Biochemical Basis of Therapeutics Flashcards

1
Q

What are the main subdivisions of the nervous system?

A

Central Nervous System (CNS):
* Comprised of the brain, spinal cord and optic nerve
* Processes sensory information and generates responses

Peripheral Nervous System (PNS):
* Somatic Nervous System - controls voluntary muscle movements
* Autonomic Nervous System (ANS) - regulates involuntary functions

Autonomic Nervous System (ANS):
* Sympathetic Nervous System (SNS) - “Fight-or-flight” response
* Parasympathetic Nervous System (PNS) - “Rest-and-digest” functions
* Enteric Nervous System (ENS) - regulates the gastrointestinal tract

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What structures make up the CNS?

A

brain
spinal cord
optic nerve

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the subdivisions of the PNS?

A

Somatic - controls voluntary muscle movements

Autonomic - regulates involuntary functions:

  • Sympathetic - Fight-or-flight” response
  • Parasympathetic - “Rest-and-digest” functions
  • Enteric - regulates the gastrointestinal tract
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What types of cells is the CNS comprised of?

A

Ependymal cells

Oligodendrocytes

Microglia

Astrocytes

Satellite Cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the function of ependymal cells?

A

Make up the lining of the ventricular system (a series of CSF-filled ventricle cavities throughout the brain and spinal cord)

Produce and regulate the secretion of cerebrospinal fluid (CSF)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the function of oligodendrocytes?

A

Form myelin insulation of axons (only in CNS)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is the function of microglia?

A

Macrophage-like cells

Important for immune defence and tissue repair in the brain and spinal cord

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the function of astrocytes?

A

Regulate and maintain chemical composition of the extracellular space

Maintain the BBB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is the function of satellite cells?

A

Small cells surrounding neurons in the sensory, sympathetic/parasympathetic ganglia

Regulate the external chemical environment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What protection features does the CNS have?

A

Enclosed by hard, bony structures
* The Brain is protected by the Cranium (skull)
* The Spinal Cord is protected by vertebral columns

Meninges - protective and nourishing membranes

Cerebrospinal fluid (CSF) - protective cushioning fluid surrounding the brain

Blood Brain Barrier (BBB)
* Highly selective
* Limits the access of bloodborne materials (including drugs) into the vulnerable brain tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How is the BBB structured?

A

The blood vessels in the brain are joined by tight junctions and supported by the feet of the astrocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What cells produce cerebrospinal fluid (CSF)?

A

Ependymal cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Name the three meninges

A

Dura mater

Arachnoid mater

Pia mater

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the excitatory neurotransmitters in the CNS?

A

Glutamate

Acetylcholine (Ach)

Serotonin (5-HT)

Dopamine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the inhibitory neurotransmitters in the CNS?

A

GABA

Serotonin (5-HT)

Dopamine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is the length of preganglionic neurons in the Sympathetic NS?

A

Short

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the length of preganglionic neurons in the Parasympathetic NS?

A

Long

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the length of postganglionic neurons in the Sympathetic NS?

A

Long

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the length of postganglionic neurons in the Parasympathetic NS?

A

Short

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the preganglionic neurotransmitter in the Sympathetic NS?

A

Acetylcholine (Ach)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the preganglionic neurotransmitter in the Parasympathetic NS?

A

Acetylcholine (Ach)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the postganglionic neurotransmitter in the Sympathetic NS?

A

Noradrenaline (NA)

or

Norepinephrine (NE)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the postganglionic neurotransmitter in the Parasympathetic NS?

A

Acetylcholine (Ach)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What type of receptors do the preganglionic neurotransmitters act on in the SNS?

A

Nicotinic cholinergic receptors (ligand-gated ion channels)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
What type of receptors do the preganglionic neurotransmitters act on in the Parasympathetic NS?
Nicotinic cholinergic receptors (ligand-gated ion channels)
26
What type of receptors do the postganglionic neurotransmitters act on in the Sympathetic NS?
Adrenergic receptors (αlpha/beta) G protein-coupled receptors (GPCRs)
27
What type of receptors do the postganglionic neurotransmitters act on in the Parasympathetic NS?
Muscarinic cholinergic receptors (M1-M5) G protein-coupled receptors (GPCRs)
28
What type of neurotransmitter is used in the Somatic NS?
Acetylcholine (Ach)
29
What type of receptor is present in the Somatic NS?
Nicotinic cholinergic receptors (ligand-gated ion channels)
30
Why is targeting sympathetic nerves in the CNS not ideal for drug intervention?
Drugs will need to penetrate the BBB which is highly selective Effects are likeley to be widespread
31
Why is targeting sympathetic ganglion not ideal for drug intervention?
Non-selective / non-specific effects Widespread effects - side effects
32
Why is targeting the neuroeffector junction ideal for drug intervention?
Allows for specific targeting of function Minimal off-target effects
33
What forms the binding site for Ach in nicotinic Ach receptors?
2 alpha subunits
34
What type of receptors are nicotinic Ach receptors?
ligand-gated non-selective cation channel
35
What are the functions/effects of sympathetic innervation?
Pupil dilation Vasoconstriction Decreased salivation Increased heart rate Increased force of heart contraction Bronchodilation Reduced motility in GI system Increased metabolism in liver (gluconeogenesis) Bladder relaxation Ejaculation
36
What are the different ways drugs can target sympathetic signalling?
Neurotransmitter synthesis (e.g. α-methyl-tyrosine) Neurotransmitter storage (e.g. reserpine, amphetamine) Autoreceptors (e.g. clonidine) Termination of signalling (e.g. moclobemide) Post synaptic receptors * Adrenergic receptor antagonists - e.g. doxazosin, propranolol, atenolol * Adrenergic receptor agonists - e.g. salbutamol
37
Give an example of how neurotransmitter storage can be used as a drug target.
Monoamine neurotransmitters (NA, Dopamine, Serotonin) are metabolised by monoamine oxidase (MAO). So to protect them, they are taken into vesicles via the vesicular monoamine transporter (VMAT). Reserpine is a drug that blocks the VMAT, preventing NA uptake into vesicles leading to its metabolisation by MAO. This decreases the nerve of NA.
38
Give an example of how neurotransmitter storage can be used as a drug target.
Monoamine neurotransmitters (NA, Dopamine, Serotonin) are metabolised by monoamine oxidase (MAO). So to protect them, they are taken into vesicles via the vesicular monoamine transporter (VMAT). Drugs such as Amphetamine, Tyramine and Ephedrine enter via the ‘uptake 1’ channel and compete with NA to enter the vesicles via VMAT displacing NA from storage causing leakage into the synaptic gap + increasing the concentration of free NA for release into the synaptic gap.
39
Give an example of how autoreceptors can be a drug target for sympathetic signalling.
Autoreceptors inhibit neurotransmitter release via exocytosis in order to regulate neurotransmission Clonidine stimulates these autoreceptors (α2AR) which inhibits neurotransmitter release.
40
Give an example of drug which targets termination of sympathetic signalling.
Signalling is terminated by the uptake of neurotransmitters followed by break down by Monoamine Oxidase (MAO) (and catechol-0-methyltransferase (COMT) for dopamine) into inactive metabolites MAO inhibitors (e.g. Moclobemide) can be utilised to prevent signal termination and increase/maintain neurotransmitter levels
41
Why can't patients on MAO inhibitors (e.g. Moclobemide) eat dairy products such as cheese and yoghurt or drink wine?
Wine and dairy products such as cheese and yoghurt contain tyrosine. Tyrosine is broken down by MAO, so MAO inhibitors prevents breakdown of tyrosine and leads to an accumulation and hypertensive crisis's as a result
42
Why is the most suitable target area for pharmacological intervention at the postsynaptic receptors?
Targeting upstream processes like neurotransmitter synthesis or degradation can have widespread effects, whilst postsynaptic receptor modulation tends to be more localised, reducing unintended systemic effects Specificity of action
43
What are the functions/effects of parasympathetic innervation?
Pupil constriction Tears Decreased heart rate Vasoconstriction Increased GI motility Bladder contraction Erection
44
What are the different ways drugs can target parasympathetic signalling?
Neurotransmitter synthesis (e.g. hemicholinium) Neurotransmitter storage (e.g. inhibitors of VAChT) Neurotransmitter release (e.g. Botulinum toxin - i.e. botox) Termination of signalling (acetylcholinesterase (AChE) inhibitors - e.g. Neostigmine, Edrophonium) Ganglia receptors (e.g. hexamethonium) Postsynaptic (muscarinic) receptors * Muscarinic receptor antagonists (anti-cholinergics) - e.g. atropine, darifenacin * Muscarinic receptor agonists
45
Describe how the SNS and PNS work together to maintain blood pressure.
Increased/high BP distends arterial wall Afferent nerve endings are stimulated by resultant stretch Sends signal to the brain (NTS) , which compares the BP signal to a set point. Brain coordinates: * Increased parasympathetic drive/innervation to heart * This leads to a decreased heart rate and decreased cardiac output (CO) + * Decreased sympathetic drive/innervation to heart, arteries, veins * This leads to a decreased heart rate and decreased force of contraction → therefore decreased cardiac output (CO) * This leads to decreased arterial constriction and venous constriction This causes BP to fall to the set point If BP falls, opposite effects occur to bring it back up to the set point
46
What are the three classes of G Protein Coupled Receptors?
Gs s = stimulate Gi i = inhibit Gq
47
Give examples of Gs receptors.
NA Receptors – β1, β2, β3 Dopamine Receptors – D1, D5 5-HT Receptors – 5-HT4, 5-HT6, 5-HT7 Histamine Receptors – H2 V2
48
Describe the cell signalling pathway of Gs receptors.
1. Binding of agonist to receptor causes conformational change 2. GDP exchanged for GTP bound to G Protein 3. Hydrolysis of GTP provides energy for dissociation of α-subunit, from β/ɣ subunits 4. α-subunit activates Adenylyl Cyclase which results in the formation of cAMP 5. cAMP activates Protein Kinase A which phosphorylates downstream target proteins Summary: ↑ Adenylyl cyclase → ↑ cAMP → activates PKA
49
Give examples of Gi receptors.
Ach Receptors – M2, M4 NA Receptors – α2 Dopamine Receptors – D2, D3, D4 5-HT Receptors – 5-HT1, 5-HT5 GABA Receptors – GABAb
50
Describe the cell signalling pathway of Gi receptors.
1. Binding of agonist to receptor causes conformational change 2. GDP exchanged for GTP bound to G Protein 3. Hydrolysis of GTP provides energy for dissociation of α-subunit, from β/ɣ subunits 4. α-subunit inhibits Adenylyl Cyclase which prevents the formation of cAMP 5. Therefore decreased Protein Kinase A activity and decreased phosphorylation of downstream target proteins 6. Also activation of potassium channels + inhibition of calcium channels Summary: ↓ cAMP → ↓ PKA, opens K⁺ channels, closes Ca²⁺ channels
51
Give examples of Gq receptors.
Ach Receptors – M1, M3, M5 NA Receptors – α1 5-HT Receptors – 5-HT2 H1 V1
52
Describe the cell signalling pathway of Gq receptors.
1. Binding of agonist to receptor causes conformational change 2. GDP exchanged for GTP bound to G Protein 3. Hydrolysis of GTP provides energy for dissociation of α- subunit, from β/ɣ subunits 4. α-subunit activates PLC which results in the formation of IP3 and DAG from PIP2. 5. IP3 mobilises Ca2+ from intracellular stores and DAG stimulates PKC ↑ intracellular Ca2+ activates Ca2+ dependent response including muscle contraction PKC phosphorylates downstream target proteins – e.g. muscle contraction Summary: ↑ PLC → ↑ IP3 + DAG → ↑ Ca²⁺ + activates PKC
53
What are the features of a typical nerve cell/ neuron?
Cell Body (Soma) Dendrites Axon Myelin Sheath Nodes of Ranvier Axon Terminals Synapse
54
What is the function of the soma (cell body) in a nerve cell?
Contains the nucleus and organelles essential for cellular function and metabolism Integrates the information received by the dendrites
55
What is the function of the dendrites in a nerve cell?
Branch-like structures that receive signals from other neurons and convey them to the cell body
56
What is the function of the axon in a nerve cell?
A long projection that transmits electrical impulses (action potentials) away from the cell body toward other neurons or muscles May or may not be insulated by myelin The axon hillock is where the action potential is initiated
57
What is the function of the Myelin Sheath in a nerve cell?
A fatty layer (formed by oligodendrocytes in the CNS and Schwann cells in the PNS) that insulates the axon and speeds up signal transmission
58
What is the function of Nodes of Ranvier in a nerve cell?
Gaps between segments of the myelin sheath where ion exchange occurs, allowing for rapid signal conduction (saltatory conduction)
59
What is the function of the axon terminals in a nerve cell?
Endings of the axon that release neurotransmitters into the synaptic cleft to communicate with target cells
60
What is the typical resting membrane potential?
~ 70 mV
61
What is resting potential?
Approximately -55 to -70mv During this state there are: * More sodium ions (Na⁺) outside than inside the neurone * More potassium ions (K⁺) inside than outside the neurone
62
How is resting potential maintained?
The neuron cell membrane is super permeable to potassium ions, and so lots of potassium leaks out of the neuron through potassium leakage channels The neuron cell membrane is partially permeable to sodium ions, so sodium atoms slowly leak into the neuron through sodium leakage channels The cell wants to maintain a negative resting membrane potential, so pumps potassium back into the cell and pumps sodium out of the cell at the same time Sumary: * Sodium-potassium pump actively transports 3 Na⁺ ions out and 2 K⁺ ions into the neuron * The membrane is more permeable to K⁺ than Na⁺, allowing K⁺ to leak out
63
Sequence of action potential phases
Resting → Depolarisation → Repolarisation → Hyperpolarisation → Resting
64
How is an action potential generated?
1. The neuron is at rest, maintaining a stable negative charge inside (~ -70mV) 2. A stimulus causes the release of neurotransmitters which bind to receptors on the dendrites **EPSPs** 3. This causes the opening of voltage-gated Na⁺ channels so that there is an influx of Na⁺ into the neurone. As a result the cell depolarises and produces an Excitatory Post-Synaptic Potential (EPSP) **Rapid depolarisation** 4. These EPSPs sum up and when the AP threshold (-55 mV) is reached more voltage-gated Na⁺ channels open, causing a large influx of Na⁺ into the neurone to become highly positive (~ +30mV) **Repolarisation** 5. Voltage-gated Na⁺ channels close, stopping Na⁺ entry and voltage-gated K⁺ channels open, allowing K⁺ to leave the neuron. This means the cell loses positively charged ions so the membrane potential becomes more negative, returning back toward resting state **Hyperpolarisation** 6. As the action potential passes through, the K⁺ channels remain open causing the membrane potential to drop below resting potential (~ -80mV). This makes the neuron temporarily less likely to fire another action potential. 7. The K⁺ channels close and the Na⁺/K⁺ pump restores the original ion balance to reestablish the resting state.
65
What are the two types of synaptic transmission?
Chemical (the most common form) Electrical
66
What triggers neurotransmitter release from the presynaptic terminal?
Influx of Ca²⁺ via voltage-gated channels
67
What is the difference between EPSPs and IPSPs?
EPSP = depolarisation, increases likelihood of action potential being fired IPSP = hyperpolarisation, decreases likelihood of action potential being fired
68
Describe the process of chemical synaptic transmission
1. A nerve impulse/action potential arrives at the presynaptic terminal, causing depolarisation 2. This depolarisation of the membrane causes voltage-gated Ca²⁺ channels to open, allowing Ca²⁺ influx into the presynaptic terminal 3. The increased Ca²⁺ concentration triggers synaptic vesicles to fuse with the presynaptic membrane and release neurotransmitters into the synaptic cleft via exocytosis 4. The released neurotransmitters diffuse across the synaptic cleft and bind to specific receptors on the postsynaptic neuron
69
What are the two main types of receptors at the synapse?
Ionotropic Metabotropic
70
What are ionotropic receptors?
Ligand-gated ion channels that mediate fast responses Agonist activation induces a conformational change, opening the ion pore
71
Give examples of ionotropic receptors
Nicotinic ACh receptors (non selective cation channel) NMDA Glutamate receptors (non selective cation channel) AMPA Glutamate receptors (non selective cation channel) 5HT3 receptors (non selective cation channel) GABAa receptors (selective chloride channel)
72
What are metabotropic receptors?
GPCRs that initiate slower, longer-lasting effects via secondary messengers Gives more selectivity of function over ionotropic receptors which are selective only to their ion Different metabotropic receptors can couple to different G proteins → different receptors can stimulate different signalling cascades → different functional outcomes
73
Give examples of metabotropic receptors.
Muscarinic ACh receptors All 5HT receptors except for 5HT3 GABAb receptors Dopamine receptors
74
How are neurotransmitters removed from the synaptic cleft?
Simple diffusion away from the synapse Reuptake Enzymatic degradation
75
Which enzyme breaks down ACh?
Acetylcholinesterase (AChE)
76
What transporter reuptakes serotonin?
SERT
77
What transporter reuptakes noradrenaline/norepinephrine?
NET transporter
78
How do amphetamines affect noradrenaline?
Displace NA from vesicles → increases release + inhibit MAO
79
What is the main neurotransmitter of the sympathetic nervous system?
Noradrenaline
80
What is the main neurotransmitter of the parasympathetic nervous system?
Acetylcholine
81
What are alpha-2 adrenergic receptors involved in?
Autoreceptor-mediated negative feedback
82
What is the absolute refractory period?
The period immediately after an action potential during which no new action potential can be initiated, regardless of stimulus strength, because voltage-gated Na⁺ channels are inactivated
83
What is the relative refractory period?
The period following the absolute refractory period during which a very strong stimulus is required to initiate an action potential, as some Na⁺ channels are reset, but K⁺ efflux is still occurring, making the membrane more negative than resting
84
What is the consensus sequence?
DNA sequence with the most common nucleotide in each position Derived from the Human Genome Project No one has the consensus sequence Comparisons are always made against the consensus sequence
85
How many genes do we have?
Approximately 25,000 genes
86
What are alleles?
Different versions of a gene that arise due to mutations in the DNA sequence
87
What are wild-type genes?
The normal, non-mutated form of a gene as it commonly occurs in a natural population
88
What is a SNP (Single Nucleotide Polymorphism)?
A type of mutation where one nucleotide is changed
89
What can cause SNPs?
Genetic mutation Ionising radiation emitted from the sun Radon leaks from the ground Background radiation
90
What are the different types of mutation?
Synonymous Non-synonymous Frame shift
91
What is a synonymous mutation?
A mutation that does not change the amino acid and typically does not affect protein function Normally occur in the the third base of a triple codon
92
What is a non-synonymous mutation?
A mutation that changes the amino acid sequence. Typically occur at the first and second base of a triple codon Can result in no functional amino acid change (E.g. Glu to Asp mutation - no/very little change in protein function) Can result in small changes in protein function (Eg. Arg114Cys in Cytochrome P450 2C9) Can result in a major change in protein function (Eg. Glu6Val in haemaglobin S)
93
What is a frameshift mutation?
A mutation from insertion/deletion that shifts the reading frame and usually inactivates the protein.
94
What is gene duplication?
Copying of the same allele multiple times, potentially leading to too much protein (e.g. CYP2D6 UM)
95
What is gene deletion?
The complete loss of a gene, resulting in no protein production (e.g. CYP2D6*5)
96
What is gene truncation?
Loss of a portion of a gene, producing a shortened, inactive protein
97
What is genotype?
The genetic makeup of an organism Refers to the specific DNA sequence or alleles an individual carries (e.g. AA, Aa, aa) Inherited from parents
98
What is phenotype?
The observable traits or characteristics of an organism, resulting from genotype and environmental interaction
99
What is the effect of being homozygous wild-type?
Two normal alleles → normal protein function
100
What is the effect of being heterozygous?
One normal and one mutated allele → reduced or partial function E.g. individuals with heterozygous genotype CYP2C9 *1/*3 may have an intermediate ability to metabolise drugs, leading to a moderate risk of drug toxicity or ineffective drug response (IM)
101
What is the effect of being homozygous mutant?
Two mutated alleles → significantly altered or lost protein function E.g. * Individuals with mutated genotype CYP2C9 *3/*3 may have a reduced ability to metabolise certain drugs, leading to toxicity (PM) * If the mutation results in an overactive protein, drug metabolism may be faster, which could lead to suboptimal drug effects (UM)
102
What are haplotypes?
A group of genes or DNA variations (like SNPs) that are inherited together from a single parent Usually found on the same chromosome segment
103
Give examples of alleles that are ethnically linked.
People of African descent have a higher prevalence of the CYP2D6*17 allele, which is associated with increased metabolism of some drugs (UM) East Asian populations have a higher frequency of the CYP2C19*2 allele, which may lead to slower drug metabolism (PM)
104
Why are some alleles ethnically linked?
Due to evolution, genetic drift, founder effects and limited gene flow across populations
105
How can gene mutations affect pharmacokinetics?
Mutations in genes that influence enzymes responsible for drug metabolism (e.g. CYP450 family) determine how quickly or slowly a drug is broken down in the body Too little activity causes too high a plasma concentration and gives rise to toxicity or result in lack of activity for prodrugs (E.g. codeine and cytochrome P450 2D6*5) Too much activity can cause treatment failure or cause toxicity if it a prodrug (E.g. codeine & cytochrome P450 2D6*2 UM)
106
How can gene mutations affect pharmacodynamics?
Mutations in genes can alter drug targets such as receptors, enzymes, or transporters, which will influence the drug's effects/efficacy: * Drug may not bind sufficiently to elicit response (agonist) * Drug may not bind sufficiently to inhibit response (antagonist) Mutations in uptake transporters can influence drug concentration in the body Mutations can cause some receptors to not work at optimal level
107
How do mutations in various genes interact to produce a patient-specific response?
Mutations can affect: * Metabolism * Uptake * Binding * Reuptake Metabolism (e.g. mutations in CYP450s) : * Ultra-rapid Metabolisers (UMs) = break down drugs too fast → low drug levels → reduced efficacy * Extensive Metabolisers (EMs) = normal drug metabolism, ideal drug levels * Intermediate Metabolisers (IMs) = slower drug metabolism → potential for drug accumulation and toxicity * Poor Metabolisers (PMs) = very slow metabolism → high drug levels → increased toxicity risk Mutations in transporters (e.g. p glycoprotein) could lead to accumulation of drug due to reduced efflux function Efficacy of drug is determined by genotype of receptors present (e.g. drug may not bind sufficiently)
108
What are the four different types of metabolisers?
Ultra-rapid Metabolisers (UMs) = break down drugs too fast → low drug levels → reduced efficacy Extensive Metabolisers (EMs) = normal drug metabolism, ideal drug levels Intermediate Metabolisers (IMs) = slower drug metabolism → potential for drug accumulation and toxicity Poor Metabolisers (PMs) = very slow metabolism → high drug levels → increased toxicity risk
109
Why might ethnicity be used as a clue in clinical settings?
Genetic screening is expensive, requires specialist equipment + expertise and is not practical in a clinical environments so using ethnic patterns can help predict likely genotypes
110
How can we overcome the effects of pharmacogenetic variation on drug efficacy and toxicity?
Alternative drugs Add on drugs Adjust dosing Therapeutic drug monitoring (TDM)
111
Define 'anterior'
Towards the front of the body
112
Define 'posterior'
Towards the back of the body
113
Define 'superior'
Towards the top of the body
114
Define 'inferior'
Towards the bottom of the body
115
Define 'medial'
Towards the midline of the body
116
Define 'median'
In the midline rather than towards the midline
117
Define 'lateral'
Away from the midline
118
Define 'proximal'
Towards the centre of the body
119
Define 'distal'
Away from the centre of the body
120
Define 'superficial'
Towards the surface of the body
121
Define 'deep'
Away from the surface of the body
122
What are the components of the integumentary system?
Hair Skin Nails Sweat + oil glands
123
What is the function of the integumentary system?
Protects deeper tissues from damage Regulation of body temperature Sensation – touch, pain, pressure, temperature Excretion of urea and uric acid (via sweat) Synthesis of Vitamin D (from cholesterol in the presence of sunlight) Assist in manipulation and grasping of small objects Scratching
124
What are the different forms of protection the integumentary system provides?
Mechanical – physical barrier Chemical – impermeable keratinized cells Bacterial – unbroken surface, antibacterial secretions UV – melanin pigment Thermal – heat, cold & pain receptors Dessication – waterproof keratinized cells
125
What is the function of the skeletal system?
Structure and movement of the body Attachment points for muscle Protects internal organs Produces blood cells Stores minerals
126
What are the different classes of bones?
Long bone – act as levers Flat bone – protect delicate organs Short bones – confers strength and flexibility Irregular bone – anchorage points
127
What are joints?
Areas where bones meet Allow movement Lubricated by synovial fluid
128
What are the three classes of muscle? Where are they found and their function?
Skeletal muscle * Moves the skeleton * Production of body heat * Facial muscles – expression, speech, mastication * Thorax and diaphragm – respiration * Abdomen – protect internal organs, contract to assist blood flow, defecation, vomiting and child birth Cardiac muscle * Found in the heart * Pumps blood throughout the body Smooth muscle * Found in walls of internal organs (e.g. stomach, intestines, blood vessels) * Helps move substances through the body (digestion, blood flow)
129
What are the three classes of levers in the muscular-skeletal system?
Class I lever: * Fulcrum (i.e. the pivot point) in the middle - like a seesaw * Weight balanced over fulcrum, therefore only a small amount of force needed * Limited in how much weight and how high can be lifted * E.g. neck joint Class II lever: * Weight is located between fulcrum and force * Can lift a considerable amount of weight * Limited in height * E.g. standing on tiptoes Class III lever: * Most common type of lever * The force is located between the weight and the fulcrum * Does not allow a great weight to be lifted * Can be lifted a great distance * Weight can be increase * E.g. bicep curl
130
What are the components of the respiratory system and their functions?
Nose + nasal cavity + sinuses: * filters, warms and moistens incoming air * produce mucus * act as resonators for sound * lighten skull Pharynx (throat) - passageway for air and food Larynx (voice box) - produces sound + routes air into trachea Trachea (windpipe): * carries air to bronchi * lined with cilia and mucus to trap dust Bronchi + bronchioles - direct air to alveoli Alveoli - air sacks where gas exchange occurs (O₂ enters blood, CO₂ leaves) Lungs Diaphragm: * dome-shaped muscle below lungs * contracts and relaxes to control breathing * enlarges thoracic cavity for inspiration
131
What is the function of the Cerebrum?
Higher brain functioning (e.g. thought, memories, personality, intelligence, language, vision)
132
What is the function of the Cerebellum?
Fine co-ordination of movements (e.g. walking)
133
What is the function of the brainstem?
Control of functions essential to life which we have no conscious control over Comprised of: The midbrain - centre for several important reflexes (e.g. heart rate, breathing, swallowing, vomiting) The pons - relay between cerebrum and cerebellum The medulla oblongata - visual reflex centre, auditory pathway Reticular formation – controls cyclic activities (e.g. sleep-wake cycle)
134
What is the function of the Diencephalon?
Body homeostasis (e.g. temperature, hunger) Comprised of: Thalamus – major sensory relay centre, influences mood and movement Subthalamus – nerve tracts and nuclei Epithalamus – olfactory stimulation, contains pineal gland Hypothalamus – major control centre for homeostasis + regulation of endocrine function
135
What is the function of the hypothalamus?
Location: Brain Links nervous and endocrine systems Control centre for homeostasis Controls the pituitary gland
136
What is the function of the Pituitary Gland?
Location: Brain “Master gland” Regulation of metabolic activities of other endocrine glands Releases hormones that regulate other glands (e.g. growth hormone, ADH) Regulation of growth
137
What is the function of the Pineal Gland?
Location: Brain Produces melatonin (regulates sleep-wake cycle) Maintains seasonal/circadian rhythm
138
What is the function of the Thyroid Gland?
Location: Neck Produces hormones (e.g. thyroxine) that regulate metabolism Induction of cytochrome P450s
139
What is the function of the Parathyroid Glands?
Location: behind the thyroid in the neck Regulate calcium and phosphate levels in blood Development of T & B cells
140
What is the function of the Adrenal Glands?
Location: On top of the kidneys Produce hormones for stress response (e.g. adrenaline, cortisol)
141
What is the function of pancreas?
Produces insulin and glucagon to regulate blood sugar
142
What is the function of the ovaries?
Produce oestrogen and progesterone which regulate the menstrual cycle and pregnancy
143
What is the function of the testes?
Produce testosterone which regulates sperm production and male traits
144
What are the four chambers of the heart?
Right atrium – deoxygenated blood from body Right ventricle - deoxygenated blood to lungs Left atrium – oxygenated blood from lungs Left ventricle – oxygenated blood to body
145
What are the vessels of the heart?
Vena cava – bring deoxygenated blood to the right atrium Pulmonary artery – carry deoxygenated blood to the lungs Pulmonary vein – return oxygenated blood to the left atrium Aorta – carries oxygenated blood from left ventricle to body
146
What are the valves of the heart?
Tricuspid Valve – separates right atrium and right ventricle Pulmonary Valve – separates right ventricle and the pulmonary artery Bicuspid (Mitral) Valve – separates left atrium and left ventricle Aortic Valve – separates left ventricle and the aorta
147
What are the components of blood and their functions?
Red blood cells (RBCs) – carry oxygen White blood cells (WBCs) – fight infection Platelets – help blood clot Plasma – fluid portion, carries hormones, nutrients, waste
148
What are the three types of blood vessels and their functions?
Arteries – carry blood away from the heart Veins – carry blood toward the heart Capillaries – tiny vessels for gas and nutrient exchange
149
What are the functions of the lymphatic system?
Removes foreign substances Combats disease Maintains body fluid balance Absorbs fats
150
What are the components of the lymphatic system and their function?
Lymph * Clear fluid that leaks out of blood capillaries into tissues * Contains white blood cells, especially lymphocytes Lymphatic Vessels * Network of tubes that carry lymph * One way flow * Similar to veins (have valves to prevent backflow) * Eventually drain into the subclavian veins, returning fluid to the bloodstream / Enter circulatory system via left or right subclavian vein Lymph Nodes * Filter lymph * Contain immune cells that trap and destroy pathogens * Found in clusters (e.g. neck, armpits, groin) Spleen * Filters blood, removes old red blood cells * Stores white blood cells Thymus * Where T cells mature * Most active during childhood, decreases in size in adults Tonsils + adenoids * Trap pathogens from mouth and nose Peyer’s patches * Found in the intestines * Monitor gut bacteria Bone Marrow * Produces lymphocytes and other blood cells
151
What are the components of the digestive system and their functions?
Mouth * Starts digestion with chewing (mechanical) * Tongue forms bolus (ball of food) * Salivary glands - secretes amylase for digestion of carbohydrates into maltose and dextrin Pharynx + esophagus * Pushes food into the stomach via swallowing and peristalsis Stomach * Contains gastric juice comprising of HCl, pepsinogen, lipase, mucus, intrinsic factor * Churns food with acid (HCl) and enzymes (e.g. pepsin) to break down proteins, carbohydrates and fats * Forms chyme (semi-liquid food). Small intestine * Main site of digestion and absorption * 3 parts: duodenum, jejunum, ileum * Villi & microvilli increase surface area for nutrient absorption Large Intestine (Colon) * Absorbs water and electrolytes * Forms and stores faeces Liver * Produces bile to break down fats * Detoxifies blood * Storage of nutrients Gallbladder * Stores and releases bile into small intestine Pancreas * Secretes enzymes (lipase, amylase, protease) and bicarbonate into the duodenum to aid digestion and neutralise stomach acid *Regulation of blood sugar
152
Describe the process of digestion.
1. Ingestion involves mouth, pharynx, oesophagus 2. Secretion and digestion * Involves mouth, pharynx, oesophagus, stomach, small intestine * Enzymes are secreted for chemical breakdown of large molecules 3. Absorption * Involves stomach, small intestine, large intestine * Results in smaller soluble molecules and water 4. Assimilation * Utilisation of small molecules by all cells of body 5. Excretion * Rejection of undigested particles + excretion of biochemical waste products
153
What are the main functions of the urinary system?
Produces, stores and eliminates urine Filters blood Reabsorbs water, salts, etc. Secretes hormones (adrenaline) Excretes waste products (main source of nitrogen excretion) Regulates blood pH and blood pressure
154
What are the components of the urinary system and their functions?
Kidneys * Filter blood to remove waste, toxins, and extra water * Produce urine and hormones (e.g. erythropoietin, renin) Ureters - thin tubes that transport urine from the kidneys to the bladder via peristalsis Bladder * Muscular sac that stores urine until it's ready to be excreted * Stretch receptors trigger the urge to urinate Urethra * Tube that carries urine from the bladder to outside the body * Shorter in females, longer in males
155
What are the components of the male reproductive system and their functions?
Testes * Production of spermatozoa (from puberty until old age) * Production of testosterone Vas deferens + Epididymis * There are two of each * Transfer spermatozoa from the testes * Seminal vesicle empties into it Seminal vesicles - secrete seminal fluid for transport of spermatozoa Prostate gland - secrete seminal fluid for transport of spermatozoa Penis * Transfers spermatozoa from male to female * Contains erectile tissue which fill with blood to produce erection
156
What are the male secondary sex characteristics?
Laryngeal changes - deep voice Growth of facial, pubic and axillary hair Receding hair at temples Increase in muscle and skeletal mass Growth spurt and characteristic male shape of body Increase in sex drive and aggression Increased sebaceous gland secretion (over secretion results in acne)
157
What are the components of the female reproductive system and their functions?
Ovaries * Production of ova/eggs (from puberty until menopause) * Production of oestrogen and progesterone Fallopian tubes - transfer ova/eggs from the ovaries Uterus - responsible for the nutrition and development of the fertilised egg cell and resulting embryo Vagina - receives spermatozoa Mammary glands - produces milk which provides nutrition for new infant after birth
158
What are the female secondary sex characteristics?
Development of breasts Characteristic female form + proportions (e.g. narrow shoulders, broad hips) Sex drive Pubic and axillary hair
159
What is homeostasis?
The process by which the body maintains a stable internal environment despite external changes
160
What variables are regulated by homeostasis?
Plasma [Na+], [K+], [Ca+] Osmolality pH Body temperature Plasma O2 and CO2 levels Arterial blood pressure
161
What is negative feedback?
When the response opposes the original stimulus to bring the system back to normal
162
How is osmolality controlled?
Hypothalamic osmoreceptors detect changes in body fluid osmotic pressure Changes in the secretion of antidiuretic hormone (induces water reabsorption, therefore less water loss) Stimulation or inhibition of thirst and drinking
163
What effect does diabetes have on osmolality?
When a patient is diabetic excess sugar gets excreted into the urine. This drives the removal of water into the urine to balance the osmotic difference This increases homeostatic regulation of water loss and thus increasing urination frequency and thirst
164
Which part of the body is responsible for maintaining body temperature?
The hypothalamus
165
How does the body respond when it is too cold?
Vasoconstriction = vessels constrict at the surface of the skin → reduction of blood flow to skin → reduction in heat loss Piloerection - traps insulating layer of air next to skin Thermogenesis * Shivering - rapid contraction of muscles generates heat * Activation of mitochondrial activity in brown adipose tissue which generates heat
166
How does the body respond when it is too hot?
Vasodilation = vessels dilate at the surface of the skin → increased blood flow to skin → increased heat transfer/loss Sweating - Evaporation of water from skin surface reduces temperature
167
What is a full agonist?
A drug that occupies receptors and fully activates them (in the same way as the endogenous ligand)
168
What is a partial agonist?
A drug that binds to receptors but produces a lower maximal effect than a full agonist
169
What is an antagonist?
A drug that binds to receptors without activating them, blocking the action of agonists
170
What is an inverse agonist?
A drug that binds to receptors but induces a pharmacological response opposite to that of the agonist (switches off activity)
171
Reversible drug interactions are mediated by ionic attractions or hydrophobic interactions Irreversible drug interactions are mediated by covalent bonds
172
What types of experiments measure drug action?
In vitro * Most common type of experiment * Use a piece of tissue dissected from an animal (or human) which is kept alive outside the body * Typical responses measured include: tension of a muscle, change in enzyme activity, change in hormone secretion, change in neurotransmitter secretion In vivo * Use a living animal (or human) * Typical responses measured include: increase in blood pressure, reduction in pain threshold, reduction in allergen, induced bronchoconstriction Ex vivo * Use a tissue or organ which has been removed from an animal that has been treated with a drug; the effects the drug has had on organ function are then tested in vitro * Typical experiments measure: whether long-term treatment with a drug induces liver damage or alters some aspect of brain biochemistry
173
What is Emax?
The maximum response (effect) the drug can produce, i.e. the “top” of the concentration-response curve Increasing the concentration of the drug will produce no greater effect
174
What is EC50?
The concentration of a drug that produces 50% of its maximum response Indicates the potency (i.e. sensitivity) of a drug The lower the EC50 the more potent the drug
175
What is the therapeutic index?
The ratio between the toxic dose of a drug and the dose producing the desired therapeutic effect The higher the therapeutic index the less chance of the drug producing toxic side-effects in therapeutic use
176
What is Kd?
The equilibrium dissociation constant The molar concentration of a drug needed to occupy 50% of receptors A measure of affinity (i.e. how tightly the receptor holds on to the drug once they come together) A lower Kd = higher affinity
177
What is efficacy?
The ability of a drug to activate the receptor once it has bound
178
What is the difference between EC50 and Kd?
EC50 is the the concentration needed to produce 50% of the maximal functional, which describes the potency of an agonist whereas Kd is the concentration at which 50% of receptors are occupied, which describes the agonists affinity for a receptor (i.e. the strength of binding) They will be equal in a simple system where there is no signal amplification and the response is directly proportional to receptor occupancy. However, in many biological systems there are spare receptors or signal amplification mechanisms (e.g. GPCRs with second messengers). In these cases, full activation can occur without all receptors being occupied, so the EC50 is lower than the Kd.
179
How will a full agonist and partial agonist look different, graphically?
A full agonist reaches a higher maximum response (Emax). A partial agonist has a lower maximum response (Emax). Full agonist produces a steeper, taller curve, whilst a partial agonist produces a shallower curve which plateaus earlier
180
What are the different forms of antagonism?
Receptor antagonism Chemical antagonism * One drug chemically inactivates another * Receptors are not involved * E.g. dimercaprol in arsenic poisoning Pharmacokinetic antagonism * One drug alters the way the body deals with another * Receptors are not involved * E.g. some antacids reduce the absorption of the antiepileptic drug phenytoin from the gut Physiological antagonism * Two drugs act to produce opposing effects, so canceling each other out * E.g. noradrenaline increases heart rate whilst acetylcholine decreases it (in this case both drugs are agonists, but acting on different receptors)
181
What are the different types of antagonists?
Competitive Non-competitive Reversible Irreversible
182
What is a competitive antagonist?
Compete with the agonist for the same site on the receptor molecule, but don’t activate Have affinity but zero efficacy
183
What is a non-competitive antagonist?
A drug which inhibits the effects of an agonist by acting at a different site on the receptor or another molecule closely associated with it
184
How does the presence of reversible competitive antagonists affect dose-response relationship?
Reversible competitive antagonists produce a parallel shift to the right of the agonist log concentration vs response curve EC50 changes but Emax remains the same because the action of the antagonist can be overcome by increasing agonist concentration
185
How does the presence of irreversible competitive antagonists affect dose-response relationship?
Irreversible competitive antagonists also produce rightward shift in the agonist log concentration vs response curve. The shift is not parallel, the Emax is lower. This is because inhibition is not overcome by increasing the agonist concentration – it is not surmountable
186
What is Bmax
Maximum binding capacity The total number of receptor sites available for binding in a sample
187
What is a dose ratio?
The ratio of the concentrations of agonist needed to produce the same effect in the presence and absence of antagonist For a reversible competitive antagonists, the dose-ratio should increase linearly with the concentration of the antagonist
188
What is pA₂?
The negative log of the antagonist concentration that doubles the EC₅₀ of an agonist/produces a dose ratio of 2 The bigger the pA₂ , the higher the affinity of the antagonist
189
How do you obtain pA₂ ?
Construct (log) concentration-response curves for the agonist in the presence of several concentrations of antagonist If there is no parallel shift seen, it is not a reversible competitive antagonist and therefore you cannot continue From the curves read the EC50 values for the agonist in the absence and presence of the different concentrations of antagonist (if using logEC50 calculate by doing inverse e to the power of reading from graph) Calculate the dose ratio for each concentration of antagonist (EC50 in presence of antagonist ÷ EC50 in absence of antagonist) Construct a Schild plot, by plotting log (dose ratio - 1) vs. log antagonist concentration The gradient = 1 The X-axis intercept = log(Kb) pA₂ = negative x-intercept ( - log Kb)
190
What is Kb?
The equilibrium dissociation constant for a competitive antagonist The lower the Kb, the higher the affinity for the receptor, the stronger the antagonist
191
What does a Schild plot show?
Whether antagonism is competitive and helps determine pA₂ and Kb which both indicate the affinity of the receptor for the antagonist
192
What are the components of the biological membrane and their functions?
Phospholipid bilayer Cholesterol - regulates membrane fluidity Glycoproteins + glycolipids - act as receptors which bind hormones or neurotransmitters triggering responses within the cell Transport proteins
193
What is the phospholipid bilayer naturally permeable to (via simple diffusion)?
Lipid-soluble molecules Oxygen Nitrogen Carbon dioxide Small uncharged molecules (e.g. urea, glycerol) Water (via aquaporins)
194
What factors effect membrane permeability?
Molecular size (ideal = small) Polarity (ideal = low) Charge (ideal = none) Solubility in oil (ideal = medium) Solubility in water (ideal = medium) Temperature
195
What are the different modes of transport across the membrane?
Simple diffusion Facilitated diffusion Active transport Endocytosis Exocytosis
196
What is simple diffusion?
The net movement of molecules from an area of high concentration to an area of low concentration Passive process - no requirement for energy Spontaneous process driven by an increase in entropy and therefore a decrease in free energy The rate of movement depends on the difference in concentrations (concentration gradient) Non-saturable
197
What is the mechanism of simple diffusion?
1. Molecule must break any bonds/interactions with the aqueous medium (e.g. hydrogen bonds) 2. Enter the lipid region 3. Cross the lipid region via diffusion 4. Leave the membrane at the other side 5. Reform any bonds with the aqueous medium
198
What types of molecules are transported via simple diffusion?
Non-polar gases (e.g. O2, CO2, N2) Hydrophobic molecules (e.g. steroid hormones, lipid soluble vitamins, small lipid soluble drugs) Urea
199
What factors affect the rate of diffusion?
The diffusion coefficient (D) Concentration gradient Thickness of the membrane Surface area
200
What is Fick's First Law of Diffusion?
J = -D x dC/dx Where * J = flux (rate) * D = diffusion coefficient * C = concentration * x = thickness * dC/dx = concentration gradient OR J = A x D x dC/dx Where A = surface area of membrane
201
What is facilitated diffusion?
The net movement of molecules from an area of high concentration to an area of low concentration, through the use of specific carrier and/or channel proteins
202
What types of proteins are involved in facilitated diffusion?
Carrier proteins - upon binding, undergo conformational changes that allow the molecule to pass through the membrane and be released on the other side Channel proteins - form open pores through the membrane, allowing the free diffusion of the molecule
203
What types of molecules are transported via facilitated diffusion?
polar and charged molecules (e.g. carbohydrates, amino acids, nucleosides, and ions)
204
What are the three classifications of transport proteins?
Uniport - transport one solute at a time Symport * Transports the solute and a co-transported solute at the same time in the same direction * S for same * E.g. uptake of sugars and amino acids into some animal cells is linked to the inward movement of Na+ Antiport * Transports the solute in (or out) and the co-transported solute in the opposite direction * i.e. one goes in the other goes out or vice-versa
205
What is active transport?
The movement of molecules from an area of low concentration to an area of high concentration, working up/against the concentration gradient and thus requiring energy
206
What is the difference between primary active transport and secondary active transport?
Primary active transport uses ATP as an energy source Secondary active transport uses an established electrochemical gradient to power the movement (always involves either symport or antiport proteins)
207
Why are membrane transporters important in pharmacy?
Drug targets E.g. SERT is the target for serotonin reuptake inhibitors (SSRIs) Drug disposition E.g. Breast Cancer Resistance Protein (BCRP) restricts intestinal absorption of anticancer drugs Disease E.g. Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) protein which is an ABC transporter
208
What is P-glycoprotein?
A type of ABC transporter which transports foreign molecules out of cells Often effects drugs (e.g. anticancer drugs, cardiac glycosides, glucocorticoids, immunosuppressive agents, HIV protease inhibitors)
209
What is catabolism?
Break down of molecules Produces/releases more energy than consumed
210
What is anabolism?
Formation of molecules Consumes more energy than produced
211
Describe the structure of ATP.
Nitrogenous base (adenine) Ribose sugar Triphosphate chain (α, β, ɣ)
212
How does ATP act as a link between anabolic and catabolic reactions in the body?
Catabolism makes ATP → ATP fuels anabolism → Anabolism builds complex molecules → repeat
213
How is ATP synthesised?
Substrate-level phosphorylation = involves transferring a high-energy phosphate group from an intermediate directly to ADP Oxidative phosphorylation = involves removal of electrons from NADH and FADH2 which are then passed through an electron transport chain (ETC) to oxygen Photophosphorylation (only occurs in chlorophyll-containing plant cells)
214
Why does ATP readily break down to ADP to release energy
There is electrostatic repulsion between the large, bulky, negatively-charged phosphate groups, which is decreased when ATP is hydrolysed. A free phosphate group enjoys resonance stabilisation, which is not possible in the ATP molecule because of the ester linkages
215
Why is carbohydrate metabolism important?
Produces glucose which is the primary source of energy for cells Glucose can be directed towards: * ATP production * Amino acid synthesis * Glycogen storage * Triglyceride formation
216
How does glucose enter the cells?
Via GluT transporters (facilitated diffusion) Insulin increases the number of transporters in the membrane which increases the rate of glucose entry into the cell Glucose then undergoes phosphorylation, trapping it inside the cell
217
What are the four stages of aerobic respiration and where do they occur?
1. Glycolysis/Fatty acid oxidation (cytoplasm) 2. Link Reaction (mitochondrial matrix) 3. Krebs Cycle/Citric Acid Cycle/The Tricarboxylic Acid Cycle (mitochondrial matrix) 4. Electron Transfer Chain (ETC)/Oxidative Phosphorylation (membrane of cristae/inner mitochondrial membrane)
218
Describe the process of glycolysis
Reaction 1 glucose + ATP → glucose-6-phosphate + ADP + H * A phosphate group is transferred from ATP to glucose, making glucose-6-phosphate * Catalysed by the enzyme hexokinase * Utilises a molecule of ATP * Traps glucose inside the cell Reaction 2 glucose-6-phosphate → fructose-6-phosphate * Six-membered pyranose ring becomes a five-membered furanose ring * Catalysed by the enzyme phosphoglucose isomerase Reaction 3 fructose-6-phosphate + ATP → fructose-1,6-bisphosphate + ADP + H * A phosphate group is transferred from ATP to fructose-6-phosphate, producing fructose-1,6-bisphosphate * This step is catalysed by the enzyme phosphofructo-1-kinase (PFK-1) * Utilises another molecule of ATP Reaction 4/5 fructose-1,6-bisphosphate → 2 glyceraldehyde-3-phosphate * Fructose-1,6-bisphosphate splits to form two three-carbon sugars: dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate * This is catalysed by the enzyme aldolase * DHAP is converted into glyceraldehyde-3-phosphate * This is catalysed by the enzyme triphoshpate isomerase (TIM) Reaction 6 2 glyceraldehyde-3-phosphate + 2NAD + 2P → 2 (1,3-bisphosphoglycerate) + 2NADH + 2H * The two molecules of glyceraldehyde-3-phosphate are oxidised and phosphorylated to form 1,3-bisphosphoglycerate * This is catalysed by the enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) * Substrate level phosphorylation * Two molecules of NAD are reduced to NADH Reaction 7 2 (1,3-bisphosphoglycerate) + ADP → 2 (3-phosphoglycerate) + 2ATP * Each 1,3-bisphosphoglycerate molecule donates one of its phosphate groups to ADP to form 3-phosphoglycerate and ATP * Catalysed by the enzyme phosphoglycerate kinase (PGK Reaction 8 2(3-phosphoglycerate) → 2(2-phosphoglycerate) * The two molecules of 3-phosphoglycerate are converted into the isomer, 2-phosphoglycerate * Catalysed by the enzyme phosphoglycerate mutase (PGM) Reaction 9 2(2-phosphoglycerate) → 2PEP(phosphoenolpyruvate) * The two molecules 2-phosphoglycerate lose a molecule of water, forming an enol, phosphoenolpyruvate (PEP) * Catalysed by the enzyme enolase Reaction 10 2PEP + ADP + H → 2Pyruvate + 2ATP * The two PEP molecules readily donate the phosphate group to ADP making 1 molecule of ATP each and forming the end product of glycolysis: pyruvate
219
What are the net products of glycolysis?
Glucose → 2 Pyruvate 2 ATP used 4 ATP produced Net gain = 4 - 2 = 2 ATP 2 NADH generated Total = 2 NADH, 2 ATP, 2 Pyruvate
220
How are the products of glycolysis formed?
Glucose uses one molecule of ATP to form glucose-6-phosphate (-1 ATP) Fructose-6-phosphate uses one molecule of ATP to form fructose-1,6-bisphosphate (-1 ATP) Glyceraldehyde-3-phosphate generates one molecule of NADH each (2 x 1 = 2 NADH) 1,3-bisphosphoglycerate generates one molecule ATP each (2 x 1 = 2 ATP) Phosphoenolpyruvate forms one molecule of pyruvate and one molecule of ATP each (2 ATP, 2 Pyruvate) Total = 2 NADH, 2 ATP, 2 Pyruvate
221
What happens to pyruvate after glycolysis?
With O₂ : Pyruvate → Acetyl-CoA (in mitochondria) Without O₂ : Pyruvate → Lactate (reduction, in cytosol, reversible by liver)
222
Describe the process of the Link Reaction/Pyruvate Oxidation
A carboxyl group is removed from pyruvate in the form of CO₂ The resultant 2 carbon molecule is oxidised by removing a hydrogen atom to form acetate. The hydrogen is picked up by NAD+ to form reduced NAD. The acetyl group is attached to Coenzyme A (CoA) an organic molecule derived from vitamin B5, to form acetyl CoA This process is catalysed by the enzyme pyruvate dehydrogenase (PDH)
223
Describe the Krebs Cycle/Tricarboxylic Acid Cycle/Citric Acid Cycle
1. Acetyl CoA (2C) joins with oxaloacetate (4C) to form citrate (6C) 2. After rearrangement the six-carbon molecule releases two of its carbons as carbon dioxide molecules, producing a molecule of NADH each time 3. The remaining four-carbon molecule undergoes a reaction which produces a molecule of ATP (in some cell GTP instead) 4. The resultant four-carbon molecule is oxidised, forming a molecule of FADH2. 5. Oxaloacetate is regenerated, forming a molecule of NADH
224
What are the net products of the Krebs Cycle?
2 x CO2 1 x ATP/GTP 1 x FADH2 3 x NADH For one molecule of glucose → 2 pyruvates → 2 acetyl CoA So Total = 2ATP, 2FADH2, 6NADH
225
Describe the process of Oxidative Phosphorylation / the Electron Transfer Chain (ETC)
All of the accumulated reduced coenzymes (NADH, FADH2) release the hydrogens which split into protons (H+) and electrons (e-) The electrons are passed down a series of electron carrier proteins embedded in the intermembrane/passed along an electron transport chain, losing energy as they move (FADH2 via Complex II & NADH via complex I) This redox energy is used to pump protons from the mitochondrial matrix into the intermembrane space. This creates a proton gradient (higher concentration in the intermembrane space than the matrix). As a result the protons will move back into the matrix through ATP synthase via chemiosmosis. The diffusion of the hydrogen ions creates a proton motive force which drives the rotation of the ATP synthase enzyme causing the phosphorylation of ADP and therefore formation of ATP Oxygen is the final electron acceptor in the electron transport chain. The oxygen combines with the electrons and the protons to form water.
226
How many ATP molecules can NADH generate?
3
227
How many ATP molecules can FADH2 generate?
2
228
Why does NADH generate more ATP than FADH2?
FADH2 donates electrons via Complex II, whereas NADH donates electrons via Complex I. Complex I is in a higher energy state than complex II so there is more energy for the production of ATP
229
What is the ATP yield from one glucose molecule?
From Glycolysis: 2 ATP + 2 NADH From Acetyl-CoA formation: 2 NADH From Krebs Cycle: 2 ATP + 6 NADH + 2 FADH2 From ETC: NADH: 3 ATP each → 10 x 3 = 30 FADH2: 2 ATP each → 2 x 2 = 4 Total = 2 + 2 + 30 + 4 = 38 (Some sources state 36 as 2 ATPs are used to move the 2 NADH formed in glycolysis from the cytoplasm into the matrix for the ETC)
230
Why is the ATP yield a theoretical maximum?
Mitochondrial membrane leakage: Protons leak back into the matrix → reduces proton motive force → decreases of ATP synthase activity → lowers ATP production Uncoupling proteins (e.g. dinitrophenol): Provide an alternative pathway for H+ to pass back across the inner mitochondrial membrane → bypass ATP synthase → dissipate energy as heat instead of generating ATP
231
Why is Dinitrophenol poisonous?
Dinitrophenol (DNP) acts as an uncoupling agent which dissolves in the inner mitochondrial membrane and carries H+ from the intermembrane space back into the matrix. This means protons bypass ATP synthase, decreasing the proton gradient and therefore the proton motive force. As a result ATP production is reduced and the energy from the ETC is released as heat, which can cause fatal hyperthermia.
232
Why is Oligomycin poisonous?
Inhibits ATP synthesis by blocking the proton channel, preventing H⁺ from flowing back into the matrix. This halts ATP production via oxidative phosphorylation leading to cellular energy failure and ultimately cell death
233
What is glycogenesis?
The process by which the body converts excess glucose into glycogen for storage Insulin stimulates hepatocytes and skeletal muscle cells to synthesise glycogen
234
What is glycogenolysis?
The process by which glycogen stored in hepatocytes is broken down into glucose and released into blood Stimulated by glucagon and epinephrine/adrenaline
235
What is gluconeogenesis?
The production of glucose from non-carbohydrate sources (glycerol, lactic acid, amino acids) Occurs in the liver Helps maintain blood glucose levels during fasting, starvation, or intense exercise Stimulated by glucagon and cortisol
236
How are lipids transported in the body?
Lipoproteins = Lipid core + apolipoprotein outer shell
237
What are the different types of lipoproteins?
Chylomicrons * Form in the small intestine mucosal epithelia * Transport dietary lipids to adipose tissue VLDL (Very Low-Density Lipoproteins) * Formed in hepatocytes (liver) * Transport endogenous lipids to adipocytes LDL (Low-Density Lipoproteins) * "bad cholesterol” * Delivered to body cells for repair and synthesis * Can deposit cholesterol to form fatty plaques HDL (High-Density Lipoproteins) * "good cholesterol" * Removes excess cholesterol from body cells + blood and delivers them to the liver for elimination
238
Which ApoE genotype is linked to Alzheimer's disease?
Three allelic variants: E2, E3, E4 Each have varying degrees of risk for the onset of AD: E4 > E3 > E2 E4/E4 genotype = High risk E2/E2 genotype = Low risk
239
What are the two sources of cholesterol in the body?
Dietary intake Endogenous synthesis in hepatocytes
240
How do statins treat high cholesterol levels?
Inhibit 3-Hmg-CoA reductase which is an enzyme found in the liver involved in the cholesterol synthesis pathway.
241
How are lipids metabolised?
Oxidised to provide ATP Stored in adipose tissue if not needed for ATP production Lipid catabolism (lipolysis / fatty acid oxidation / β-oxidation) * Breaks triglycerides → glycerol + fatty acids * Generates acetyl-CoA, which enters the Krebs cycle to produce ATP * Occurs in mitochondria * Promoted by epinephrine/norepinephrine Lipid anabolism (lipogenesis / fatty acid synthesis): * Converts glucose/amino acids → fatty acids * Occurs in cytosol of hepatocytes and adipocytes * Happens in calorie surplus (when more calories are consumed than needed for ATP production)
242
Give examples of drugs which affect β-oxidation.
Caffeine * Stimulates β-oxidation * Commonly found in high-performance energy gels used by runners Mildronate (meldonium) - Inhibits fatty acid oxidation Valproate - Inhibits fatty acid oxidation
243
What is amino acid anabolism?
The synthesis of new proteins in ribosomes, used for enzymes, hormones, and structural proteins
244
What are the three main fates of excess amino acids during catabolism?
Excess amino acids cannot be stored and are: Oxidised to produce ATP or intermediates for metabolic cycles Converted into glucose (via gluconeogenesis) Converted into fatty acids (via lipogenesis) and stored as triglycerides in adipose tissue
245
When does gluconeogenesis from amino acids occur?
During prolonged starvation, fasting, or low-carb intake
246
What must happen before amino acids are catabolised?
They must undergo deamination - i.e. removal of the amino group
247
Where does deamination mainly occur?
Liver
248
What does deamination produce?
Produces ammonia (NH₃), which is toxic and converted into urea by the liver and excreted in urine by the kidneys
249
What is transamination?
The process where an amino group (-NH₂) is transferred from one amino acid to a keto acid to form a new amino acid Catalysed by enzymes called transaminases or aminotransferases E.g. ALT (alanine aminotransferase), AST (aspartate aminotransferase) Requires the coenzyme pyridoxal phosphate (PLP), derived from vitamin B6
250
What is function of transamination?
Synthesise amino acids that are not obtained from the diet (non-essential amino acids) Converts amino acids into intermediates that can enter the Krebs cycle or be used in gluconeogenesis
251
What are the key molecules of metabolism?
Glucose 6-phosphate Pyruvate Acetyl-CoA
252
Why is glucose 6-phosphate important for metabolism?
Produced shortly after glucose enters the cell “traps” the glucose inside the cell Has many uses: Glycolysis (important for energy/ATP production) Synthesis of glycogen (energy stores) Release of glucose into blood stream Synthesis of nucleic acids Synthesis of UDP-glucuronic acid for Phase II metabolism
253
Why is pyruvate important for metabolism?
The end product of glycolysis Enters the Krebs cycle, in the presence of sufficient oxygen (oxidative phosphorylation) When there is a lack of oxygen can be used for: * Production of lactic acid * Production of alanine (transamination) * Gluconeogenesis
254
Why is Acetyl-CoA important for metabolism?
The entry molecule into the Krebs cycle Used to synthesise fatty acids and cholesterol (lipogenesis)
255
What is the absorptive state?
It's the period during and shortly after eating when ingested nutrients (glucose, amino acids, triglycerides in chylomicrons) enter the bloodstream
256
What is the primary energy source during the absorptive state?
Glucose is readily available and used for ATP production
257
What happens to excess fuel during the absorptive state?
It is stored in hepatocytes, adipocytes, and skeletal muscle cells.
258
What hormone is released during the absorptive state?
Pancreatic beta cells release insulin which promotes entry of glucose and amino acids into cells
259
When does the postabsorptive state occur?
About 4 hours after eating, when nutrient absorption is complete
260
Describe the postabsorptive state?
Blood glucose levels starts to fall Energy is derived from stored fuels
261
What are the two main ways the body maintains blood glucose levels in the postabsorptive state?
Glucose production Glucose conservation
262
What are the sources of glucose production in the postabsorptive state?
Breakdown of liver glycogen Lipolysis Gluconeogenesis (from lactate and/or amino acids)
263
How is glucose conserved during the postabsorptive state?
Oxidising fatty acids, lactate, amino acids and ketone bodies Breaking down muscle glycogen
264
Give examples of minerals used in the body?
Calcium Copper Iodine Iron
265
What are the two classes of vitamins?
Fat-soluble vitamins * Vitamins A, D, E, and K Water-soluble vitamins * Vitamins B1 (thiamine), B2 (riboflavin), B3 (niacin), B5 (pantothenic acid), B6, B7 (biotin), B9, B12 (cobalamin) * Vitamin C (L-ascorbic acid)
266
What is metabolic syndrome?
A disorder of energy usage and storage Diagnosed by the clustering of at least three of the following: * Central obesity * High blood pressure * High blood sugar * High serum triglyceride * Low HDL
267
What are inborn errors of metabolism?
Genetic defects in enzyme-encoding genes
268
What is Von Gierke’s Disease?
Glucose-6-phosphatase deficiency (carbohydrate metabolism disorder)
269
What is Forbe’s Disease?
Glycogen debranching enzyme deficiency (carbohydrate metabolism disorder)
270
What is Andersen Disease?
Glycogen branching enzyme deficiency (carbohydrate metabolism disorder)
271
What is Phenylketonuria (PKU)?
Autosomal recessive amino acid metabolism disorder Deficiency in phenylalanine hydroxylase (PAH) Causes build-up of Phe, which competes for the LNAA transporter in the blood-brain-barrier, reducing entry of key amino acid used to synthesise neurotransmitters and neuronal proteins Reduces Tyr synthesis – reduced dopamine, norarenaline and adrenaline synthesis
272
How is PKU treated?
Treated with a diet low in foods containing phenylalanine and aspartame (which is metabolised into Phe)
273
What are the complications of PKU?
Intellectual disability Seizures Behaviour problems Microcephaly
274
What is MCADD?
Medium-chain acyl-coenzyme A dehydrogenase deficiency An autosomal recessive lipid metabolism disorder Prevents breakdown of fatty acids into acetyl-CoA Managed by avoiding fasting and using glucose supplements during illness
275
What are the symptoms of diabetes?
Frequent urination Increased thirst Increased hunger Diabetic ketoacidosis
276
What are some of the long term complication of diabetes?
Cardiovascular disease Kidney disease Foot ulcers
277
What are the three types of diabetes?
Type 1 * Pancreatic failure to produce insulin due to loss of beta-cells * No known cause * Treated using insulin injections Type 2 * Insulin resistance * Can lead to lack of insulin * Caused by obesity and insufficient exercise * Treated using diet Gestational diabetes - occurs in pregnant women
278
What is diabetic ketoacidosis (DKA)?
When the body shifts to fat metabolism (fatty acid oxidation) due to insulin deficiency Produces ketone bodies (β-hydroxybutyrate (main) + Acetoacetate) Occurs mainly in type 1 diabetes
279
What are the symptoms of DKA?
Nausea Vomiting Thirst Excessive urination Kussmaul breathing (a deep, rapid, and labored breathing pattern) Fruity breath Cerebral oedema (in children) Coma Death
280
What are the three stages of DKA?
Mild * pH 7.2 – 7.3 * Serum bicarbonate 10 – 15 mM * Person is alert Moderate * pH 7.1 – 7.2 * Serum bicarbonate 5 – 10 mM * Person is mildly drowsy Severe * pH < 7.1 * Serum bicarbonate < 5 * Patient is in a coma
281
How is DKA treated?
IV fluids IV insulin Potassium (replacement of potassium lost due to osmotic diuresis)
282
What are the main types of lipids?
Fats Oils Waxes Phospholipids Glycolipids Steroids
283
Describe the structure of lipids.
Glycerol + 3 fatty acids = Triacylglycerol (Triglyceride) Ester bond
284
What are the different types of fatty acids?
Saturated (e.g. stearic acid) Monounsaturated (e.g. oleic acid) Polyunsaturated (e.g. - E.g. Linoleic Acid, Omega-3 and Omega-6)
285
What are the differences between saturated and unsaturated fatty acids?
Saturated: * no double bonds * form straight chains * tightly packed + strong intermolecular forces → high melting points → solid at room temperature * found in butter, animal fats Unsaturated * Contain double bond(s) * Mono = one double bond * Poly = multiple double bond * Kink/bending → lower melting points → liquid at room temp
286
Which geometric configuration or most fatty acids found in?
Cis configuration - creates more fluidity Trans fats are linked to coronary heart disease
287
What are waxes?
Esters formed from long-chain fatty acids + alcohols No double bonds High melting points Chemically inert Provide a waterproof coating on the surfaces of many organisms
288
Describe the structure of phospholipids
glycerol 2 fatty acids a phosphate group which is usually linked to a small ionic nitrogen-containing group (e.g. choline, serine, ethanolamine)
289
What are glycolipids?
Comprised of: glycerol + 2 fatty acids + a sugar Involved in cell recognition and cell signalling
290
What are steroids?
Organic compounds containing four fused rings Includes: * Cholestanes (e.g. Cholesterol) * Estranes (e.g. Estradiol) * Androstanes (e.g. Testosterone) * Pregnanes (e.g. Progesterone)
291
What are the functions of cholesterol?
Structural role in membranes Precursor of steroid hormones (e.g. corticosteroids from adrenal cortex) Vitamin D synthesis Bile acid synthesis
292
What are the functions of fats?
Energy storage Carrier for fat-soluble vitamins (A, D, E, K) Membrane fluidity + signalling Immune response (e.g. via Eicosanoids)
293
What is the difference between D and L Isomers?
Based on the position of the OH group on the last chiral carbon D-isomer: OH group is on the right L-isomer: OH group is on the left
294
What are monosaccharides?
Simple sugars made of a single sugar unit. E.g. glucose, fructose, galactose, ribose
295
What are disaccharides?
Carbohydrates made of two monosaccharide units linked by a glycosidic bond E.g. Sucrose = glucose + fructose Lactose = glucose + galactose
296
What are polysaccharides?
Carbohydrates made of many monosaccharide units linked by glycosidic bonds E.g. starch, glycogen, cellulose
297
Most natural sugars are in which stereoisomer form?
D-isomer
298
What is the difference between enantiomers and diastereomers?
Both are stereoisomers - i.e. have same molecular formula and connectivity but different spatial arrangement Enantiomers are non-superimposable mirror images while diastereomers are not mirror images Enantiomers have identical physical properties (except for optical rotation), but diastereomers do not
299
How can calculate the number of stereoisomers a molecule with multiple chiral centres will have?
A molecule with n chiral centers has 2ⁿ stereoisomers
300
What happens to monosaccharides in aqueous solution?
They can form cyclic structures via an internal reaction between the carbonyl group and a hydroxyl group
301
What does cyclisation of a monosaccharide create?
A new chiral center at carbon 1, known as the anomeric carbon
302
What are the two anomeric forms of cyclic monosaccharides?
α-anomer: OH on C1 is down, opposite to CH₂OH on C5 β-anomer: OH on C1 is up, same side as CH₂OH
303
What is the structure formed when an aldose cyclises?
A hemiacetal forming a pyranose (6-membered ring)
304
What is an aldose?
A monosaccharide that contains an aldehyde group (-CHO) at carbon 1 E.g. glucose
305
What is a ketose?
A monosaccharide that contains a ketone group (C=O) E.g. fructose
306
What structure does a ketose form when cyclised?
A hemiketal, forming a furanose (5-membered ring)
307
What are the two types of glycosidic bond?
O-linked * Sugar is linked to another molecule via oxygen (OH + OH) N-linked * Sugar is linked via a nitrogen atom (OH + NH)
308
What are the biological functions of carbohydrates?
Energy source + storage (e.g. glucose, glycogen) Membrane structure + recognition and signaling (e.g. glycolipids) Nucleic acid structure (ribose in RNA, deoxyribose in DNA) Protein modification: * N-linked glycosylation (to Asn in Asn-X-Ser/Thr motif) * O-linked glycosylation (to Ser or Thr)
309
What roles do carbohydrates play in medicine?
As drugs themselves E.g. streptomycin (antibiotic) In drug delivery systems * Modified carbohydrates (e.g. cyclodextrins) improve solubility and bioavailability As excipients E.g. Lactose and starch are used as binders + fillers As conjugates for enhanced targeting or uptake Vaccines Diagnostics * Carbohydrate markers used in blood tests Scaffold in drug discovery
310
Why are enzymes important in pharmacy?
Enzymes are often drug targets Affect drug metabolism
311
Examples of enzyme cofactors
metal ions - Zn2+ , Mg2+ , Mn2+
312
Examples of coenzymes
Nicotinamides - e.g. NADP(H), NAD(H) Flavins - e.g. FMN, FADs, prFMN
313
What is an apoenzyme?
An enzyme without a cofactor
314
What is a holoenzyme?
An enzyme with a cofactor
315
How are enzyme-substrate complexes stabilised?
Van der Waals interactions Hydrogen bonding Electrostatic interactions Entropic contributions due to “hydrophobic effect”
316
What are the different classes of enzymes?
Oxidoreductases (EC1) Transferases (EC2) Hydrolases (EC3) Lyases (EC4) Isomerases (EC5) Ligases (EC6) Translocases (EC7)
317
What are oxidoreductases?
Enzymes which catalyse oxidation and/or reduction reactions E.g. dehydrogenases, reductases
318
What are transferases?
Enzymes which catalyse the transfer of a functional group/moiety from donor to acceptor E.g. aminotransferases, methyltransferases, prenyltransferases
319
What are lyases?
Enzymes which catalyse the cleavage of various bonds not via hydrolysis and oxidation E.g. ammonia lyases
320
What are isomerases?
Enzymes which catalyse isomerisation within a single molecule
321
What are ligases?
Enzymes which link two molecules together at a specific site, usually with covalent bonds E.g. C-N ligases, DNA ligases
322
What are translocases?
Enzymes which mediate the movement of molecules/ions across cell membranes or their separation within a membrane E.g. ADP/ATP translocase
323
What is the lock and key model?
States that an enzyme's active site has a specific, rigid shape that exactly matches the shape of its substrate - just like a key fits into a specific lock Highly stereospecific
324
What is the induced fit model?
States that when a substrate begins to bind, it induces a conformational change in the enzyme that results in a more precise fit Takes into account the flexibility of proteins
325
What are the most common enzyme catalytic mechanisms?
Acid-Base Catalysis Covalent Catalysis Electrostatic Catalysis
326
What is the catalytic triad of serine proteases?
Ser 195 * The serine -OH group acts as a nucleophile and attacks the carbonyl carbon of the peptide bond of the substrate His 57 * The histidine nitrogen accepts the hydrogen in the OH group of serine hence coordinating the attack of the peptide bond Asp 102 * Aspartic acid contains a carboxyl group which hydrogen bonds with the more electronegative histidine Nitrogen
327
What are the essential residues of reductive aminases?
D169 - deprotonates amines Y177 - anchors ketone group
328
What factors effect enzyme activity?
Temperature pH Enzyme concentration Substrate concentration Presence of inhibitors or activators
329
What is the effect of temperature on enzymatic activity?
Enzyme activity increases with temperature up to an optimum. Beyond the optimum, denaturation occurs, where the enzyme loses its structure and therefore function
330
What is the effect of pH on enzymatic activity?
Each enzyme has an optimal pH (e.g., pepsin ~pH 2, amylase ~pH 7) Extremes of pH will denature the enzyme and so destroy activity This is because they are made of proteins containing many ionisable groups, which will exist in a whole series of different states of ionisation depending on the pH This will have its greatest effect in the active site where ionization may affect shape and any ionic binding of substrate The ionization of the substrate(s) must also be considered
331
What is the Michaelis-Menten Equation?
Describes how the reaction rate varies with substrate concentration v = Vmax[S] ÷ Km + [S] Where * v = V0 = initial reaction rate * Vmax = maximum reaction rate (when enzyme is saturated) * Km = Michaelis constant (substrate concentration at half Vmax)
332
What assumptions are made for the Michaelis-Menten model to be valid?
The formation of the enzyme-substrate (ES) complex is reversible The concentration of ES reaches a steady state (doesn't change over time)
333
How is the Michaelis-Menten model graphically represented?
Plot V₀ vs. [S]
334
Describe the Michaelis-Menten plot (V₀ vs. [S])
At relatively low concentrations of substrate, V0 increases almost linearly with an increase in [S] At higher substrate concentrations, V0 increases by smaller and smaller amounts in response to increases in [S] Finally, a point is reached beyond which increases in V0 are vanishingly small as [S] increases. This plateau-like V0 region is close to the maximum velocity, Vmax
335
What does Km tell you?
The substrate concentration at which half the enzyme active sites are occupied by the substrate Indicates substrate affinity Lower Km = higher affinity (and tighter binding)
336
What is Kcat?
The number of substrate molecules converted to product in a given unit of time on a single enzyme molecule when the enzyme is saturated with substrate The turnover number
337
What is the specificity constant?
Kcat ÷ Km A measure of catalytic efficiency
338
What are the different ways enzyme kinetics can be visualised graphically?
Michaelis-Menten Plot = V₀ vs [S] Lineweaver-Burke Plot = 1/V₀ vs 1/[S] Hanes-Woolf Plot = [S]/V₀ vs [S] Eadie-Hofstee Plot = V₀ vs V₀/[S]
339
What is the Lineweaver-Burke Plot?
1/V₀ vs 1/[S] Gradient = Km/Vmax y-intercept = 1/Vmax x-intercept = -1/Km
340
What is the Hanes-Woolf Plot?
[S]/V₀ vs [S] Gradient = 1/Vmax y-intercept = Km/Vmax x-intercept = -Km
341
What is the Eadie-Hofstee Plot?
V₀ vs V₀/[S] Gradient = -Km y-intercept = Vmax x-intercept = Vmax/Km
342
How do you choose which enzyme in a pathway to target?
The enzyme which catalyses the first step in the metabolic scheme The enzyme which catalyses the rate-limiting step The enzyme which is functionally the most critical
343
What are the types of enzyme inhibition?
Reversible competitive Reversible uncompetitive Reversible non-competitive (mixed) Irreversible
344
What is reversible competitive enzyme inhibition?
Inhibitor competes with substrate at active site Increases Km Vmax unchanged E.g. Captopril (ACE inhibitor), Trimethoprim
345
What is reversible uncompetitive enzyme inhibition?
Inhibitor binds only after substrate binds → traps enzyme in ES form Decreases both Km and Vmax E.g. Finasteride, Methotrexate
346
What is reversible non-competitive (mixed) enzyme inhibition?
Inhibitor binds to enzyme or ES complex at a different site Both inhibitor and substrate can bind to the enzyme simultaneously Vmax decreases Km unchanged or variable E.g. Etoposide, Tacrine
347
What is irreversible enzyme inhibition?
Forms covalent bond with enzyme, permanently inactivating it E.g. Penicillin, Diisopropyl fluorophosphate (DIFP)
348
What is Ki?
Dissociation constant of enzyme-inhibitor complex Difference in gradients = 1 + [I]/Ki Lower Ki = more potent inhibitor (tighter binding)
349
What is Ki'?
Dissociation constant for enzyme-substrate-inhibitor complex Difference in gradients = 1 + [I]/Ki' Lower Ki = more potent inhibitor (tighter binding)
350
How can you identify/determine inhibition type using enzyme kinetic data?
1. Measure initial rates, v at varying substrate concentrations [S], with and without inhibitor 2. Construct a Lineweaver-Burk Plot (1/v vs 1/[S]) using the data obtained For: Competitive - Lines intersect at y-axis (same Vmax, increased Km) Uncompetitive - Parallel lines (lower Vmax and Km) Mixed - Lines intersect left of y-axis (lower Vmax, variable Km) For all increasing [I] = steeper/lower gradient (upwards)
351
What is the structure of an amino acid?
Composed of: * An amino group * Carboxylic acid * Hydrogen atom * R-group/ side chain which is variable (can be non polar, polar uncharged, polar charged, etc.)
352
What are the different types of amino acid?
Non polar aliphatic amino acids E.g. Glycine, Alanine, Proline, Valine, Leucine, Isoleucine, Methionine Aromatic amino acids E.g. Phenylalanine, Tyrosine, Tryptophan Polar uncharged amino acids E.g. * Serine (hydroxyl group) * Threonine (hydroxyl group) * Asparagine (amide group) * Glutamine (amide group) * Cysteine (sulfhydryl group - a weak acid, mostly uncharged at neutral pH, can form disulphide bridges) Positively charged amino acids E.g. Lysine, Arginine, Histidine Negatively charged amino acids E.g. Aspartate (carboxyl group), Glutamate (carboxyl group)
353
What happens to an amino acid at low pH?
The carboxylic acid becomes protonated, and the molecule is positively charged
354
What happens to an amino acid at high pH?
The amino group loses its proton, making the molecule negatively charged
355
What is the isoelectric point?
The pH at which an amino acid has no net charge and exists as a zwitterion (positively charged amino group and negatively charged carboxyl group) pI = (pKa1 + pKa2) ÷ 2
356
What is a zwitterion?
A molecule with both a positive and negative charge but an overall neutral charge
357
How many pKa values do amino acids with uncharged side chains have?
Two one for the α-carboxyl group and one for the α-amino group E.g. glycine
358
How many pKa values do amino acids with ionisable side chains have?
Three one for the α-carboxyl group, one for the α-amino group and one for the side chain (R group) E.g. lysine, arginine, glutamic acid, aspartic acid
359
What are the four levels of protein structure?
Primary Secondary Tertiary Quaternary
360
What is the primary structure of a protein?
The number, type and sequence of amino acids joined together by strong covelant peptide bonds in a condensation reaction to form a polypeptide chain
361
What acts as the nucleophile during peptide bond formation?
The amino group of one amino acid displaces the hydroxyl group of another
362
How is a peptide bond formed?
The amino group of one amino acid acts as a nucleophile to displace the hydroxyl group of another amino acid
363
What is a peptide bond?
A covalent bond formed between the carboxyl group of one amino acid and the amino group of another
364
What causes resonance in the peptide bond?
The lone pair on the nitrogen is delocalised onto the carbonyl oxygen, creating partial double bond character
365
What is the conformational structure of a peptide bond?
Planar
366
Can peptide bonds rotate?
No, rotation around the peptide bond is not permitted due to partial double bond character
367
What is the more stable geometric isomer of the peptide bond, cis or trans?
The trans form which is more stable (exception = proline)
368
What is the N-terminus of a protein?
The end with a free NH₂ group, not involved in a peptide bond
369
What is the C-terminus of a protein?
The end with a free COOH group, not involved in a peptide bond
370
How are amino acids numbered in a protein?
From the N-terminus to the C-terminus
371
What is the secondary structure of a protein?
The three dimensional structure determined solely by hydrogen bonds formed between the carbonyl oxygen and the amino nitrogen
372
What is an α-helix?
A right-handed coil where the C=O group of one amino acid hydrogen bonds with the NH group four residues ahead
373
Which amino acids promote α-helix formation?
Small hydrophobic residues like alanine and leucine due to low steric hindrance
374
Which amino acids break α-helices and why?
Proline: No rotation around N–Cα bond Glycine: Lacks an R group, allowing other conformations
375
What is a β-pleated sheet?
A secondary structure where hydrogen bonds form between NH and CO groups of adjacent polypeptide chains
376
What is the difference between parallel and antiparallel β-sheets?
Parallel: Chains run in the same direction Antiparallel: Chains run in opposite directions and can form loops or U-turns
377
Which amino acids are favoured in β-sheets?
Larger bulky residues (e.g. valine, isoleucine, phenylalanine) which can stabilise the extended structure through hydrogen bonding and packing interactions
378
What are turns in protein secondary structure?
Short regions allowing the protein backbone to change direction, often connecting α-helices and β-sheets.
379
What is a β-turn?
A 4-residue turn stabilised by a hydrogen bond between the CO of residue 1 and NH of residue 4.
380
Which amino acids are common in β-turns and why?
Proline at position 2 (rigidity) Glycine at position 3 (flexibility)
381
What is the tertiary structure of a protein?
The three-dimensional structure is determined by interactions between R groups and their properties
382
What types of interactions are involved in tertiary structure?
Hydrogen bonds (between polar R groups) Ionic bonds (between charged R groups) Hydrophobic interactions (non-polar side chains) Electrostatic interactions (e.g. aspartate, lysine) Van der Waals forces Disulfide bridges (e.g. cystine)
383
What are disulfide bridges and where can they form?
Covalent bonds between sulfur atoms of two cysteine residues Can be intramolecular (within one polypeptide chain) or intermolecular (between chains)
384
What are the two major classes of tertiary protein structure?
Fibrous proteins Globular proteins
385
What are key characteristics of fibrous proteins?
Insoluble (hydrophobic exterior) Made from a single secondary structure Much less compact than globular proteins Play important roles in providing structural rigidity and in contractile movement E.g. Collagen, Keratin, Fibrin, Myosin
386
What are key characteristics of globular proteins?
Hydrophobic side chains fold inward (forming water-free pockets) Polar side chains face outward (hydrophilic exterior) Function as enzymes, transporter, antibodies, etc. E.g. Hemoglobin, Amylase, Insulin, Immunoglobulins
387
What is the quaternary structure of a protein?
When a protein contains more than one polypeptide chain (subunits) Can be identical subunits (homo) or different (hetero) Not all proteins have a quaternary structure
388
What factors can cause protein denaturing?
Heat pH extremes Organic solvents (e.g. alcohols) - interfere with hydrophobic interactions Reducing agents - convert disulphides into thiols Detergents - disrupt hydrophobic interactions Salts - causes aggregation and precipitation UV radiation Urea
389
What are some examples of protein functions?
Signalling (hormones) Catalysis (enzymes) Genetics (e.g. histones, transcription factors) Defence (antibodies) Movement (e.g. actin, myosin) Structure (e.g. collagen, elastin) Transport (e.g. membrane proteins, myoglobulin, albumin, haemoglobin)
390
Describe the structure of myoglobin
Globular protein ~ 153 amino acids Monomeric (8 α-helices) Contains a heme prosthetic group in a hydrophobic pocket which binds oxygen
391
What is the function of myoglobin?
Stores and transports oxygen within muscles
392
Describe the structure of albumin
Globular protein Highly water-soluble Contains multiple hydrophobic pockets for binding
393
What is the function of albumin?
Maintains osmotic pressure in blood Acts as a buffer for pH and ions Transports fatty acids, hormones (e.g. thyroxine), drugs (e.g. warfarin), bilirubin, etc.
394
Describe the structure of immunoglobulins (antibodies)?
Globular proteins Y-shaped glycoproteins Consist of 4 polypeptide chains, which are connected by disulphide bonds: * 2 heavy chains * 2 light chains
395
What is the function of the cell membrane?
Controls the movement of substances in and out of the cell
396
What is function of the nucleus?
Controls and regulates the activities of the cell (e.g. growth, metabolism, etc.) Carries genetic information (DNA)
397
Describe the structure of the nucleus?
Nucleolus * contains chromatin, proteins and RNA * synthesises ribosomes Chromatin - tangled form of DNA bounded to histones (proteins which act like spools for thread) Nuclear membrane/envelope - a double membrane which controls exchange Nuclear Pores - allow mRNA and ribosomes to pass through Nucleoplasm - full of chromatin, contains chromosomes
398
What is the function of ribosomes?
The site of protein synthesis
399
What is the structure and function of the Rough Endoplasmic Reticulum (RER)?
Function: Folds and processes proteins made on the ribosomes Strcuture: A series of flattened sacs enclosed by a membrane (called cisternae) with ribosomes on the surface
400
What is the structure and function of the Smooth Endoplasmic Reticulum (SER)?
Structure: a network of tubules Function: produces and processes lipids
401
What is the function of mitochondria?
Site of aerobic respiration, producing ATP
402
Describe the structure of mitochondria.
Double Membrane: * Inner Membrane - contains folds/finger-like projections called Cristae which increases surface area * Outer membrane - more permeable, contains proteins called porins to form aqueous channels Matrix - contains the enzymes needed for respiration
403
What is the structure and function of the Golgi apparatus?
Function: processes, modifies and packages proteins and lipids + produces lysosomes Structure: a series of fluid-filled, flattened and curved sacs with vesicles surrounding the edges
404
What are the three components of the cytoskeleton and their functions?
Microfilaments * provides shape and structure to the cell * facilitates cell division and muscle contraction Intermediate filaments * provides structural stability to the cell * anchors organelles in place Microtubules * component of flagella, cilia, and centrioles * facilitate cell division
405
What is the structure and function of lysosomes?
Function: breaks down cellular debris Structure: membrane-bound vesicles which contain lysozyme, a hydrolytic enzyme
406
What is the function of centrioles?
Produce the spindle fibres during cell division
407
What is the function of peroxisomes?
generate and destroy hydrogen peroxide away from the rest of the cell where it would cause damage
408
How are epithelial cells classified?
By their shape (squamous, cuboidal, columnar) and arrangement (simple, stratified, pseudostratified)
409
What are the types of epithelial cells based on shape?
Squamous: Flat and sheet-like Cuboidal: Cube-shaped (equal width, height, depth) Columnar: Tall and column-like (taller than wide)
410
What are the types of epithelial cell arrangements?
Simple: One layer of cells Stratified: Multiple layers Pseudostratified: One layer that appears layered due to cell height variation
411
What are the types of epithelial membranes?
Mucous membrane: * Contains goblet cells, which secrete mucus * Lines external cavities (e.g. respiratory and digestive tract) Serous membrane: * Made up of simple squamous epithelium/ * Lines internal cavities (e.g. pleura)
412
Where is simple squamous epithelium found and what is its function?
Location: alveoli, blood vessels, lymphatics, serous membranes Function: allows substances to pass through by diffusion and filtration + secretes lubricating substances
413
Where is simple cuboidal epithelium found and what is its function?
Location: glandular tissue, kidney tubules Function: absorption and secretion
414
Where is simple columnar epithelium found and what is its function?
Location: bronchi, uterus, digestive tract Function: protection, absorption, secretion of mucus and enzymes Can be ciliated
415
Where is stratified squamous epithelium found and what is its function?
Location: esophagus, mouth, vagina, skin Function: protection against abrasion, pathogens, and chemicals
416
Where is stratified cuboidal epithelium found and what is its function?
Location: sweat glands, salivary glands, mammary glands Function: protection and secretion
417
Where is stratified columnar epithelium found and what is its function?
Location: male urethra, salivary gland ducts Function: protection and secretion
418
Where is pseudostratified columnar epithelium found and what is its function?
Location: trachea, upper respiratory tract, male reproductive ducts Function: mucus secretion and movement Usually ciliated
419
What is transitional epithelium?
Stratified cuboidal cells that flatten (squamous-shaped) when stretched Location: the urinary tract and bladder Function: Stretch readily to accommodate the different volume of liquids
420
What is keratinised epithelium?
Dead epithelial cells which are devoid of the nucleus and cytoplasm and filled with keratin Function: protection against abrasion
421
What are the main types of cell junctions in epithelial tissue?
Tight junctions Adherens Junctions Desmosomes Gap junctions Hemidesmosomes
422
What is the structure and function of tight junctions?
Structure: composed of proteins called claudins and occludins which are arranged into strands that "stitch" adjacent cells together Function: * Forms an impermeable barrier that prevents the passage of molecules and ions through the space between adjacent cells * Maintains the polarity of the cells (apical and basolateral) * Regulates selective permeability (e.g. in the intestines and blood-brain barrier)
423
What is the structure and function of adherens junctions?
Structure: formed by cadherin proteins that link to the actin cytoskeleton inside the cell and extend to the neighbouring cell Function: * Provide mechanical strength * Anchor cells together and allowing them to function as a unit, especially in tissues that experience stress (e.g. epithelial layers)
424
What is the structure and function of desmosomes?
Structure: composed of proteins called cadherins (desmoglein and desmocollin) linked to intermediate filaments, forming strong button-like adhesions between cells Function: * Pin adjacent cells together * Resists mechanical stress, especially in tissues that stretch (e.g. skin, cardiac muscle)
425
What is the structure and function of gap junctions?
Structure: comprised of of connexin proteins that form channels connecting adjacent cells Function: * Allow direct communication between cells by permitting the passage of ions, small molecules and electrical signals * Important for tissue coordination (e.g. in cardiac and smooth muscle)
425
What is the structure and function of hemidesmosomes?
Structure: comprised of integrin proteins (α6β4 integrins) that connect the cell’s intermediate filaments (keratin) to the basement membrane Function: attach epithelial cells to extracellular matrix for structural support
426
What are the components of the extracellular matrix in connective tissue?
Ground substance - a jelly like substance made of glycosaminoglycans Protein fibers: * Collagen * Reticular fibres * Elastic fibres
427
Name some of the different types of cells present in connective tissue and their function.
Fibroblasts - synthesise extracellular matrix components like collagen and ground substance Macrophages - engulf pathogens and debris (immune defence) Plasma cells - produce antibodies Mast cells - release histamine and other mediators during allergic and inflammatory reactions Adipose cells: * Store energy as fat * Provide insulation and cushioning
428
What are the three types of loose connective tissue?
Areolar Adipose Reticular
429
What is the structure and function of areolar connective tissue?
Location: under the skin + around blood vessels, nerves and organs Structure: loose, irregular arrangement of fibres + a variety of cells (fibroblasts, macrophages, mast cells) Function: provides cushioning, support, and flexibility to tissues and organs + immune defence
430
What is the structure and function of adipose tissue?
Location: under the skin + around organs Two types - white adipose tissue + brown adipose tissue Composed mainly of adipocytes Function: * Insulation/maintaining body temperature * Provides cushioning and protection to organs * Stores energy
431
What is dense regular connective tissue?
Location: tendons (connecting muscles to bones) + ligaments (connecting bones at joints) Structure: * Composed of parallel bundles of collagen fibers with fibroblasts squeezed between them * The alignment of the fibres provides tensile strength in one direction Function: Provides strong resistance to pulling forces in one direction
432
What is dense irregular connective tissue?
Location: the dermis of the skin + organs of the digestive tract Structure: * Irregularly arranged collagen fibers that are thicker and interwoven, with fibroblasts scattered throughout * The disordered fiber arrangement allows resistance to tension in multiple directions Function: Provides strength and support in tissues that experience stress from many directions, helping to prevent tearing or damage
433
What are the two types of bone tissue?
Compact (outer layer) Spongy (inner layer containing bone marrow)
434
What are the different types of muscle?
Skeletal * Location: Skeletal muscle * Voluntary contraction * Structure: Long cylindrical fibres, highly striated * Function: Movement + maintenance of posture + heat production Smooth * Location: Walls of hollow organs + blood vessels * Involuntary contraction * Structure: Spindle shaped fibres, nonstriated Function: Peristalsis, contraction + relaxation of vessels and bronchi Cardiac * Location: Heart * Involuntary contraction * Structure: Short branching fibres, finely striated * Function: Circulation of blood
435
What is the biomechanics of skeletal muscle contraction?
1. Nerve impulse arrives at the neuromuscular junction, releasing acetylcholine (ACh) 2. ACh binds to receptors on the muscle cell membrane, triggering an action potential 3. The action potential travels down the T-tubules, stimulating the sarcoplasmic reticulum to release calcium ions (Ca²⁺) 4. Calcium binds to troponin, causing a shift in tropomyosin, which exposes the binding sites on actin 5. Myosin heads bind to actin, forming cross-bridges 6. Using energy from ATP, the myosin head performs a power stroke, pulling actin filaments inward and shortening the sarcomere 7. A new ATP molecule binds to myosin, causing it to detach from actin and reset. 8. This cycle repeats as long as calcium and ATP are present, leading to muscle contraction. 9. When stimulation ends, calcium is pumped back into the sarcoplasmic reticulum, tropomyosin re-covers binding sites, and the muscle relaxes
436
How does smooth muscle contraction occur?
1. Stimulation occurs via ANS or local factors (e.g. stretch) 2. This causes an influx of calcium ions (Ca²⁺) into the smooth muscle cell from the extracellular space or sarcoplasmic reticulum 3. Ca²⁺ binds to a protein called calmodulin 4. The calcium–calmodulin complex activates myosin light chain kinase (MLCK) 5. MLCK phosphorylates myosin, allowing it to bind to actin and initiate contraction 6. Relaxation occurs when calcium levels fall, MLCK activity stops, and myosin is dephosphorylated by myosin phosphatase Contraction is slower and more sustained than in skeletal muscle
437
How is smooth muscle contraction innervated?
Controlled by autonomic nervous system Sympathetic Innervation (e.g. blood vessels, bronchi): 1. Preganglionic neurons release ACh onto nicotinic receptors in sympathetic ganglia 2. Postganglionic neurons release noradrenaline/ norepinephrine 3. Noradrenaline binds to adrenergic receptors on smooth muscle: α₁ receptors (e.g. in blood vessels) → Gq pathway, increases IP₃ → increases Ca²⁺ release → increases contraction β₂ receptors (e.g. in bronchi) → Gs pathway, increases cAMP → inhibits MLCK → relaxation Parasympathetic Innervation (e.g. gut, bladder): 1. Preganglionic neurons release acetylcholine (ACh) onto nicotinic receptors in autonomic ganglia 2. Postganglionic neurons release ACh onto muscarinic M3 receptors on smooth muscle 3. This activates Gq pathway, increases IP₃ , which increases Ca²⁺ release from the sarcoplasmic reticulum 4. Ca²⁺ binds calmodulin → activating MLCK → increased contraction
438
How is cardiac muscle contraction innervated?
Controlled by the autonomic nervous system Sympathetic Innervation: 1. Innervation of the SA node is via the cardiac nerve which releases noradrenaline 2. Noradrenaline binds to β1 adrenergic receptors which activates Gs pathway → increases cAMP which: * Activates PKA → increases Ca²⁺ release → positive inotropy, i.e. greater force of contraction * Speeds up depolarisation → positive chronotropy, i.e. faster rate - more beats per second Parasympathetic Innervation 1. Innervation of the SA node is via the vagus nerve which releases acetylcholine 2. Acetylcholine binds to M2 muscarinic receptors which activates Gi pathway → inhibits adenylate cyclase + opens K+ channels → increases time taken to reach the threshold for an action potential → negative chronotropy, i.e. slower rate
439
What are germ cells?
Reproductive cells (sperm in males, ova in females) Function: * carry genetic information * involved in sexual reproduction * combine during fertilisation to form a zygote
440
What is the structure and function of sperm cells?
Head containing the nucleus with tightly packed DNA - delivers paternal genetic material to the ovum Acrosome - contains enzymes which help penetrate the ovum's outer layers during fertilisation Midpiece is packed with mitochondria - produces ATP to power movement of the tail Tail (flagellum) - long, whip-like structure which propels the sperm forward for mobility through the female reproductive tract
441
What is the structure and function of the ovum?
Nucleus - contains maternal DNA Cytoplasm - rich in nutrients and organelles which supports early development after fertilisation Zona pellucida - glycoprotein layer surrounding the cell membrane which protects the ovum and regulates sperm binding Corona radiata - layer of follicular cells surrounding the zona pellucida which supplies nutrients and supports the ovum's journey through the fallopian tube
442
What is ovulation?
The release of a mature egg from the ovary, triggered by a surge in luteinising hormone (LH), typically around day 14 of the menstrual cycle
443
What is fertilisation?
When a sperm cell penetrates the ovum’s membrane, combining genetic material to form a diploid zygote, usually in the fallopian tube
444
What is embryogenesis?
The process by which a fertilised egg (zygote) develops into an embryo, involving cell division + differentiation and the formation of early tissues and organs
445
What are stem cells?
Undifferentiated cells that can divide and develop into different types of specialised cells
446
What are the different types of stem cells?
Totipotent - can form all cell types including placenta Pluripotent - can form any cell of the body (not placenta) Multipotent - can form limited cell types (e.g. blood cells) Unipotent - can form only one cell type
447
What is haematopoiesis?
The process by which all blood cells are formed from haematopoietic stem cells in the bone marrow
448
What is the structure and function of erythrocytes (red blood cells)?
Structure: biconcave, lacking nuclei, contain haemoglobin Function: transport oxygen
449
What are neutrophils?
A type of WBC that engulf and destroy pathogens through phagocytosis
450
What is the function of eosinophils?
Fight larger parasites Modulate allergic inflammatory responses
451
What is the function of basophils?
Regulate allergic inflammatory response Release histamine
452
What are lymphocytes and their function?
B cells - produce antibodies + assist in activation of T cells T cells - kill pathogens and regulate immunity
453
What are monocytes and their function?
Large white blood cells that circulate in the blood They migrate into tissues and differentiate into macrophages
454
What is the function of macrophages?
Engulf pathogens through phagocytosis
455
How are platelets formed?
Fragmentation of megakaryocytes in the bone marrow
456
Briefly summarise the process of clot formation
When a vessel is injured, platelets adhere and form a plug Clotting factors activate the coagulation cascade, leading to fibrin formation, which stabilises the clot and prevents blood loss
457
Briefly summarise the history of the discovery of DNA and its structure.
Friedrich Miescher (1869): Discovered a new molecule in the nucleus called nuclein (later known as DNA), rich in phosphorus Oswald Avery (1944): Showed that DNA, not protein, is the genetic material using bacterial transformation experiments Erwin Chargaff: * Found base pairing rules: [A]=[T], [G]=[C] * DNA composition varies between species Linus Pauling: Proposed an incorrect DNA model with bases on the outside Rosalind Franklin & Maurice Wilkins: Used X-ray diffraction to reveal DNA's helical structure James Watson & Francis Crick (1953): Proposed the double helix model of DNA (B-form), integrating Franklin's data and Chargaff’s rules.
457
Describe the structure of DNA
A double-stranded right-handed helix, with an ionised sugar-phosphate backbone on the outside and bases on the inside
458
What is the importance of the sugar-phosphate backbone in DNA?
It is ionised, allowing favourable interactions with water, which contributes to DNA's solubility and structural stability
459
What does it mean that DNA strands are antiparallel?
Each strand runs in opposite directions (one 5' to 3', the other 3' to 5'), allowing complementary base pairing
460
What holds the two strands of DNA together?
Hydrogen bonds between complementary base pairs: * A pairs with T (2 H-bonds) * G pairs with C (3 H-bonds)
461
What are the grooves in DNA and why are they important?
DNA has a major groove and a minor groove Proteins typically bind at the major groove where more chemical information is exposed
462
What are the hydrogen bonding patterns in DNA grooves?
a = H-bond acceptor, d = H-bond donor Minor Groove: * CG and GC = a/d/a * AT and TA = a/a Major Groove: * CG = d/a/a * GC = a/a/d * TA = hydrophobic/a/d/a * AT = a/d/a/hydrophobic
463
What is a nucleotide made of?
A phosphate group A pentose sugar (β-D-2’-deoxyribose) A nitrogenous base (Adenine, Guanine, Cytosine, Thymine)
464
What are the different types of base in DNA?
Purines: * Adenine (A) + Guanine (G) * Two rings, bond via N9 Pyrimidines: * Cytosine (C) + Thymine (T) * One ring, bond via N1
465
Why does DNA have a consistent diameter of ~2 nm?
Because purines (larger) always pair with pyrimidines (smaller), maintaining uniform spacing between the strands
466
What stabilises the helical structure of DNA besides hydrogen bonds?
π–π stacking (van der Waals interactions) between adjacent base pairs
467
Why is DNA polyanionic and what are the consequences?
The phosphodiester backbone has a pKa ~1, making DNA negatively charged at physiological pH This gives it water solubility and allows interaction with counter-ions like Na⁺, K⁺, Mg²⁺, spermine, and spermidine
468
What role does water play in DNA structure?
Water forms a spine of hydration in the grooves and stabilises the DNA through hydrophilic interactions with the backbone
469
Describe the tertiary structure of DNA.
Supercoiling, which compacts the DNA and reduces strain during replication or transcription Looping and scaffolding within the chromosome Association with histone proteins forming nucleosomes and further folding into chromatin
470
What are introns and exons?
Introns are non-coding regions of DNA (23% of human genome) Exons are coding regions of DNA (1.1% of human genome)
471
How does the structure of DNA relate to its role in inheritance?
Role: store of information Structure: DNA's sequence of nitrogenous bases (A, T, C, G) encodes genetic instructions, Link: allowing it to function as a stable, long-term repository of information Role: replication mechanism Structure: The complementary base pairing (A with T, C with G) enables semi-conservative replication, where each strand serves as a template for a new one. Link: This ensures that genetic information is accurately copied and passed from one generation to the next during cell division and reproduction.
472
What are the differences between RNA and DNA?
DNA * Deoxyribose sugar * Thymine nucleobase * Double stranded * Larger in size * More stable * Chargaff rule applies RNA * Ribose sugar * Uracil nucleobase (doesn't have a methyl group) * Single stranded * Smaller in size * Less stable * Chargaff rule does not apply
473
Why is RNA's lower stability important?
RNA is less stable than DNA Because of the hydroxyl group, under slightly basic conditions the phosphate can be attacked leading to cleavage of the backbone This is important because it means that RNA can be broken down easily allowing for gene expression to be regulated
474
Describe the structure of RNA
Single-stranded polymer of nucleotides Can fold into various shapes (e.g. hairpins, loops, bulges, etc.), through regional/local complementary bases pairing, which facilitates its diverse functions in the cell
475
What does semi-conservative replication mean?
Two identical copies of the original DNA molecule is produced, each of which contains: one of the original strand and one newly synthesised strand Each newly formed DNA molecule consists of one original (parental) strand and one newly synthesised strand
476
What happens during the initiation stage of DNA replication?
Helicase unwinds the DNA helix by breaking hydrogen bonds between bases, forming a replication fork. Single-strand binding proteins prevent rejoining of strands. Topoisomerase relieves supercoiling by creating temporary cuts in DNA
476
How does Meselson and Stahl’s experiment (1958) show that DNA replication is semi-conservative?
E. coli was grown in heavy nitrogen (N-15) and then transferred to light nitrogen (N-14) DNA samples were centrifuged using a CsCl density gradient to separate DNA based on density Generation 0: * DNA formed a single heavy band (all N-15). Generation 1: * DNA formed a single intermediate band, showing hybrid DNA (N-15/N-14) * Ruling out conservative replication Generation 2: * Two bands appeared — one intermediate and one light * Supporting semi-conservative replication, where each new DNA molecule contains one old and one new strand.
477
What is the role of primase in DNA replication?
Primase synthesises a short RNA primer (8–10 nucleotides) to provide a 3'-OH group for DNA polymerase. This is important because DNA polymerase cannot initiate synthesis without a primer as it can only add nucleotides to an existing strand
478
How does elongation occur during DNA replication?
DNA polymerase extends from the primer, adding nucleotides in the 5’ → 3’ direction The leading strand is synthesised continuously toward the replication fork The lagging strand is synthesised in Okazaki fragments away from the fork, each needing a new primer DNA polymerase also proof-reads as it moves along by removing incorrectly matching nucleotides that are accidentally added
479
What happens to the RNA primers during DNA replication?
The RNA primers are removed by the enzyme exonuclease and the resultant gaps are filled with nucleotides by DNA polymerase I
480
What is the role of DNA ligase in replication?
DNA ligase joins the Okazaki fragments on the lagging strand, creating a continuous strand of DNA
481
Describe the process of DNA replication
1. Helicase unwinds the DNA helix by breaking hydrogen bonds between bases, forming a replication fork. 2. Single-strand binding proteins prevent the strands from rejoining of strands and topoisomerase relieves tensions by creating temporary cuts in DNA, preventing supercoiling 3. Primase synthesises a short RNA primer (8-10 nucleotides long), providing a starting point for DNA synthesis 4. DNA polymerase attaches to the primer and starts forming the new strands 5. One strand (leading strand) is continuous, in the same direction as the replication fork 6. The other strand (lagging strand) is made in small pieces (Okazaki fragments), requiring a new primer for each, moving away from the replication fork 7. Exonuclease removes RNA primers and the resultant gaps are filled by DNA polymerase I 8. DNA ligase joins the Okazaki fragments together 9. Once both strands are fully replicated, the replication process ends, resulting in two identical DNA molecules, each containing one old strand and one new strand
482
How are leading and lagging strand synthesis co-ordinated, despite synthesis proceeding in different directions topologically?
The synthesis of the leading and lagging strands is coordinated by the replisome, using the trombone model The lagging strand forms a loop, allowing its polymerase to stay near the leading strand polymerase This loop enables discontinuous synthesis of Okazaki fragments in the 5' → 3' direction The loop grows and releases with each fragment, maintaining synchronised replication of both strands at the replication fork
483
Activity of topoisomerase is both essential and toxic. Why?
Topoisomerase activity is essential because it resolves the topological stress that arises during DNA replication and transcription. When helicase unwinds the DNA double helix, it creates tension in the DNA molecule. This tension leads to supercoiling (over-winding) ahead of the fork. Topoisomerases prevent this by creating temporary cuts and rejoining DNA strands to relieve supercoiling and untangle knots. However, topoisomerase activity can also be toxic. If a topoisomerase introduces breaks in the DNA but fails to reseal them due to malfunction or interference, it leaves behind persistent DNA breaks. These breaks can lead to genome instability, mutations, or cell death.
484
What can prevent topoisomerase toxicity?
The enzyme DNA ligase can help prevent toxicity by resealing nicks and breaks left by topoisomerase activity, ensuring that transient DNA cleavage events do not become permanent
485
How do polymerase inhibitors target DNA replication to treat cancer?
E.g. Gemcitabine, Mitomycine C Polymerase inhibitors block DNA polymerase, the enzyme responsible for synthesising new DNA strands. By doing so, they: * Prevent the addition of nucleotides to the growing DNA chain * Cause DNA damage or incomplete replication, triggering cell death
486
How do bulky adducts interfere with DNA replication to treat cancer?
E.g. Cisplatin, Mitomycine C Large chemical groups that attach to DNA strands, preventing proper base pairing and blocking DNA polymerase progression
487
What is Polymerase Chain Reaction (PCR) and how does it work?
Polymerase Chain Reaction (PCR) is a laboratory technique used to amplify specific DNA sequences, making them easier to study Process: 1. Denaturation: The DNA template is heated to around 94-98°C, causing the double-stranded DNA to separate into two single strands. 2. Annealing: The reaction temperature is lowered to 50-65°C, allowing short DNA primers to bind to the complementary sequences on the single-stranded DNA. 3. Extension: DNA polymerase adds nucleotides to the primers, extending the DNA strand in the 5’ to 3’ direction, at around 75-80°C. 4. These steps are repeated for 20-40 cycles, resulting in the exponential amplification of the target DNA sequence.
488
How are chromosomes characterised?
Size Pattern of G-bands Location of centromere: * Metacentric = centromere in the middle, arms of equal length * Submetacentric = centromere slightly off-center * Acrocentric = centromere close to one end
489
What is human karyotype?
The human karyotype is 46 chromosomes, including 22 pairs of autosomes and 1 pair of sex chromosomes
490
What is the difference between sister chromatids and homologous chromosomes
Sister chromatids: * Identical copies of a single chromosome, formed after DNA replication in the S phase * Held together at the centromere * Exact genetic replicas * Present only during and after DNA replication - separate during anaphase of mitosis or anaphase II of meiosis Homologous chromosomes: * A pair of chromosomes, one from each parent * Similar in size, shape and genetic content but not identical * Do not share a centromere * Separate during meiosis I * Present in diploid cells
491
What is a locus?
The specific physical location or position of a gene on a chromosome Each gene is located at a specific locus on a chromosome, which is consistent across individuals of the same species E.g. the gene for eye colour in humans is located at a particular locus on chromosome 15
492
What is Mendel's Law of Segregation?
Every inherited characteristic/trait is defined by a gene pair Due to the meiosis process, each offspring receives one allele from each parent to make a gene pair
493
What is Mendel's Law of Independent Assortment?
Genes for different traits are sorted separately from one another so that the inheritance of one trait is not dependent on the inheritance of another
494
What is Mendel's Law of Dominance?
An organism with alternate forms of a gene will express the form that is dominant. (One gene is dominant and one is recessive)
495
What is the difference between monogenic and polygenic disorders?
Monogenic disorders are caused by a mutation in a single gene (E.g. Cystic fibrosis, Sickle cell anaemia, Huntington's disease) Polygenic disorders are caused by multiple gene mutations and environmental factors (E.g. Type II diabetes, Rheumatoid arthritis, Schizophrenia)
496
What are the different modes of inheritance for monogenic disorders?
Autosomal recessive Autosomal dominant X-linked recessive X-linked dominant Mitochondrial
497
What is an autosomal recessive disease?
Both copies of the gene (one from each parent) must be mutated for the disorder to occur If an individual inherits only one mutated allele, they are a carrier and typically do not show symptoms Affects males and females equally E.g. Cystic Fibrosis, Sickle Cell Anaemia
498
What is an autosomal dominant diseas?
Only one copy of the mutated gene, inherited from either parent, is sufficient to cause the disorder Affects males and females equally E.g. Huntington's disease
499
What is an X-linked recessive disease?
The mutated gene is located on the X chromosome Two copies of the mutated gene (one on each X chromosome) are needed for females to express the disorder Males, having only one X chromosome, will express the disorder if they inherit the mutated gene Affected fathers cannot pass the disorder to their sons but will pass it to daughters E.g. Hemophilia A and B, Red-green color blindness
500
What is an X-linked dominant disease?
The mutated gene is located on the X chromosome, and only one copy of the mutated gene is needed to cause the disorder Affects males and females equally
501
What is a mitochondrial disease?
Passed exclusively from mothers to their offspring, as only the egg contributes mitochondria to the zygote Affects males and females equally
502
How do we assess the contribution made by genetics vs environment for a given disorder?
Twin Studies Compare the concordance rates (likelihood of both individuals having the disorder) between identical twins (monozygotic - sharing 100% of their DNA) and fraternal twins (dizygotic - sharing 50% of their DNA) If identical twins have a much higher concordance rate than fraternal twins, the disorder is likely to have a strong genetic component If concordance rates are similar, environmental factors may play a larger role
503
What are some things to look out for during pedigree analysis?
If a generation is skipped (i.e. unaffected parents have affected offspring), dominant inheritance is unlikely → likely recessive If male offspring do not inherit the condition from an affected mother, the condition is unlikely to be X-linked Autosomal dominant traits typically appear in every generation Consistent transmission from fathers to sons suggests autosomal, not X-linked inheritance
504
What is the link between malaria and sickle cell anaemia?
Malaria is caused by a parasite which infects the red blood cells. Individuals who carry the mutated β-globin gene are protected against malaria because the parasite is less able to survive and reproduce in sickle-shaped red blood cells