhancers, silencers, and insulators.

Example: CaSR Gene Composition

  • ~95% intronic DNA, only ~5% exonic.
  • Demonstrates the dominance of non-coding sequence even within protein-coding genes.

TLDR:

The human genome contains vast amounts of non-coding DNA, including introns, repetitive sequences, and pseudogenes. While only ~1–2% of the genome encodes proteins, the rest plays structural, regulatory, or unknown roles—illustrating the complexity beyond simple coding functions.


2. Describe the Packaging of DNA with Histones to Form Nucleosomes, Nucleosome Structure

Chromatin and Nucleosome Structure

Feature Description
Chromatin Complex of DNA + proteins (primarily histones).
Nucleosome The fundamental unit of chromatin: ~145 bp of DNA wrapped around a histone octamer.
Histone Octamer 2 each of H2A, H2B, H3, and H4.
Linker DNA ~50 bp between nucleosomes; bound by H1 histone.

DNA-Histone Interaction

  • Histone tails (rich in lysine and arginine) are positively charged.
  • These interact with the negatively charged phosphate backbone of DNA via electrostatic attraction.
  • Not sequence-specific: allows for dynamic wrapping/unwrapping as needed.

TLDR:

DNA is tightly wrapped around histone octamers to form nucleosomes, the basic units of chromatin. Electrostatic interactions between positively charged histone tails and negatively charged DNA enable compact storage of ~2 meters of DNA in a ~10 μm nucleus, while still allowing access for gene expression.


3. Describe How Histone Modifications Can Influence Gene Expression

Key Modifications and Their Effects

Modification Enzymes Effect on DNA Packing Impact on Gene Expression
Acetylation HATs (Histone Acetyltransferases) Loosens DNA-histone interaction (neutralises lysine’s positive charge) Activates transcription by increasing accessibility
Deacetylation HDACs (Histone Deacetylases) Restores positive charge on histones → tighter DNA wrapping Represses transcription
Methylation HMTs (Histone Methyltransferases) Depends on which residues are modified Can activate or repress, depending on context and reader proteins

Conceptual Analogy:

  • Acetylation = loosening the grip = “open book”
  • Deacetylation = tightening the grip = “closed book”
  • Methylation = “waving a flag” to recruit specific proteins (activators or repressors)

TLDR:

Histone modifications such as acetylation and methylation modulate how tightly DNA is packed, thereby controlling access to genetic information. Acetylation generally enhances transcription, while methylation’s effect depends on context. These modifications are reversible and dynamic, enabling fine-tuned regulation of gene expression.


4. Understand How Histone Acetyltransferases (HATs) and Histone Deacetylases (HDACs) Influence the Binding Interaction Between Histones and the DNA Backbone

Key Molecular Players

Enzyme Function Mechanism
HATs (Histone Acetyltransferases) Add acetyl groups to lysine residues on histone tails Neutralise positive charge → weaken interaction with DNA
HDACs (Histone Deacetylases) Remove acetyl groups Restore positive charge on lysine → strengthen interaction with negatively charged DNA

Electrostatic Basis:

  • DNA phosphate backbone = negatively charged
  • Histone tails = positively charged lysines
  • Acetylation neutralises lysine → reduces affinity → loosens chromatin
  • Deacetylation restores charge → tightens chromatin

Functional Consequences

  • HAT activity → chromatin opens → transcription factors can bind → gene activation
  • HDAC activity → chromatin condenses → reduced accessibility → gene repression

TLDR:

HATs and HDACs modify histone charge states to regulate DNA-histone interactions. Acetylation loosens chromatin for gene expression; deacetylation restores tight packing and silences genes. This electrostatic regulation is central to dynamic transcriptional control.


5. Explain How Histone Modifications Can Be Used to Remodel Regions of Chromatin to Increase or Decrease How Tightly It Is Packed

Types of Chromatin

Chromatin State Features Gene Activity
Euchromatin Loosely packed, accessible Active transcription
Heterochromatin Tightly packed, inaccessible Repressed transcription

Histone Code Hypothesis

  • Specific combinations of histone modifications (the “histone code”) recruit remodelling complexes.

  • These proteins interpret the code and restructure chromatin by:

    • Sliding nucleosomes
    • Ejecting nucleosomes
    • Replacing histone variants

Key Modifications and Their Impact

Modification Example Effect
H3K9 acetylation Promotes open chromatin, active transcription
H3K9 methylation Recruits HP1, promotes heterochromatin formation
H3K27 methylation Repressive mark, key in development (e.g., Polycomb silencing)

Role of Remodelling Complexes

  • ATP-dependent chromatin remodellers respond to histone marks and reposition nucleosomes to:

    • Open chromatin for gene expression
    • Close chromatin for gene silencing

TLDR:

Histone modifications act as molecular signals that recruit chromatin remodelling proteins. These complexes reshape nucleosome positioning to either loosen or condense chromatin structure, modulating accessibility and regulating gene expression accordingly.


6. Describe How Pluripotent Stem Cells and Terminally Differentiated Cells Differ in Their Ability to Access and Transcribe Certain Genome Regions

Epigenetic Landscape: Open vs Closed

Cell Type Chromatin State Gene Access
Pluripotent Stem Cells Broadly open chromatin Can access and potentially express most genes
Terminally Differentiated Cells Highly restricted chromatin Many genes are silenced and inaccessible

Stem Cells

  • Possess global euchromatin
  • Express pluripotency transcription factors (e.g. Oct4, Sox2)
  • Can transcribe genes for many cell lineages
  • High H3K4 methylation and H3K9 acetylation → active marks

Differentiated Cells

  • Lineage-specific gene expression
  • Other regions become permanently silenced (heterochromatin)
  • Increased H3K27 or H3K9 methylation (repressive)

Implications

  • Plasticity in stem cells relies on open chromatin
  • Differentiation involves epigenetic locking of transcriptional states
  • Cell identity is maintained by histone modifications, not just DNA sequence

TLDR:

Pluripotent stem cells have open chromatin and can access most of the genome, enabling them to differentiate into any cell type. Terminally differentiated cells have restricted chromatin, limiting gene access to only lineage-appropriate genes. Histone modifications enforce this transcriptional memory and cell identity.


L24: Prokaryotic Transcription

1. Stages and Basic Mechanisms of Prokaryotic Transcription

Overview of Transcription

Transcription is the DNA-to-RNA step of the central dogma, carried out in prokaryotes by a single RNA polymerase enzyme.

Three Main Stages:

1. Initiation

  • RNA polymerase (with σ subunit) binds to a promoter upstream of the gene.
  • The σ subunit scans DNA, finds the promoter, and facilitates tight binding.
  • DNA unwinds, forming the transcription bubble.
  • First few nucleotides are added; once ~10 nt are added, σ is released.

2. Elongation

  • RNA polymerase moves along DNA, synthesising RNA 5′ → 3′.
  • It uses ribonucleotides (ATP, UTP, CTP, GTP).
  • No primer is required; synthesis is de novo.
  • A short RNA-DNA hybrid (~8 nt) forms in the transcription bubble.
  • Elongation rate: ~50 nt/sec (slower than DNA replication).
  • RNA pol has limited proofreading via backtracking and excision.

3. Termination

  • Signals to stop transcription arise from the RNA, not DNA.

  • Two major mechanisms:

    • Intrinsic (Rho-independent): GC-rich hairpin + U-rich tail → polymerase pauses and dissociates.
    • Rho-dependent: Rho binds to RNA, travels along it, and knocks RNA pol off the DNA when it reaches it.

TLDR:

Transcription in prokaryotes involves three stages: initiation (promoter recognition by RNA polymerase + σ), elongation (RNA synthesis 5′→3′, no primer), and termination (via intrinsic or Rho-dependent signals in the RNA). It’s a precise but faster and slightly more error-prone process than DNA replication.


2. Features of Promoters

What Is a Promoter?

  • A DNA sequence upstream of a gene where RNA polymerase binds to begin transcription.
  • It contains specific sequence motifs that direct binding and positioning.

Key Promoter Elements in Bacteria:

  • –10 region (TATAAT): Also called the Pribnow box.
  • –35 region (TTGACA)
  • These sequences are based on a consensus, derived from many bacterial promoters.

Positioning: –35 and –10 sequences are ~17 bp apart (optimal spacing for RNA polymerase binding).


Promoter Recognition

  • The σ subunit of RNA polymerase recognises these sequences.
  • It decreases general DNA binding affinity, allowing RNA pol to search efficiently for proper promoters.

Consensus Sequence Example:

5′ - TTGACA ------17bp------ TATAAT - +1 - 3′
                     ↑                   ↑
                 –35 region         –10 region

TLDR:

Prokaryotic promoters contain conserved –10 and –35 regions upstream of the transcription start site. These are recognised by the RNA polymerase σ subunit and are essential for correct and efficient transcription initiation.


3. Role and Function of Promoters

Promoter Role:

  • Defines where transcription starts.
  • Controls transcription frequency — the “volume knob” for gene expression.

Promoter Strength:

  • Determined by:

    • How closely the sequence matches the consensus.
    • The spacing between the –10 and –35 regions (ideal: 17 bp).
  • Strong promoters:

    • Closely match consensus
    • → High transcription frequency (e.g., every 2 seconds)
  • Weak promoters:

    • Diverge from consensus
    • → Low transcription frequency (e.g., every 10 minutes)

Analogy used in lecture: A familiar-looking bus stop (strong promoter) vs. an ambiguous one (weak promoter) affects how likely RNA pol is to stop.


TLDR:

Promoters determine where and how frequently a gene is transcribed. Strong promoters closely match the consensus and promote frequent transcription; weak ones lead to low expression levels. The promoter’s sequence and spacing are critical for efficient RNA polymerase binding.


4. Effect of Promoter Strength on Transcription Rate

Definition of Promoter Strength:

  • Promoter strength refers to how efficiently RNA polymerase can recognise, bind, and initiate transcription at a promoter.

Influencing Factors:

Factor Effect
Match to consensus sequences Closer match = stronger binding = more frequent transcription
Spacing between –10 and –35 regions Optimal spacing (~17 bp) allows best positioning of RNA pol
DNA supercoiling and local sequence context May influence accessibility or σ subunit recognition

Transcription Rate:

  • Strong promoters → transcription initiation every ~2 seconds
  • Weak promoters → initiation every ~10 minutes

Important note: Promoter strength is constitutive, meaning it’s intrinsic to the DNA sequence. Regulation (e.g. via repressors) is layered on top of this.


TLDR:

Promoter strength determines how often transcription is initiated, based on the match to consensus and spacing of the –10/–35 boxes. Strong promoters drive frequent transcription; weak ones yield rare transcription.


5. Transcriptional Regulation of Lac and trp Operons

Operons = Units of Coordinated Gene Expression

Feature Lac Operon trp Operon
Inducible or repressible? Inducible (turned on when needed) Repressible (turned off when product abundant)
Default state Off (blocked by repressor) On (until trp builds up)
Inducer/Corepressor Allolactose (removes repressor) Tryptophan (activates repressor)
Repressor binding site Operator (overlaps with promoter) Operator

Lac Operon (Catabolic Regulation)

  • Purpose: Encodes enzymes to metabolise lactose.
  • In absence of lactose: LacI repressor binds operator → blocks RNA pol.
  • When lactose is present: allolactose binds repressor → causes it to fall off → RNA pol can transcribe.

trp Operon (Anabolic Regulation)

  • Purpose: Encodes enzymes to synthesise tryptophan.
  • When tryptophan is scarce: repressor inactive → transcription ON.
  • When tryptophan is plentiful: it binds and activates repressor → binds operator → transcription OFF.

Diagrams:

Lac Operon:

No lactose → repressor binds → transcription OFF  
Lactose → allolactose binds repressor → repressor releases → transcription ON

trp Operon:

Low Trp → repressor inactive → transcription ON  
High Trp → Trp binds repressor → repressor binds operator → transcription OFF

TLDR:

The lac operon is inducible—turned ON in the presence of lactose, while the trp operon is repressible—turned OFF in the presence of tryptophan. These classic operons demonstrate how bacteria regulate gene expression based on metabolic needs through repressor-operator interactions.


6. Role and Function of Repressors

Repressors = Negative Regulators

  • Bind to operator regions, which overlap or are near promoters.
  • Block access or progress of RNA polymerase, preventing transcription.

Mechanism of Action:

Type Example Binding Trigger Result
Inducible Lac repressor Inactivated by allolactose Transcription ON
Repressible trp repressor Activated by tryptophan Transcription OFF

Features:

  • Repressors are usually DNA-binding proteins.
  • Bind to operator sequences (not the core promoter itself).
  • Often part of feedback regulation loops (e.g. product of the operon affects its own transcription).

TLDR:

Repressors are proteins that bind to operators and prevent RNA polymerase from initiating transcription. Their activity is modulated by small molecules (e.g. allolactose or tryptophan) to regulate operons like lac and trp in response to environmental conditions.


L25: Eukaryotic Transcription

1. Differences Between Prokaryotic and Eukaryotic Transcription

Feature Prokaryotic Transcription Eukaryotic Transcription
Location Cytoplasm Nucleus (then mRNA exported to cytoplasm)
RNA Polymerase One type Three distinct polymerases (I, II, III)
Initiation Requires σ factor Requires many general transcription factors (TFII family)
Promoters Simple: –10 (TATAAT), –35 (TTGACA) Complex: TATA box, Inr, CAAT, GC, DPE
Transcription and Translation Coupled (simultaneous) Separated in space and time
RNA Processing None (mRNA often used immediately) Extensive: 5′ capping, splicing, 3′ poly-A tail
Termination Rho-dependent or intrinsic Cleavage + polyadenylation; distinct mechanisms

TLDR:

Eukaryotic transcription is more complex than prokaryotic transcription due to compartmentalisation, extensive RNA processing, multiple RNA polymerases, and reliance on large protein complexes. Prokaryotes streamline transcription with a single RNA polymerase and direct mRNA translation.


2. Types and Roles of Eukaryotic RNA Polymerases

RNA Polymerase Location Function Sensitivity to α-amanitin
Pol I Nucleolus Synthesises 45S rRNA precursor → 18S, 5.8S, 28S rRNAs Insensitive
Pol II Nucleoplasm Synthesises mRNA, some snRNA, miRNA Highly sensitive
Pol III Nucleoplasm Synthesises tRNA, 5S rRNA, some snRNA Moderately sensitive
  • Pol II is the key polymerase for transcription of protein-coding genes.
  • Each polymerase has distinct promoter preferences and subunit compositions.
  • Pol I and III are specialised and operate in restricted contexts (e.g., ribosome biogenesis, tRNA processing).

TLDR:

Eukaryotic cells use three RNA polymerases: Pol I (rRNA), Pol II (mRNA and snRNA), and Pol III (tRNA, 5S rRNA). Pol II is the most important for gene expression and is highly sensitive to transcriptional inhibitors like α-amanitin.


3. Types of Eukaryotic Promoter Elements

General Features:

  • Promoters are cis-acting DNA elements located near the transcription start site.
  • Recognised by transcription factors, not RNA polymerase directly.

Core Promoter Elements for RNA Polymerase II:

Promoter Element Location Function
TATA box ~–25 Highly conserved; facilitates transcription initiation via TBP (TATA-binding protein)
CAAT box –40 to –150 Enhancer-like; binds regulatory proteins
GC box –40 to –150 Often found in housekeeping genes; binds Sp1 TF
Initiator (Inr) ~+1 Defines the transcription start site; can function without TATA box
Downstream Promoter Element (DPE) +30 Found in TATA-less promoters; functions with Inr

Promoter combinations vary, adding flexibility and complexity to gene regulation.


Visual Summary:

5’ ——– GC/CAAT —— TATA —— +1 (Inr) —— DPE —— 3’

TLDR:

Eukaryotic promoters are more diverse and modular than prokaryotic ones. They often include a TATA box, CAAT or GC boxes, and additional downstream elements. Their arrangement affects transcription strength, tissue specificity, and RNA Pol II recruitment.


4. Role of Enhancers

What Are Enhancers?

  • Cis-regulatory elements that increase the rate of transcription initiation.

  • Can act:

    • Upstream or downstream
    • Thousands of base pairs away from the promoter
    • In either orientation (5′→3′ or 3′→5′)

How They Work:

  • Bind activator proteins (enhancer-binding transcription factors).

  • Through DNA looping, they are brought into proximity with the core promoter, facilitating:

    • Recruitment of coactivators
    • Stabilisation of general transcription machinery (e.g. TFIID)
    • Enhanced assembly of the pre-initiation complex

Enhancer vs Promoter:

Feature Enhancer Promoter
Location Can be distant Close to TSS
Orientation Works bidirectionally Orientation-specific
Role Regulatory – boosts transcription Core – initiates transcription

TLDR:

Enhancers are distant regulatory DNA elements that increase transcription by looping into contact with the promoter. They work by binding activators that help stabilise the transcriptional machinery, making them crucial for gene-specific regulation.


5. Role of Transcription Factors

Two Main Classes:

Type Function
General Transcription Factors (GTFs) Required for all Pol II transcription (e.g., TFIID, TFIIB, TFIIE, TFIIF, TFIIH)
Regulatory (Specific) Transcription Factors Bind enhancers, silencers, or response elements to regulate gene-specific transcription

Key General TFs in the Pre-Initiation Complex:

Factor Role
TFIID Includes TBP (TATA-binding protein); binds the TATA box
TFIIH Helicase activity to unwind DNA; kinase activity to phosphorylate RNA Pol II
TFIIB, TFIIE, TFIIF Recruit and stabilise RNA Pol II at the promoter

Regulatory TF Domains:

Domain Function
DNA-binding domain (DBD) Recognises specific DNA sequences (e.g., zinc fingers, helix-turn-helix)
Activation/repression domain Interacts with coactivators or corepressors to modulate transcription
Dimerisation domain Allows homo-/heterodimer formation for increased binding specificity

TLDR:

Transcription factors are proteins that control gene expression by guiding RNA polymerase to specific promoters (GTFs) or regulating its activity via enhancers (regulatory TFs). Their modular structure enables DNA recognition and interaction with other regulatory proteins.


6. Hormone Regulation of Gene Expression

Hormones as Transcriptional Signals:

  • Steroid hormones (e.g., cortisol, estrogen, testosterone) are lipid-soluble and pass through membranes.
  • Bind to nuclear hormone receptors (NHRs) that act as ligand-activated transcription factors.

Mechanism:

1. Hormone diffuses into cell
2. Binds nuclear receptor → conformational change
3. Receptor-hormone complex binds **hormone response element (HRE)** in DNA
4. Recruits coactivators or corepressors → modifies gene expression

Examples:

Hormone Receptor Response Element Target Effect
Cortisol Glucocorticoid receptor GRE Anti-inflammatory genes
Estrogen Estrogen receptor ERE Reproductive and growth genes

TLDR:

Steroid hormones regulate gene expression by binding to intracellular receptors that act as transcription factors. These hormone–receptor complexes target specific DNA response elements to turn genes on or off based on physiological signals.


7. Drugs That Target Hormone Receptors to Alter Gene Expression

Mechanism of Action:

  • Many drugs mimic or block hormone action by binding to nuclear hormone receptors.
  • These drugs modulate gene expression in hormone-responsive cells (e.g., breast, prostate, immune cells).

Drug Examples:

Drug Target Type Effect
Tamoxifen Estrogen receptor (ER) SERM (Selective Estrogen Receptor Modulator) Antagonist in breast → used in ER+ breast cancer
Dexamethasone Glucocorticoid receptor Agonist Suppresses inflammation by activating anti-inflammatory genes
RU486 (Mifepristone) Progesterone receptor Antagonist Blocks progesterone → used in medical abortion

TLDR:

Hormone-based drugs modulate gene expression by targeting nuclear receptors. Some (like dexamethasone) mimic hormones to stimulate gene expression, while others (like tamoxifen or RU486) block hormone receptors to suppress specific gene programs, including those involved in cancer and inflammation.


L26: Post-Transcriptional Processing

1. Processing Events for rRNA

Overview:

  • rRNAs (18S, 5.8S, 28S) are transcribed by RNA polymerase I as a single precursor: 45S pre-rRNA.
  • Processing occurs in the nucleolus.

Steps in rRNA Processing:

Step Description
1. Nucleotide Modification Methylation and pseudouridine formation on specific bases or sugars; enhances ribosome stability and function
2. Assembly with Ribosomal Proteins Forms large ribonucleoprotein (RNP) complex
3. Cleavage 45S pre-rRNA is cleaved into 18S (small subunit), 5.8S and 28S (large subunit components)

5S rRNA is transcribed separately by RNA polymerase III, outside the nucleolus.


TLDR:

The 18S, 5.8S, and 28S rRNAs are transcribed as a 45S precursor by RNA Pol I, modified, assembled with proteins, and cleaved into mature forms. The 5S rRNA is transcribed separately by RNA Pol III. These steps ensure proper ribosome assembly and function.


2. Processing Events for tRNA

Overview:

  • tRNAs are transcribed by RNA polymerase III and undergo multiple processing steps before becoming mature.

Steps in tRNA Processing:

Step Description
1. 5′ and 3′ End Trimming RNases remove extra sequences from both ends
2. Addition of CCA at 3′ End Universal 3′-CCA tail is added enzymatically (not encoded) → site of amino acid attachment
3. Base Modifications Includes methylation and pseudouridylation (modifications enhance stability and function)
4. Intron Removal and Ligation If present, intron is spliced and exons ligated to form a functional anticodon loop

TLDR:

Pre-tRNAs undergo end trimming, 3′ CCA addition, base modifications, and splicing of introns. These steps are essential to produce a mature tRNA capable of delivering amino acids during translation.


3. Processing Events for mRNA

Overview:

  • In eukaryotes, mRNAs are extensively processed in the nucleus before being exported to the cytoplasm.
  • Processing begins co-transcriptionally (while RNA is still being synthesised).

Major mRNA Processing Events:

1. 5′ Capping

Feature Description
When After ~25 nt are transcribed
How 3 enzyme steps → adds a 7-methylguanosine cap via 5′–5′ linkage
Function Protects mRNA from 5′ exonucleases; enhances translation efficiency

2. 3′ Polyadenylation

Feature Description
Signal AAUAAA motif triggers cleavage
Process Cleaved by nucleases, then ~250 adenosines added by poly(A) polymerase (not transcribed from DNA)
Function Enhances mRNA stability and translation; does not affect nuclear export
Exception Histone mRNAs: no poly(A) tail → form stem-loop structures instead

3. Splicing

Feature Description
Purpose Remove non-coding introns and join exons
Signals 5′ splice site (GU), branch point (A), 3′ splice site (AG)
Catalysed by Spliceosome (snRNAs + proteins)
Coupled with transcription Via phosphorylation of RNA Pol II CTD tail

TLDR:

Eukaryotic mRNAs are capped at the 5′ end, polyadenylated at the 3′ end, and spliced to remove introns. These modifications protect mRNA, promote translation, and are essential for proper gene expression. All processing steps are tightly coupled with transcription.


4. Roles of 5′ Capping and 3′ Polyadenylation

5′ Capping

Feature Description
Structure 7-methylguanosine (m⁷G) cap linked via 5′–5′ triphosphate bridge
Added by Capping enzyme complex soon after transcription begins (~25 nt)
Functions
• Protects mRNA from degradation
• Aids nuclear export
• Required for ribosome recognition and translation initiation (via cap-binding proteins)

3′ Polyadenylation

Feature Description
Triggered by AAUAAA signal sequence upstream of cleavage site
Process
  1. Cleavage by endonucleases
  2. Addition of ~250 A residues by poly(A) polymerase
  3. Bound by poly(A)-binding proteins (PABPs) | | Functions | • Stabilises mRNA (protects from 3′ exonucleases) • Enhances translation via PABP interactions • Does not affect nuclear export (unlike 5′ cap) • Exceptions: histone mRNAs are not polyadenylated

TLDR:

The 5′ cap protects mRNA and is essential for translation initiation, while the 3′ poly(A) tail stabilises mRNA and boosts translation efficiency. Both are added co-transcriptionally and are crucial for proper gene expression in eukaryotic cells.


5. Features and Importance of Splicing

Splicing Overview:

Feature Description
Purpose Removes introns (non-coding) and joins exons (coding)
Signals
• 5′ splice site: GU
• Branch point: A
• 3′ splice site: AG
Catalysed by Spliceosome (complex of snRNAs: U1, U2, U4, U5, U6 + proteins)
Coupled with transcription Via phosphorylation of RNA Pol II C-terminal domain (CTD)
Energetics Requires ATP, though cleavage and ligation are transesterification reactions

Spliceosome Action Summary:

1. U1 binds 5′ splice site
2. U2 binds branch point A
3. U4/U5/U6 recruited
4. Intron looped and cut → lariat structure
5. Exons joined, lariat degraded

Why Splicing Matters:

  • Ensures correct reading frame
  • Prevents synthesis of non-functional proteins
  • Allows post-transcriptional regulation of gene expression

TLDR:

Splicing removes introns and joins exons via the spliceosome, using conserved GU–AG boundaries and a branch point A. This editing step is critical for generating functional proteins and is tightly linked to transcription.


6. Alternative Splicing as a Means to Generate Protein Diversity

What Is Alternative Splicing?

  • The process by which different combinations of exons are joined to produce multiple mRNA variants from a single gene.

Types of Alternative Splicing:

Type Example
Exon skipping/inclusion Exon 3 included in one isoform but skipped in another
Mutually exclusive exons Only one of two exons used
Alternative 5′/3′ splice sites Different start/end sites for the same exon
Intron retention Introns may be retained and translated in some contexts

Regulation:

  • Controlled by splice enhancers and silencers (cis-elements)
  • Bound by SR proteins (activators) and hnRNPs (repressors)

Biological Significance:

  • Increases proteomic diversity without increasing gene count
  • Critical for cell-type-specific expression and developmental regulation
  • Errors can cause disease (e.g., spinal muscular atrophy, β-thalassemia)

TLDR:

Alternative splicing allows a single gene to produce multiple protein isoforms. It is regulated by RNA-binding proteins and is essential for tissue specificity, adaptability, and biological complexity in eukaryotes.


7. mRNA Transport After Processing

Overview of mRNA Export:

Step Description
Export form Mature mRNA is exported as messenger ribonucleoprotein (mRNP) complex
Gate Transported through nuclear pore complexes (NPCs)
Signals Cap-binding proteins (CBPs), exon junction complexes (EJCs), and poly(A)-binding proteins help mark mRNA as export-ready
Directionality Export is unidirectional (nucleus → cytoplasm)
Post-export changes CBPs are replaced by eIF4E, allowing translation initiation in cytoplasm

Quality Control:

  • Improperly processed mRNA is retained in the nucleus or degraded.
  • Export is tightly linked to successful splicing, capping, and polyadenylation.

TLDR:

Once capped, polyadenylated, and spliced, mRNA is exported from the nucleus through nuclear pores as a mature mRNP. Export is highly regulated to ensure only fully processed transcripts reach the cytoplasm for translation.


L27 & L28: Translation in Prokaryotes and Eukaryotes

1. Properties of Nucleic Acids vs Amino Acids

Property Nucleic Acids Amino Acids
Function Store and transmit genetic information Build functional proteins, enzymes, signalling molecules
Structure Similar backbones with variable bases (A, U/T, C, G) Diverse side chains (charged, polar, nonpolar, aromatic, etc.)
Chemical Makeup Sugar-phosphate backbone + nitrogenous bases Central carbon with amino, carboxyl, hydrogen, and R-group
Charge Negatively charged due to phosphate groups Varies (acidic, basic, neutral) depending on R-group
Role in Biology Information storage & transfer (DNA/RNA) Functional diversity in metabolism and cellular structure

TLDR: Nucleic acids are chemically uniform and function as information carriers, while amino acids are structurally diverse to support a wide range of biochemical roles.


2. Features of the Genetic Code

Feature Description
Triplet Code 3 nucleotides = 1 codon = 1 amino acid
Universal Used by nearly all organisms with few exceptions (e.g. protozoa)
Non-overlapping Codons are read in strict triplets, without overlaps
No punctuation No nucleotide set aside as a “stop” base; stop codons are specific triplets
Degeneracy Most amino acids are encoded by >1 codon (e.g. serine: 6 codons)
Directionality Always read 5′ to 3′

Why Degeneracy Matters:

  • Minimizes mutation impact – changes in the 3rd base often don’t alter the amino acid (silent mutation).
  • Provides error-buffering during transcription/translation.

TLDR: The genetic code is a robust, redundant system that translates RNA into protein with high reliability. Degeneracy buffers against harmful mutations.


3. tRNA as an Adaptor Molecule

tRNA Characteristics:

Feature Detail
Length ~73–93 ribonucleotides
Structure Cloverleaf 2D shape, L-shaped 3D conformation
Key Sites Anticodon loop (binds mRNA), 3′ CCA tail (binds amino acid)
Post-transcriptional Modifications Methylation, base alterations help recognition

Function:

  • Bridges the language of nucleic acids (codons) and the language of proteins (amino acids).
  • Each amino acid has ≥1 matching tRNA.

Anticodon-Codon Interaction:

  • Anticodon binds to codon in antiparallel fashion (codon: 5′–3′; anticodon: 3′–5′).

TLDR: tRNAs act as molecular adaptors, matching mRNA codons to their respective amino acids. Their structure ensures compatibility with ribosomes, mRNA, and synthetases.


4. tRNA Binding to Codons and Amino Acids (Wobble Pairing)

Simple Base Pairing Model:

  • First two bases of codon pair strictly with anticodon.
  • Third base is more flexible — the wobble position.

Wobble Hypothesis:

First Anticodon Base (5′ end of tRNA) Recognised 3rd Codon Bases (3′ end of mRNA)
C G
A U
U A or G
G U or C
I (Inosine) U, C, or A

Key Concept:

  • Wobble allows one tRNA to recognise multiple codons that code for the same amino acid.
  • Reduces the number of distinct tRNA species needed.

Important Caveat:

  • Wobble only occurs at the third codon base / first anticodon base.
  • No wobble at the first two codon positions — strict Watson-Crick pairing.

TLDR: Wobble base pairing explains how cells use fewer tRNAs than codons. It provides flexibility in codon recognition without compromising translational accuracy.


5. Role of Aminoacyl-tRNA Synthetases

Function:

  • Enzymes that “charge” tRNAs with the correct amino acid.
  • Each amino acid has its specific synthetase (20 in total).

Charging Process:

Amino acid + ATP → aminoacyl-AMP + PPi
Aminoacyl-AMP + tRNA → aminoacyl-tRNA + AMP
Feature Description
Substrate Specificity Recognises both amino acid and tRNA anticodon loop
Proofreading Uses double-sieve mechanism to prevent incorrect charging
Energy Use Requires ATP (2 high-energy bonds used per aminoacylation)
Impact on Translation Errors at this step cause misincorporation of amino acids, not corrected during translation

TLDR: Aminoacyl-tRNA synthetases ensure the genetic code is correctly interpreted by precisely pairing each tRNA with its corresponding amino acid, forming aminoacyl-tRNA in an ATP-dependent, highly specific process.


6. Role of Ribosomes in Protein Synthesis

Prokaryotic Ribosome:

Subunit Composition Function
30S 16S rRNA + proteins Binds mRNA, decodes codons
50S 23S + 5S rRNA + proteins Catalyses peptide bond formation
70S 30S + 50S (assembled unit) Coordinates initiation, elongation, termination

Three Binding Sites:

Site Function
A (Aminoacyl) Incoming aminoacyl-tRNA binds here
P (Peptidyl) Holds the growing peptide chain
E (Exit) Where deacylated tRNA exits the ribosome

TLDR: Ribosomes orchestrate translation by positioning mRNA and tRNAs, forming peptide bonds, and ensuring directionality and fidelity of protein synthesis through their structured A, P, and E sites.


7. Stages of Translation and Its Features

Flowchart of Prokaryotic Translation:

1. Initiation
   - 30S binds Shine-Dalgarno (mRNA)
   - Initiator tRNA (fMet) binds P site
   - 50S joins → 70S complex forms

2. Elongation (repeats per codon)
   - EF-Tu brings aminoacyl-tRNA to A site
   - Peptide bond formed (23S rRNA)
   - EF-G drives ribosome translocation

3. Termination
   - Stop codon enters A site
   - Release factors hydrolyse peptide-tRNA bond
   - Ribosome dissociates
Factor Role
IF1, IF2, IF3 Initiation factors (assembly & accuracy)
EF-Tu, EF-G Elongation (delivery and movement)
RF1, RF2, RF3 Termination (release of peptide)

TLDR: Translation consists of initiation (ribosome assembly), elongation (codon-by-codon peptide synthesis), and termination (release of finished protein). Each stage uses GTP and accessory factors to maintain speed and accuracy.


8. Differences Between Prokaryotic and Eukaryotic Translation

Feature Prokaryotes Eukaryotes
mRNA recognition Shine-Dalgarno sequence pairs with 16S rRNA Ribosome scans from 5′ cap to start codon (AUG in Kozak sequence)
Initiator tRNA fMet-tRNA (formylated) Met-tRNA (non-formylated)
Coupling Transcription and translation coupled Occur in different compartments (nucleus vs cytoplasm)
mRNA structure Often polycistronic Always monocistronic
Ribosome size 70S (30S + 50S) 80S (40S + 60S)

TLDR: Prokaryotic translation is faster and coupled with transcription, using fMet and Shine-Dalgarno sequences. Eukaryotic translation is separated in space and time, involves scanning from a cap, and uses more initiation factors.


9. Inhibition of Protein Synthesis as a Mechanism of Drugs and Toxins

Selective Translation Inhibitors

Drug / Toxin Target Mechanism Application / Effect
Streptomycin 30S subunit Inhibits initiation, causes misreading Antibiotic (bactericidal)
Tetracycline 30S subunit Blocks A-site tRNA binding Antibiotic
Chloramphenicol 50S subunit Inhibits peptidyl transferase Broad-spectrum antibiotic
Erythromycin 50S subunit Blocks peptide exit tunnel Macrolide antibiotic
Cycloheximide 80S (eukaryotic) Inhibits elongation Lab use; toxic to humans
Ricin 28S rRNA (eukaryotic) Depurinates rRNA, inactivates ribosome Lethal toxin
Diphtheria toxin eEF-2 (eukaryotic) Blocks elongation by ADP-ribosylation Causes cell death

TLDR: Many antibiotics exploit differences in ribosomes to block bacterial translation. Others, like ricin and diphtheria toxin, target eukaryotic translation and can be lethal. These mechanisms underpin drug action and biotoxin lethality.


L29: Translational Regulation

1. Features and Application of Alternative Translation Start Sites

Key Concept:

  • Most eukaryotic mRNAs require a 5′ cap for ribosome binding and translation initiation.

  • Some mRNAs (especially viral or mitochondrial) lack a 5′ cap but still undergo translation using:

    • IRES: Internal Ribosome Entry Sites.

IRES: Internal Ribosome Entry Site

Feature Description
Structure Complex RNA loop in 5′ UTR before start codon
Function Recruits initiation factors or 40S subunit directly, bypassing the 5′ cap
Discovered in Polio virus (cap-independent translation)
Benefit to virus Can still be translated even if host cap-dependent translation is shut down

Experimental Application:

  • Co-expression of multiple proteins from a single mRNA (poly-cistronic in eukaryotes):

    • Gene of interest → IRES → reporter gene.
    • Allows independent translation of both proteins without interference.

Used in tracking gene expression in real time (e.g. with GFP or luciferase reporters).


TLDR:

Alternative translation start sites via IRES enable cap-independent translation. Found in viruses and essential host genes, IRES sequences are also exploited experimentally for bicistronic expression in eukaryotes.


2. Features of Nonsense-Mediated mRNA Decay (NMD)

What It Detects:

  • mRNAs with premature stop codons (due to nonsense mutations)

Mechanism:

  1. Splicing deposits Exon Junction Complexes (EJCs) downstream of exon-exon boundaries.

  2. During the first round of translation:

    • Normal mRNA: ribosome removes all EJCs.
    • Faulty mRNA: ribosome hits a premature stop codon before EJCs are removed.
  3. Remaining EJCs signal decay:

    • Recruit proteins that remove the 5′ cap.
    • mRNA is then degraded by 5′→3′ exonucleases.

TLDR:

NMD removes faulty mRNAs containing early stop codons. Exon junction complexes downstream of the premature stop mark them for degradation, preventing translation of truncated, potentially harmful proteins.


3. Features of Non-Stop mRNA Decay

What It Detects:

  • mRNAs that lack a stop codon, often due to mutations or processing errors.

Mechanism:

  1. Ribosome fails to encounter a stop codon.
  2. Translation continues into the poly(A) tail, producing a C-terminal polylysine tail.
  3. Ribosome stalls at the 3′ end → recruits decay proteins (e.g., Ski7).
  4. mRNA is degraded from 3′ to 5′ by exonucleases.
  5. The resulting defective protein (with polylysine tail) is also degraded by proteases.

TLDR:

Non-stop decay removes transcripts that lack a stop codon by recognizing stalled ribosomes at the 3′ end. Both the faulty mRNA and its abnormal protein product are rapidly degraded.


4. Features of No-Go mRNA Decay

What It Detects:

  • mRNAs where the ribosome stalls before a stop codon.

Causes of Ribosome Stalling:

Cause Example
Rare codons No matching tRNAs in host cell
Secondary structure Stable stem-loops block ribosome movement
Chemical damage Modified bases interfering with translation

Mechanism:

  1. Ribosome stalls mid-translation.
  2. Stalling triggers recruitment of rescue factors.
  3. Ribosome dissociates; mRNA is cleaved near the block site.
  4. Both 5′→3′ and 3′→5′ exonucleases degrade the fragments.

TLDR:

No-go decay eliminates mRNAs that cause ribosome stalling (e.g., due to rare codons or structured RNA). It rescues stalled ribosomes and degrades the faulty mRNA to maintain translation fidelity.


5. Mechanisms of Controlling Cellular Iron Levels

Overview:

Cells must tightly regulate iron levels to avoid:

  • Deficiency → impaired enzyme and oxygen transport function
  • Excess → toxic ROS via Fenton chemistry

Two Key Proteins:

Protein Function
Ferritin Iron storage protein (synthesised when Fe²⁺ is high)
Transferrin receptor (TfR) Iron uptake receptor (synthesised when Fe²⁺ is low)

Regulation Mechanism:

  • Iron Response Element (IRE) = conserved RNA hairpin structure
  • Iron Regulatory Proteins (IRPs) bind to IREs in mRNA depending on iron availability
Iron Level IRP Activity Effect on mRNAs
Low Iron IRPs bind IREs Block ferritin mRNA translation (don’t store iron); stabilize TfR mRNA (more iron uptake)
High Iron IRPs do not bind Ferritin translated (store excess iron); TfR mRNA degraded (reduce uptake)

TLDR:

Cellular iron homeostasis is regulated post-transcriptionally. IRPs bind IREs on ferritin and transferrin receptor mRNAs depending on iron levels, controlling translation or stability to balance iron storage and uptake.


6. Roles of the Iron Response Element (IRE) in Ferritin and Transferrin Receptor Synthesis

Ferritin mRNA:

Feature Detail
IRE location 5′ UTR
Low iron IRP binds IRE → blocks ribosome scanning → no ferritin made
High iron IRP detaches → ribosome initiates translation → ferritin made to store excess iron

Transferrin Receptor mRNA:

Feature Detail
IRE location 3′ UTR (multiple IREs)
Low iron IRP binds → protects mRNA from endonuclease cleavage → stable mRNA → more receptor
High iron IRP dissociates → mRNA degraded → less receptor → reduced iron import

Diagram Summary:

[Low Iron]
- IRP binds IREs on ferritin mRNA → translation OFF
- IRP binds IREs on TfR mRNA → stabilisation → translation ON

[High Iron]
- IRP released from IREs → ferritin mRNA translated
- TfR mRNA unprotected → degraded

TLDR:

The position of IREs dictates their effect: 5′ UTR binding blocks translation (ferritin), while 3′ UTR binding stabilizes mRNA (TfR). This elegant switch allows rapid post-transcriptional regulation of iron metabolism.


7. Roles of Small RNAs in Regulating mRNA Stability

Small RNAs = Post-Transcriptional Regulators

Class Size Function
miRNA (microRNA) ~21–25 nt Endogenous regulators of gene expression
siRNA (short interfering RNA) ~21 nt Often exogenous; used in RNA interference
piRNA (piwi-interacting RNA) ~24–30 nt Suppress transposons in germ cells

miRNA-Mediated Silencing (via RISC Complex):

  1. miRNA is transcribed, processed by Drosha and Dicer.

  2. Incorporated into RISC (RNA-Induced Silencing Complex).

  3. Guides RISC to target mRNAs by partial base pairing (usually 3′ UTR).

  4. If match is:

    • Perfect → mRNA cleavage and degradation
    • Imperfect → translation inhibition and deadenylation

Functions:

  • Regulate mRNA stability
  • Control translation rates
  • Essential in development, differentiation, and immune responses

Clinical Insight:

  • Dysregulated miRNAs are involved in cancer, metabolic diseases, and neurodegeneration.

TLDR:

Small RNAs (like miRNAs) regulate gene expression post-transcriptionally by binding mRNAs and either blocking translation or promoting degradation. They enable fine-tuned, rapid control of protein synthesis in response to cellular signals.


L30: Molecular Techniques I

1. Identify Some Uses of Recombinant DNA Technology

Recombinant DNA (rDNA) technology involves manipulating and recombining DNA from different organisms to produce new genetic combinations.

Key Applications:

Application Area Examples
Protein production Insulin, growth hormone, clotting factors, hepatitis B vaccine
Research tools Restriction enzymes, polymerases, glucose oxidase, fluorescent proteins (GFP), antibodies
Visualisation techniques Immunofluorescence, Western blotting, optogenetics, in vivo GFP tagging (neuronal networks, transgenic animals, tissue-specific expression)
Commercial products GloFish, fluorescent proteins in fish and mice
Agriculture Bt cotton (Cry proteins from Bacillus thuringiensis), golden rice (β-carotene genes), sterile mosquito vectors

TLDR:

Recombinant DNA technology is used across medicine, research, and agriculture. It enables protein production, creation of visualisation tools, and genetic enhancement of crops and animals. Examples include insulin, fluorescent tagging, Bt cotton, and sterility-based mosquito control.


2. Explain the Implications of the Universal Genetic Code for Cloning

Concept:

  • The genetic code is universal, meaning the same codons code for the same amino acids across all organisms.
  • This allows genes from one organism to be expressed in another, e.g., a jellyfish gene expressed in bacteria.

Implications:

  • A gene from humans (e.g., insulin) can be inserted into E. coli and the bacteria will produce human insulin.
  • Essential for cloning, GFP tagging, transgenic models, and synthetic biology.

TLDR:

Because the genetic code is universal, genes from any organism can be expressed in any other, enabling recombinant techniques. This foundational principle underpins all genetic engineering across species.


3. Describe the Differences Between Genomic DNA and cDNA

Feature Genomic DNA Complementary DNA (cDNA)
Source Entire genome (nucleus) Synthesised from processed mRNA (cytoplasm)
Contains introns Yes No (introns removed via mRNA splicing)
Represents Full genetic potential (all genes) Only expressed genes at the time of RNA extraction
Use cases GWAS, mutation analysis Gene expression studies, cloning for bacteria
Usability in bacteria Not suitable directly (contains introns) Ideal for prokaryotic expression (no introns)

TLDR:

Genomic DNA includes all genetic material, both coding and non-coding. cDNA is made from mRNA and contains only spliced, expressed genes, making it ideal for recombinant expression in systems like bacteria.


4. Describe How cDNA Is Generated

Step-by-Step Overview:

1. Extract Total RNA

  • Lyse cells and isolate total RNA (includes mRNA, tRNA, rRNA, etc.).

2. Isolate mRNA

  • Use oligo(dT) beads or columns that bind the poly(A) tails unique to mRNA.

3. Reverse Transcription

  • Use reverse transcriptase (an RNA-dependent DNA polymerase, RdDp) to synthesize first-strand cDNA.

4. Amplify cDNA

  • Add random primers (for whole transcriptome) or gene-specific primers (for targeted amplification).

  • Perform PCR to amplify cDNA, creating either:

    • cDNA libraries (multiple transcripts)
    • Specific gene products (targeted cloning)

Key Enzymes:

Enzyme Function
Reverse Transcriptase Synthesizes DNA from RNA (RdDp)
Taq Polymerase Amplifies DNA (DdDp)

TLDR:

cDNA is created by extracting mRNA, isolating it using its poly(A) tail, and reverse-transcribing it into DNA using reverse transcriptase. This cDNA can then be amplified for expression studies or cloning, especially useful in systems that cannot process introns (e.g., bacteria).


L31: Molecular Techniques II

1. Identify the Components of Cloning Vectors and Explain Their Functions

Cloning Vectors = Engineered Plasmids

Used to carry and propagate foreign DNA in host cells (e.g., E. coli).

Key Components:

Component Function
OriV (Origin of Replication) Allows plasmid to replicate independently within host cell
Antibiotic Resistance Gene Provides selectable marker; only transformed cells survive in antibiotic
Multiple Cloning Site (MCS) Cluster of unique restriction sites to insert gene of interest
Promoter Drives transcription of inserted gene (can be constitutive or inducible)

Diagram Summary:

[ OriV ]───[ Antibiotic Resistance ]───[ Promoter ]───[ MCS ]───(Insert Gene Here)
  • Optional: Fluorescent or reporter gene markers may be included downstream for screening recombinants.

TLDR:

Cloning vectors like plasmids are engineered with OriV for replication, an antibiotic resistance gene for selection, a promoter to drive gene expression, and a multiple cloning site for gene insertion.


2. Describe the Procedures Used to Generate Recombinant DNA Molecules

Step-by-Step Molecular Cloning Workflow

1. Isolate Plasmid DNA

  • Alkaline lysis with NaOH/SDS disrupts cell membranes.
  • Potassium acetate neutralizes pH; chromosomal DNA aggregates and precipitates.
  • Plasmid DNA remains soluble due to smaller size and re-annealing capacity.

2. Cut DNA with Restriction Enzymes

  • Restriction enzymes cleave DNA at specific sequences, usually palindromic.

  • Cuts can result in:

    • Sticky ends (preferred): Overhanging single strands allow specific base pairing.
    • Blunt ends: Less specific, harder to ligate directionally.

3. Insert DNA of Interest

  • Ligation: DNA ligase joins insert and plasmid via phosphodiester bond formation.

4. Transform Competent Cells

  • Competency induced with CaCl₂ and heat shock:

    • Ca²⁺ ions neutralise negative charges on DNA and membranes.
    • Cold → rigid membrane → heat → transient pores → DNA entry.
  • Very low efficiency (<10%) but sufficient for downstream work.

5. Select for Transformants

  • Grow bacteria on antibiotic-containing plates.
  • Only bacteria with plasmids (antibiotic resistance gene) survive.

6. Screen for Recombinants

  • Not all surviving plasmids are recombinant.
  • Further screening methods (e.g., blue/white screening, PCR, sequencing) required.

TLDR:

Recombinant DNA is generated by isolating plasmids, cutting both vector and insert with restriction enzymes, ligating them, and transforming them into bacteria. Selection uses antibiotics; screening distinguishes recombinants from empty vectors.


3. Describe the Properties of Restriction Enzymes and Apply These Properties to Cloning Contexts

Key Features of Restriction Enzymes (REs):

Property Description
Source Naturally found in bacteria as a defense against phages
Recognition Sites Usually palindromic DNA sequences
Cutting Mechanism Hydrolyse phosphodiester bonds in DNA backbone
DNA Methylation Sensitivity Bacteria methylate their own DNA to prevent self-digestion
End Type Create sticky ends (5′/3′ overhangs) or blunt ends

Application in Cloning:

Feature Purpose in Cloning
Sticky ends Promote directional and specific ligation
Blunt ends Less efficient, more versatile but may accept any insert
Multiple Cloning Site (MCS) Contains many unique RE sites to give flexibility in cloning
Tail Addition via PCR Artificial RE sites added to primers for compatibility

Flowchart: Using Restriction Enzymes in Cloning

[Plasmid DNA] + [Restriction Enzyme] → Cut at MCS → Insert Gene with Matching Ends → Ligate → Transform

TLDR:

Restriction enzymes are sequence-specific DNA cutters originally from bacteria. They are essential in cloning for preparing plasmids and inserts. Sticky ends allow specificity; blunt ends allow flexibility. The MCS ensures safe, targeted insertion.


L32: Molecular Techniques III

Identify and Describe the Procedures Used to Screen and Validate Recombinant DNA Molecules

Recombinant screening ensures that plasmids carry the intended DNA insert, not just the vector backbone. Since not all transformed bacteria are recombinant, several validation techniques are used.


1. Blue-White Screening (Qualitative, Visual)

Principle:

  • Uses the LacZ gene (encodes β-galactosidase) with a multiple cloning site (MCS) inserted within it.
  • Insertion of foreign DNA disrupts LacZ, preventing β-galactosidase production.
  • Substrate X-gal is cleaved by active β-galactosidase → turns colonies blue.
Colony Colour Interpretation
White Recombinant plasmid (insert disrupted LacZ)
Blue Non-recombinant (LacZ intact)

Key Reagents:

  • Ampicillin: Selects transformed cells (plasmid contains resistance gene).
  • X-gal: Chromogenic substrate, cleaved to produce blue colour.
  • IPTG (if used): Induces LacZ expression.

Limitations:

  • Remnants of LacZ still present → may interfere in downstream applications.
  • Overuse of X-gal may lead to false blue or pale blue colonies (spontaneous hydrolysis).
  • Not definitive—requires further validation.

2. Colony PCR (Quick, DNA-specific)

Principle:

  • Pick colonies → add directly to PCR mix with primers flanking the insert.
  • First PCR step (95 °C) lyses cells.
  • Presence and size of product tells you whether the insert is present.
Result Interpretation
Larger band Recombinant (insert included)
Smaller/no band Non-recombinant or no insert

Advantages:

  • Fast (~1 hour total).
  • Sensitive to small size differences (~100 bp).
  • Good for screening many colonies at once.

3. Restriction Digest (Semi-quantitative, Structural Check)

Principle:

  • Extract plasmid DNA → digest with restriction enzymes flanking the insert.
  • Run fragments on gel to assess insert size.
Outcome Meaning
Two fragments (larger insert) Recombinant
Two fragments (smaller, no insert) Non-recombinant

Advantages:

  • Easy setup.
  • Confirms insert size and correct ligation site usage.
  • Still requires plasmid preparation (plasmid mini-prep).

4. Sequencing (Definitive, Gold Standard)

Principle:

  • Extract plasmid → use Sanger sequencing (or other platforms) to confirm insert sequence and orientation.
Feature Benefit
Fluorescent nucleotides Direct sequence readout
Template specific primers Targeted verification

Limitations:

  • Time-consuming and expensive.
  • Typically reserved for final validation after narrowing candidates with other methods.

Screening Summary Table:

Method Confirms Insert? Speed Cost Drawback
Blue-white screen Indirect Fast Low False positives, qualitative only
Colony PCR Yes Very fast Low Can fail if primers poorly designed
Restriction digest Yes (size only) Medium Low Cannot confirm sequence or orientation
Sequencing Yes (definitive) Slow High Costly for large-scale screening

TLDR:

Screening recombinant DNA involves a tiered validation strategy:

  • Use blue-white screening to identify likely recombinants.
  • Confirm presence of insert with colony PCR or restriction digest.
  • Validate insert identity and orientation by sequencing. This layered approach balances efficiency, accuracy, and cost.

Theory of Practical


🔬 Spectrophotometry & ELMA


1. 🔹 Generate an absorption spectrum using a spectrophotometer and interpret the results

Explanation: An absorption spectrum plots absorbance against wavelength for a substance. By running a scan (typically from 650 nm to 350 nm), you identify the λmax, the wavelength at which the compound absorbs the most light. This value is crucial because measuring absorbance at λmax provides the most sensitive and reliable data.

Procedure Highlights:

  • Use distilled water as a blank to baseline the spectrophotometer.
  • Measure the absorbance of a known dye (e.g., green, blue, red, yellow) across the spectrum.
  • Record and analyse λmax and secondary peaks.

Example Diagram: Example Absorption Spectrum

Video: Absorption Spectra Explained – YouTube (Kurzgesagt style)


2. 🔹 Select appropriate wavelengths for measuring the absorbance of an unknown compound in a mixture

Explanation: To accurately quantify a compound, measure at or near its λmax, where sensitivity is highest and interference is minimized. In mixtures, choose wavelengths where the target compound’s absorbance is significant and others’ is minimal.

Example: When mixing red and blue dyes, both may have overlapping spectra. Use unique λmax values to deconvolute contributions.

Key Point: Choose λmax based on the compound of interest, while minimizing overlapping absorbance from other substances.


3. 🔹 Apply the Beer-Lambert Law to estimate concentrations of compounds from absorbance data

Equation: \(A = \varepsilon \cdot c \cdot l\) Where:

  • \(A\) = Absorbance
  • \(\varepsilon\) = Molar extinction coefficient (M⁻¹·cm⁻¹)
  • \(c\) = Concentration (mol/L)
  • \(l\) = Path length (usually 1 cm)

Example Calculation: If \(A = 0.5\), \(\varepsilon = 5\, \text{mM}^{-1}\text{cm}^{-1}\), and \(l = 1\, \text{cm}\), then:

\[ c = \frac{A}{\varepsilon \cdot l} = \frac{0.5}{5 \cdot 1} = 0.1\, \text{mM} \]

Useful Tool: You can calculate these in Excel or lab software.


4. 🔹 Prepare standard curves in Excel with absorbance data obtained in the lab

Steps:

  1. Prepare serial dilutions (e.g., 0, 20, 40, 60, 80, 100 nmol/mL).
  2. Measure absorbance of each at λmax.
  3. Plot: x-axis = concentration, y-axis = absorbance.
  4. Add linear trendline and show equation + R².

Key Point: The linearity (R² close to 1.00) confirms reliable measurement. Use this curve to interpolate unknown concentrations.

Diagram Example: Standard Curve

Excel Guide Video: Create a Standard Curve in Excel


5. 🔹 Use standard curves and spectrophotometry to interpolate concentrations of unknown compounds

Explanation: Using the equation from your standard curve (e.g., \(A = 0.01c\)), you can solve for unknown concentration:

\[ c = \frac{A}{\text{slope of the curve}} \]

Example: If the standard curve equation is \(y = 0.0104x\) and an unknown has an absorbance of 0.52:

\[ x = \frac{0.52}{0.0104} \approx 50\, \text{nmol/mL} \]


6. 🔹 Explain why it is important to consider the working range when determining the concentration of a compound

Explanation:

  • Working range: The span of absorbance values where the spectrophotometer provides reliable, linear data (typically between 0.1 and 1.0).

  • Outside this range:

    • < 0.1: Signal-to-noise ratio too low.
    • > 1.0: Non-linearity due to excessive absorption and light scatter.

Example: If your unknown’s absorbance is 1.7, you should dilute it to bring it into the optimal range (~0.5).


7. 🔹 Apply the principles of spectrophotometry to biological contexts

Application in ELMA (Glucose Assay):

  • Measures glucose via a coupled enzyme reaction:

    • Glucose oxidase → H₂O₂
    • Peroxidase + 4-AAP + phenol → red quinoneimine (read at 500 nm)

In practice:

  • Run a glucose standard curve.
  • Use it to determine unknown blood glucose concentration.
  • Diagnostic context: helps identify Type I vs Type II diabetes.

🧪 Summary Table

Objective Key Technique Example
Generate absorption spectrum λ scan 650–350 nm Blue dye: λmax = 628 nm
Select λ Choose λmax Yellow dye = 426 nm
Beer-Lambert Law \(A = \varepsilon c l\) Use ε = 10 to find c
Standard curve Serial dilutions, Excel y = 0.0104x
Interpolate unknowns Use standard curve eq. A = 0.52 → x = 50
Working range 0.1 ≤ A ≤ 1.0 Above or below = error
Biological context ELMA glucose test Measures diabetes state

DNA Genotyping, PCR & Electrophoresis


🔹 1. Explain the steps required to isolate DNA

Goal: Extract human DNA (from cheek cells) in a form suitable for PCR.

Key Steps:

  1. Cell collection – Mouth rinse or cheek swab collects epithelial cells.
  2. Centrifugation – Pellets the cells.
  3. Lysis – Chelex + heat (100°C) disrupts membranes and denatures proteins.
  4. Chelation – Chelex chelates Mg²⁺ ions → inhibits nucleases that degrade DNA.
  5. Debris removal – High-speed centrifugation removes Chelex, ions, and cell debris.
  6. DNA supernatant collection – Only clean DNA-containing solution is retained.

Critical Notes:

  • Avoid Chelex carryover → it inhibits Taq polymerase.
  • Do not vortex DNA post-isolation; shear forces can fragment it.

🖼️ Diagram: DNA Isolation Workflow – ThermoFisher


🔹 2. Describe the function of PCR components

Component Function
Template DNA Source DNA (e.g. D1S80 locus from cheek cells)
Primers Short DNA oligos that define the amplified region
Taq polymerase Heat-stable enzyme that synthesises new DNA
dNTPs Free nucleotides used to build new DNA strands
Buffer (Tris, KCl) Maintains pH and salt conditions for enzyme activity
MgCl₂ Essential cofactor for DNA polymerase
Water To bring the final volume to 50 µL

🧪 PCR Master Mix includes everything except primers and template.


🔹 3. Predict the effects of adding too much, too little, or omitting PCR components

Component Too Much Too Little Omitted
Template DNA Inhibition due to impurities or competition Weak product No amplification
Primers Non-specific binding, primer-dimer formation Incomplete reactions No amplification
Taq polymerase Non-specific products, background bands Inefficient amplification No amplification
Mg²⁺ Increased error rate, non-specific bands Reduced yield No activity
dNTPs Incorporation errors Incomplete synthesis No DNA synthesis
Buffer pH imbalance Poor enzyme function No amplification

🔹 4. Estimate nucleic acid purity using the absorbance at 260 nm and 280 nm

Principle:

  • A260: Nucleic acids absorb maximally at 260 nm.
  • A280: Proteins (especially tryptophan) absorb at 280 nm.

A260/A280 Ratio:

  • ~1.8 → pure DNA
  • ~2.0 → pure RNA
  • <1.6 → protein or phenol contamination likely

🖼️ Nanodrop Spectrum Example (p. 20–21)


🔹 5. Identify likely contaminants in a DNA preparation (such as protein) on a UV spectrum

Contaminants and Their UV Absorbance Patterns:

Contaminant Absorbance Signature
Protein Elevated 280 nm peak → low A260/A280
Phenol High 270 nm absorbance, distorts spectrum
Chelex Does not show UV absorbance but can inhibit PCR if carried over

🔹 6. Evaluate whether the contaminants will impact results of a PCR

Contaminant Impact on PCR
Protein Binds DNA or enzyme, reducing yield
Phenol or SDS Denatures Taq polymerase
Chelex Chelates Mg²⁺, blocking DNA polymerase
Ethanol/salts Interfere with enzyme activity and denature DNA

Conclusion: Even small amounts of contaminants can fully inhibit PCR. Always remove Chelex and centrifuge thoroughly to avoid inhibitory carryover.


🔹 7. Compare the information we obtain from spectrophotometric (Nanodrop) and electrophoretic methods and reasons for combining the two

Method What it tells you Limitations
Nanodrop (spectrophotometry) DNA concentration, purity (A260/A280) Doesn’t show DNA degradation or specific fragment size
Agarose gel electrophoresis Fragment size, integrity, presence of bands Doesn’t give precise concentration or purity

🔁 Why use both?

  • Nanodrop confirms quantity and purity.
  • Gel confirms presence, size, and quality of PCR product.

Example:

  • Nanodrop says you have DNA, but no band = likely degraded or inhibitory contaminants.
  • Clear band at expected size = successful PCR, fragment ~300–700 bp = D1S80 alleles.

🖼️ Gel electrophoresis image – example of VNTR PCR


🧪 Summary Table of Objectives

Objective Tool/Technique Outcome
DNA isolation Cheek cells, Chelex PCR-compatible DNA
PCR components Master mix + DNA + primers Amplification of D1S80
Troubleshooting Vary each component Understand function
Nanodrop A260/A280 ratio Purity + concentration
UV spectrum Identify contaminants Decide if DNA is usable
Gel electrophoresis Separation by size Visual confirmation
Combined analysis Spectro + Gel Reliable, interpretable genotyping

PCR Electrophoresis & Plasmid Mapping


🔹 1. Estimate the size of PCR amplicons

Method:

  • Use agarose gel electrophoresis with a DNA ladder (size standard).
  • Measure the migration distance (mm) of your PCR band and compare it to known fragment sizes.

Key Concept:

  • DNA migrates inversely with the log10 of its size. A standard curve of log10(size) vs distance allows interpolation.

🧰 Excel Tip: Plot log10(bp) (y-axis) vs migration (mm) (x-axis) → trendline equation gives a tool to calculate unknown sizes.

📊 Example Tool: Bioline DNA HyperLadder IV: https://www.bioline.com/sg/hyperladder-100bp.html


🔹 2. Determine the number of copies in a VNTR using the size of a PCR product

D1S80 locus structure:

  • Flanking (non-repetitive) region: ~164 bp (forward primer to start of repeat) + ~160 bp (after last repeat)
  • Each repeat unit: 16 bp
  • Total length = 324 bp + (16 × number of repeats)

Formula:

\[ \text{Number of repeats} = \frac{\text{Amplicon size} - 324}{16} \]

Example: If amplicon size is 624 bp →

\[ \frac{624 - 324}{16} = 18 \text{ repeats} \]


🔹 3. Appreciate the uses of DNA genotyping

Applications:

  • Forensic identification (e.g. matching VNTR profiles in crime scenes)
  • Paternity testing (VNTR inheritance)
  • Disease gene tracking (e.g. Huntington’s trinucleotide repeats)
  • Population genetics and ancestry testing
  • Identity confirmation in tissue/organ transplantation

🎥 Video: What is DNA profiling?


🔹 4. Explain how positive and no-template controls help troubleshoot PCR

Control What it contains Expected result Purpose
Positive control Working PCR mix + known DNA Visible band Confirms PCR reagents and thermocycler work
No-template control (NTC) PCR mix with water instead of DNA No bands Confirms no contamination or primer-dimers

Common errors if controls fail:

  • NTC shows a band → contamination
  • Positive control fails → enzyme or thermocycler problem

🔹 5. Identify homozygous and heterozygous loci using PCR and electrophoresis

Principle:

  • Humans inherit one D1S80 allele from each parent.

  • Gel with:

    • One band → homozygous
    • Two bands → heterozygous

🔹 6. Appreciate inheritance of alleles and haplotypes

Key Concept:

  • Alleles at a locus are inherited one from each parent.
  • A haplotype is a group of alleles (or genes) inherited together from a single parent.

Example:

  • Mother: 18 + 20 repeats
  • Father: 22 + 24 repeats → Child: could inherit any one from each → e.g. 18 + 22

🧬 Video: DNA inheritance explained


🔹 7. Interpret plasmid maps and identify restriction endonuclease cleavage sites

Plasmid Map Basics:

  • Circular DNA diagram with gene and restriction site locations
  • Restriction enzymes (e.g. EcoRI, PstI) cut at specific sequences

🧭 Use SnapGene or NEB Cutter to:

  • View cut sites
  • Predict fragment sizes

📍 Example:

  • EcoRI at 1000 bp and 6000 bp → produces fragments of 5000 bp and 1000 bp

🧬 Video: Interpreting plasmid maps


🔹 8. Analyse and interpret restriction fragment gel electrophoresis

Steps:

  1. Digest plasmid with one or more enzymes
  2. Run fragments on a gel
  3. Compare band sizes to known ladder

Double digest → more precise location of cut sites

Interpretation:

  • If digest gives two bands at 5000 bp and 1000 bp → infer distance between two cut sites is 1000 bp, rest of plasmid is 5000 bp

🔹 9. Identify an unknown plasmid by restriction mapping

Process:

  1. Perform single and double digests (e.g., EcoRI, PstI, EcoRI + PstI)
  2. Measure fragment sizes from gel
  3. Match pattern to predicted outcomes for Plasmid A, B, or C

🧩 Matching tip: Use your pre-drawn restriction maps from prework!


🔹 10. Predict the sizes of DNA fragments after restriction enzyme digestion

Example:

  • Plasmid size = 7000 bp
  • EcoRI cuts at 1500 bp and 6000 bp → Fragments = 1500 bp, 4500 bp, 1000 bp

Considerations:

  • Circular vs linear DNA
  • Cut site locations

🔹 11. Predict the results of a gel electrophoresis experiment

Key Predictions:

  • Smaller fragments → run further
  • Supercoiled plasmid → runs faster than linear
  • Circular uncut plasmid → runs erratically (often multiple bands)

Band Pattern Example:

  • Lane 1: Ladder
  • Lane 2: Uncut plasmid (supercoiled)
  • Lane 3: EcoRI digest → 1 band
  • Lane 4: EcoRI + PstI → 2–3 bands (depending on sites)

🎥 Video: Gel electrophoresis interpretation


Molecular and Solution Calculations


🔹 1. Understand the relationship between amounts given in weights and moles and the concentration of a solution


Key Concepts

  • Mass (g), moles (mol), and concentration (mol/L or M) are related through the volume of the solution and the molecular weight (MW) of the solute.

\[ \text{Moles (mol)} = \frac{\text{Mass (g)}}{\text{Molecular Weight (g/mol)}} \]

\[ \text{Concentration (mol/L)} = \frac{\text{Moles (mol)}}{\text{Volume (L)}} \]


📘 Example 1:

How many moles are in 180 mg of glucose (MW = 180.16 g/mol)?

Convert mg to g:

\[ 180\, \text{mg} = 0.180\, \text{g} \]

\[ \text{Moles} = \frac{0.180}{180.16} \approx 0.001\, \text{mol} = 1\, \text{mmol} \]


📘 Example 2:

What is the concentration if you dissolve 0.5 g of NaCl (MW = 58.44 g/mol) in 250 mL of water?

\[ \text{Moles} = \frac{0.5}{58.44} \approx 0.00855\, \text{mol} \]

\[ \text{Concentration} = \frac{0.00855}{0.250} = 0.0342\, \text{mol/L} = 34.2\, \text{mM} \]


🔹 2. Use molecular weight to convert between moles and mass of a compound in a given solution


Key Formula

\[ \text{Mass (g)} = \text{Moles} \times \text{Molecular Weight (g/mol)} \]


📘 Example 3:

How much Tris base (MW = 121.14 g/mol) do you need to make 100 mL of a 0.5 M solution?

\[ \text{Moles} = 0.5\, \text{mol/L} \times 0.1\, \text{L} = 0.05\, \text{mol} \]

\[ \text{Mass} = 0.05 \times 121.14 = 6.06\, \text{g} \]


📘 Example 4:

How many grams of EDTA (MW = 292.24 g/mol) are required for 500 mL of 20 mM solution?

\[ \text{Moles} = 0.020\, \text{mol/L} \times 0.5\, \text{L} = 0.01\, \text{mol} \]

\[ \text{Mass} = 0.01 \times 292.24 = 2.9224\, \text{g} \]


🔹 3. Calculate final concentrations from stock solutions and volumes


Key Formula: Dilution Equation

\[ C_1 V_1 = C_2 V_2 \]

Where:

  • \(C_1\) = stock concentration
  • \(V_1\) = volume of stock solution to use
  • \(C_2\) = desired (final) concentration
  • \(V_2\) = final total volume

📘 Example 5:

You have 10X TBE buffer and need 500 mL of 1X buffer.

\[ 10 \times V_1 = 1 \times 500 \Rightarrow V_1 = \frac{500}{10} = 50\, \text{mL} \]

Mix 50 mL of 10X TBE with 450 mL water to make 500 mL of 1X.


📘 Example 6:

You have a 1 M NaOH stock. How much is needed to make 200 mL of 0.1 M NaOH?

\[ 1 \times V_1 = 0.1 \times 200 \Rightarrow V_1 = \frac{20}{1} = 20\, \text{mL} \]

Use 20 mL of 1 M NaOH, top up with 180 mL water to get 200 mL of 0.1 M.


🔍 Key Unit Conversions

Unit Equivalent
1 L 1000 mL
1 mM 0.001 M
1 µL 0.001 mL
1 g 1000 mg

🧪 Summary Table

Concept Formula Key Units
Mass ↔︎ Moles \(\text{mass} = \text{mol} \times \text{MW}\) g, mol, g/mol
Moles ↔︎ Concentration \(\text{C} = \frac{\text{mol}}{\text{L}}\) mol/L
Dilution \(C_1 V_1 = C_2 V_2\) M, mL or L