Cardiovascular Flashcards

1
Q

What is the normal value for end systolic volume?

A

75 mL

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

What is the normal value for stroke volume?

A

100 mL or less

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

What is the normal value for end diastolic pressure?

A

15 mmHg

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

What is the normal value for early diastolic pressure?

A

5 mmHg

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

Why does the gradient of the diastolic left ventricular pressure-volume curve increase more steeply with increase left ventricular volume?

A

Ventricle gets filled to its physical capacity > exponentially greater increase in pressure

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

What is the Frank-Starling relationship?

A

Greater pre-load on cardiac muscle fibres prior to contraction > increase in force of contraction
Within limits, more the heart fills with blood during diastole, the greater the force of contraction during systole
Pre-load is proportional to end diastolic volume
Therefore, the greater the end diastolic volume, the more forceful the contraction

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

What determines end diastolic volume?

A

Duration of ventricular diastole

Venous return

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

Why can the duration of ventricular diastole be short?

A

Heart rate increase > less filling time

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

What will happen to contractility if there is acidosis?

A

Decreases

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

What will happen to contractility if there is parasympathetic nerve deactivation?

A

Increases to small extent, as parasympathetic nervous system more important in heart rate and atrioventricular node firing

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

What will happen to contractility if there is caffeine intake?

A

Increases to small extent

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

What will happen to contractility if there is hypercapnia?

A

Decreases

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

Why does left ventricular pressure never reach maximal capacity during systole?

A

Aortic valve releases pressure when left ventricular pressure is greater than in aorta
In effect, aorta acts as release valve

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

Describe the blood distribution in the circulation

A
Veins = 65%
Arteries = 13%
Capillaries = 5%
Lungs = 10%
Heart = 7%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Why is atrial pressure more sensitive to changes in volume than venous pressure?

A

Veins more compliant > taking blood out of veins will have less effect on pressure change

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

What is mean circulatory filling pressure?

A

Eventual average systemic pressure if heart stopped and blood settled in vessels

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

What is mean circulatory filling pressure determined by?

A

Volume of blood

Compliance of vessels

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

What effect would an increase in cardiac output have on venous pressure?

A

Decrease venous pressure

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

What effect would an increase in total peripheral resistance have on the volume of blood in the arterial and venous systems?

A

No effect

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

What effect would decreasing venous pressure have on cardiac output?

A

Decrease cardiac output due to reduced filling pressure

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

What is central venous pressure?

A

Pressure in great veins just outside heart

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

Why might central venous pressure rise?

A

Systemic venous congestion during right heart failure

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

Why might central venous pressure fall?

A
Shock = systemic vasodilation
Venous return poor
- Blood loss
- Upright posture
- Inadequate muscle and respiratory pumps
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What effect would an increase in contractility have on cardiac output and venous pressure?

A

Increase cardiac output

Decrease venous pressure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What substances produced by the endothelium alter muscle contraction?

A

Nitric oxide = vasodilator
Endothelin = vasoconstrictor
Prostacyclin = vasodilator

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is antagonist potency mostly determined by?

A

Potency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Which antagonists stop binding?

A

Competitive antagonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Which antagonists stop activation?

A

Non-competitive antagonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What does propranolol do?

A

Competitive antagonist of beta-adrenoceptors = beta-blocker

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the factors affecting drug distribution?

A

Molecular size
Ability to bind plasma proteins
Lipid solubility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is volume of distribution?

A

Volume of fluid in which amount of drug in body would need to be uniformly distributed to produced observed concentration in blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

How do you work out the volume of distribution?

A

Total amount of drug in body/drug blood plasma concentration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What does a small volume of distribution indicate?

A

Drug binds strongly to plasma proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What does a large volume of distribution indicate?

A

Drug take up by cells and binds to tissues > greater distribution

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

How does pH affect lipid solubility and thus the passage of drugs?

A

Drugs more lipid-soluble if uncharged

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

At which pH are acidic drugs more lipid soluble?

A

Low pH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

At which pH are basic drugs more lipid soluble?

A

High pH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

How is acetylcholine synthesised?

A
  1. Choline transported into cell by choline carrier
  2. Choline + acetyl-CoA > acetylchole
  3. Acetylcholine loaded into synaptic vessel
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

How is noradrenaline synthesised?

A
  1. Tyrosine transported into cell
  2. Converted to L-DOPA
  3. Converted to dopamine
  4. Transported into synpatic vesicle
  5. Converted to noradrenaline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

How is adrenaline synthesised?

A

Noradrenaline converted to adrenaline in synpatic vesicle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is co-transmission?

A

Autonomic nerves often release more than one neurotransmitter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

How is acetylcholine inactivated?

A

Inactivated and degraded by acetylocholinesterase on effector tissue surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

How is noradrenaline inactivated?

A

Uptake 1 = reuptake via high affinity receptor

Uptake 2 = taken up via low affinity receptor by post-synaptic tissue > degraded

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What is phenylephrine?

A

Alpha-adrenergic receptor agonist

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

How does adrenaline increase blood pressure?

A

Alpha and beta-adrenergic receptor agonist > vasoconstriction and tachycardia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

How do anitcholinesterases work?

A

Decreased breakdown of acetylcholine > increased levels of acetylcholine in synaptic cleft > increased acetylcholine action

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is myasthenia gravis?

A

Autoimmune condition where Abs produced against nicotinic acetylcholine receptors (on neuromuscular junctions)
Abs bind to receptors
- Receptor internalisation and degradation
- Activation of complement > destruction of neuromuscular junction synapse architecture

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

How can myasthenia gravis be treated?

A

Use anticholinesterases
Effective in early stages
Decreased breakdown of acetylcholine > more in synpase > more chance of binding to remaining receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What are the effects of muscarinic receptor agonism?

A
Salivation
Lacrimation
Urination
Defecation
Sweating
Bradycardia
Vasodilation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are indirectly acting sympathomimetics?

A

Taken up by neuron > displace noradrenaline from synaptic vesicles > more noradrenaline displaced into synapse

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are some stimuli that induce histamine release?

A
Ag via IgE
Complement fragments C3a and C5a
Neuropeptides
Cytokines and chemokines
Bacterial components
Physical trauma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What is the triple response to histamine?

A

Reddening
Wheal
Flare

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What causes reddening in response to histamine?

A

Vasodilation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What causes a wheal in response to histamine?

A

Increased vessel permeability > local oedema

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What causes a flare in response to histamine?

A

Spreading response through sensory fibres

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What type of histamine receptors do classic antihistamines act on?

A

H1 receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Why do H1 receptor antagonists cause sleepiness?

A

Antagonise H1 receptors in cerebral cortex > reduce neuronal excitation
Bloch acetylcholine receptors > excess acetylcholine > drowsiness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What was the problem with using terfenadine as a non-sedative H1 receptor antagonist?

A

Cause rare, sudden ventricular arrhythmia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

From which cells in the gastrointestinal tract is histamine released?

A

Enterochromaffin-like (ECL) cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What two things does bradykinin mediate?

A

Pain

Inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What triggers release of bradykining?

A

Pre-kallikrein cleaved to kallikrein by activated factor XII
Kallikrein becomes kininogen
Kininogen activated to bradykinin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What are the actions of bradykinin?

A
Potent endothelium-dependent vasodilator
Contraction of non-vascular smooth muscle in bronchus and gut
Increase of vascular permeability
Sensitises pain nerve endings
Natriuresis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the effects of bradykinin on blood pressure and how?

A

Decreases blood pressure through

  • Vasodilation
  • Natriuresis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

How do ACE inhibitors cause cough?

A

ACE normally breaks down bradykinin

ACE blocked > build up of bradykinin > bronchoconstriction > dry cough

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What are the physiological roles of nitric oxide?

A

Flow-dependent vasodilation
Inhibits platelet adhesion and aggregation
Neurotransmitter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

How is arachidonic acid stored?

A

Esterified and stored in membrane phospholipids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

How is arachidonic acid liberated from membranes?

A

Increase in intracellular Ca > hospholipase A2 activated > breaks bond between arachidonic acid and phospholipid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is the difference in expression between COX1 and COX2?

A

COX1 expression constitutive
COX2 expression inducible
- Expressed in response to inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Which cells express high levels of lipoxygenase?

A

Inflammatory cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What are the stable prostaglandins?

A

PGE2
PGD2
PGF2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are the functions of PGE2?

A

Vasodilation
Pyrogenesis
- In hypothalamus, increases temperature set-point
Angiogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What are the indications for non-steroidal anti-inflammatory drugs (NSAIDs)?

A

Anti-inflammatory
Analgesia
Antipyretic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Why is paracetamol often preferred over aspirin as an anti-pyretic?

A

Highly selective for COX2

Won’t cause gastric irritation/ulceration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Describe how bradykinin and PGE2 work in synergy to intensify pain

A

PGE2 > vasodilation > increased blood flow
Bradykinin > vascular leakiness
PGE2 increases sensitivity to pain caused by bradykinin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

How is the set point raised during fever originating from inflammation?

A

Inflammation > macrophage activation > cytokines > PGE2 synthesis in hypothalamus > increase temperature set point

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What are the gastro-protective roles of PGE2?

A

Promote blood flow
Increase mucus secretion
Reduce gastric acid secretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

How do NSAIDs cause gastric irritation?

A

Inhibit PGE2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Where is prostacyclin (PGI2) produced?

A

Endothelial cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the function of PGI2?

A

Reduce platelet activation

Vasodilation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Where is TXA2 produced?

A

Platelets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What is the function of TXA2?

A

Increase platelet activation

Vasoconstriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

How do PGI2 and TXA2 differ in function?

A

Oppose actions of each other

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

How does aspirin differ to other NSAIDs?

A

Irreversibly inactivates COX

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What effect can aspirin have on COX in platelets?

A

Irreversibly inactivate COX in platelets

Since platelets have no nucleus, won’t resynthesise COX for entire lifespan of 8 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What effect does inactivation of COX in platelets by aspirin have?

A

TXA2 levels reduced more than PGI2 levels > endothelium-mediated vasodilation and decreased platelet activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are aspirin-triggered lipoxins?

A

Acetylation of COX2 alters its activity > produces aspirin-triggered lipoxins
Active

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What is the suggested mechanism for cardio-protection from intake of fish oil?

A

Fish oil increases synthesis of PGI3

PGI3 has prostacyclin-like activity > suggested that it decreases incidence of coronary artery disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What types of cells is 5-lipoxygenase activity only seen in?

A

Inflammatory cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What is the link between leukotriene activity and asthma?

A

Leukotrienes potent bronchoconstrictors, also causing bronchospasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What is the link between leukotriene and hayfever?

A

Allergic response > leukotriene release > nasal tissue oedema > nasal congestion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

How can leukotriene action be blocked?

A

Leukotriene receptor antagonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Why do coaguation factors normally not cause thrombosis in vessels?

A

Circulate as pro-enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are the three phases of haemostasis?

A

Primary haemostasis
Secondary hamostasis
Fibrinolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What happens in primary haemostasis?

A

Vasoconstriction
Platelet adhesion
Platelet aggregation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What happens in secondary haemostasis?

A

Activation of coagulation factors

Formation of fibrin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What happens in fibrinolysis?

A

Activation of fibrinolysis

Lysis of clot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What makes up Virchow’s triad?

A

Endothelial injury
Abnormal blood flow
Hypercoagulability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What is required to trigger the coagulation system?

A

Tissue factor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What molecules can inactivate thrombin?

A

Protein C

Anti-thrombin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What is the pharmacological mechanism of warfarin?

A

Inhibits reduction of vitamin K > no activation of vitamin K-dependent factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What is the pharmacological mechanism of heparin?

A

Binds antithrombin > doesn’t cleave thrombin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is the difference between risk and rate?

A
Risk = probability of disease occurring in a disease-free population during a specified time period
Rate = probability of disease occurring in disease-free population during sum of individual follow-up periods
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What is risk a measure of?

A

Incidence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What is person-time?

A

Makes explicit time population at risk actually spent being at risk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What are the benefits of using person-time?

A

Adjusts for loss of participants to follow

Risk can potentially underestimate true incidence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is hazard, in relation to rate?

A

Type of rate that’s continuously updated as longitudinal study progresses
Rate that applies to an exact point in time = instantaneous rate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

If absolute risk is isolated, how is it possible to provide an indication of association between a risk and an outcome?

A

Generate relative risk = risk ratio

Relative risk = risk among exposed/risk among unexposed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is attributable risk?

A

Indicates absolute magnitude of change in risk/rate of outcome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

How is attributable risk calculated?

A

Risk among exposed - risk among unexposed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What does attributable risk percentage represent?

A

Percentage of incident disease among exposed people that was due to exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is population attributable risk?

A

Additional/excess risk of outcome in population, due to exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

How do you calculate population attributable risk?

A

Risk in whole population - risk among unexposed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What does the population attributable risk percentage represent?

A

Proportion of incident disease among the whole population due to exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What is the population attributable risk percentage also known as?

A

Preventable fraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

When do chemoreceptors start to act in the context of blood pressure control?

A

When blood pressure falls below 60 mmHg

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Where are baroreceptors located?

A

Carotid sinus
Aortic arch
Pre-glomerular arterioles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is the carotid sinus?

A

Thin spot of wall in internal carotid artery
More compliant
More sensitive to changes in stretch due to change in pressure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Where are chemoreceptors located?

A

Carotid and aortic bodies outside arteries

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What do chemoreceptors respond to?

A

Low flow
Low O2
High CO2
Low pH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What components of the cardiovascular system are responsive to parasympathetic signals?

A

Heart rate

Conduction speed at the sinoatrial node

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How can a stroke occur from an atrial thrombus?

A

Atria starts beating at sinus rhythm, may have to pump against higher pressure due to decreased ventricular compliance
Thrombus may dislodge and travel up to brain
Cerebral embolism > stroke

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

How do systolic and diastolic blood pressures vary with age?

A

Systolic pressure increases

Diastolic pressure increases until 60, then decreases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

Why does pulse pressure increase with age?

A

Decreasing compliance > increased systolic and decreased diastolic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

Why does compliance decrease with age?

A

In arteries

  • More collagen
  • Less elastin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

How does blood pressure vary with sex?

A

Males > females

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

How does blood pressure vary with body size?

A

Larger body size > higher blood pressure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the diurnal variation in blood pressure?

A

Lower at night

Less variability at night

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What is the population paradox?

A

More deaths occur in people at moderate risk (large group) than in people at high risk (small group)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are social permissions?

A

What doctors are allowed to do, that others can’t

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

Why are explicit ethical principles needed?

A

To address vulnerability inherent in patient’s position, by affirming particular ethical obligations of doctors to protect and respect patients
Provide counter to negative culture of medicine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are the basic ethical principles?

A
Beneficence
Non-maleficence
Respect for autonomy
Respect for privacy
Justice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is the difference between moral confusion and moral distress?

A

Moral confusion = feeling unsure what’s right thing to do, and not knowing how to resolve it
Moral distress = feeling unable to do what you think is right and feeling compelled to do what you think is wrong

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What are the targets for parasymathetic control of heart rate?

A

Sinoatrial and atrioventricular nodes via acetylcholine action on muscarinic cholinergic receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What is the effect of muscarinic receptor blockade on heart rate in healthy young individuals?

A

Increases it

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are the targets for sympathetic control of heart rate?

A

Sinoatrial node
Conducting pacemaker tissue
Myocardial cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is the effect of sympathetic release of noradrenaline and adrenaline on the heart?

A

Bind to beta-adrenoceptors > directly increase heart rate and contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What are the phases of the sinoatrial node pacemaker action potential?

A
Phase 0
- Depolarisation
- Ca in
Phase 3
- Repolarisation
- K out
Phase 4
- Spontaneous depolarisation
- Na and Ca in
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Why don’t the pacemaker cells have a stable resting membrane potential?

A

Leaky funny current Na channel

Uses Ca for depolarisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

Describe the mechanism for parasymathetic slowing of the sinoatrial node

A

Acetylcholine acts on muscarinic receptors on sinoatrial node > decrease in cAMP > K channels open > K efflux > slows Na and Ca fluxes > slower phase 4 > takes longer to reach threshold potential > slows rate of conduction of atrioventricular node

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

Describe the mechanism of sympathetic acceleration of the sinoatrial node

A

Noradrenaline and adrenaline act on beta1-adrenoceptors on sinoatrial node > increase in cAMP > opening of Ca channels > increased slope of phase 4 at sinoatrial and atrioventricular nodes > increased rate of firing from sinoatrial node and more rapid conduction to atrioventricular node

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the phases of the ventricular pacemaker action potential?

A
Phase 0
- Depolarisation
- Na in
Phase 1
- Rapid repolarisation
- K out
Phase 2
- Plateau
- Ca in
- K out
Phase 3
- Repolarisation
- K out
Phase 4
- Stable membrane potential
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What are the mechanisms underlying dysrhythmias?

A

Altered impulse formation
Altered impulse conduction
Triggered activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What is altered impulse formation?

A

Abnormal generation of action potentials at sites other than sinoatrial node

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What is altered impulse conduction?

A

Conduction block

Re-entry > extra beats

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What triggers activity in dysrhythmias?

A

Early/late after-depolarisations due to excess sympathetic activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What is the molecular mechanism of class 1 anti-dysrhythmics?

A

Na channel block > reduces phase 0 slope and peak of ventricular action potential

  • 1a = moderate block
  • 1b = weak block
  • 1c = strong block
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What is the molecular mechanism of class 2 anti-dysrhythmics?

A

Beta-adrenoceptor antagonism > decrease rate and conduction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What is the molecular mechanism of class 3 anti-dysrhythmics?

A

K channel blockade > delay phase 3 of ventricular action potential

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What is the molecular mechanism of class 4 anti-dysrhythmics?

A

Ca channel blockade > reduces rate and conduction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What effects do beta-adrenoceptor antagonists have on Purkinje fibres?

A

Membrane stabilising effects - local anaesthetic-type action

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What are patients on K channel inhibitors at an increased risk of experiencing and why?

A

K channel inhibition > K takes longer to leave cells > prolonged repolarisation time > prolonged action potential > more chance of triggered events

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What is the drug class of lignocaine?

A

Class 1b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What do the side effects of lignocaine depend on?

A

Concentration of drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are the side effects of lignocaine, with increasing concentrations?

A
Lip and tongue numbness
Light headedness
Visual disturbance
Muscular twitching
Convulsions
Coma
Respiratory arrest
Cardiovascular depression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Other than as an anti-dysrhythmic, what else is lignocaine used as?

A

Anti-dysrhthmic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are the adverse effects of beta-adrenoceptor antagonists?

A
Bradycardia
Reduced exercise capacity
Hypotension
Atrioventricular conduction block
Bronchoconstriction
Hypoglycaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

Other than as an anti-dysrhythmic, what else are beta-adrenoceptor antagonists used in?

A

Hypertension
Heart failure
Angina
Anxiety

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the drug class of amiodarone?

A

K channel inhibitor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are the side effects of amiodarone?

A

Reversible photosensitisation
Skin discolouration
Hypothyroidism
Pulmonary fibrosis with long term use

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What is the dug class of verapamil?

A

Ca channel blocker

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What are the side effects of calcium channel blockers?

A
Facial flushing
Peripheral oedema
Dizziness
Bradycardia
Headache
Nausea
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

Other than as an anti-dysrhythmic, where else are calcium channel blockers used?

A

Hypertension

Angina

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What are the goals for the treatment of hypertension?

A

Reduce blood pressure to <140/85 mmHg

If diabetes/renal disease present, reduce blood pressure to <130/80 mmHg

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What are the controlled variables contributing to blood pressure?

A

Heart rate
Stroke volume
Total peripheral resistance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What are the effects of angiotensin II?

A

Vasoconstriction
Aldosterone > salt and water retention
Cell growth
Increased sympathetic activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What is the mechanism of action of ACE inhibitors?

A
Block angiotensin I and angiotensin II conversion
- Reduce vascular tone > vasodilation
- Reduce aldosterone productionn
- Reduce cardiac hypertrophy
Prevent breakdown of bradykinin
- Increase action of bradykinin
   - Vasodilation
   - Cardioprotection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What are the adverse effects associated with ACE inhibitors?

A
1st dose hypotension
Dry cough
Loss of taste
Hyperkalaemia
Acute renal failure
Pruritis
Foetal malformations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What are the types of angiotensin receptor antagonists?

A

AT1 antagonists - clinically useful

AT2 antagonists - not clinically useful

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What do AT1 antagonists do?

A

Reduce vasoconstriction
Reduce aldosterone release
Reduce cardiac hypertrophy
Reduce sympathetic activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What are the adverse effects of angiotensin receptor antagonists?

A

Hyperkalaemia
Headache
Dizziness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

How do beta-adrenoceptor antagonists decrease blood pressure?

A

Reduce cardiac output
- Reduce heart rate
- Reduce contractility
Reduce renin release

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

How do different beta-adrenoceptor antagonists vary?

A

Selectivity
Intrinsic sympathomimetic activity
Lipid solubility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

What are the adverse effects of using beta-adrenoceptor antagonists?

A

Cold extremities
Fatigue
Dreams, insomnia
Bronchoconstriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

When is the use of beta-adrenoceptor antagonists contraindicated?

A
Diabetes
Asthma
Take care with
- Heart failure
- Metabolic syndrome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What type of calcium channels to calcium channel blockers block?

A

Voltage-gated L-type Ca channels in

  • Vasculature
  • Myocardium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What are the adverse effects of calcium channel blockers?

A

Oedema
Flushing
Headache
Bradycardia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

How do thiuzide diuretics act to lower blood pressure?

A

Inhibit Na/Cl co-transporter in distal tubule > decrease Na and Cl reabsorption > increases Na and water excretion from kidney and K from collecting ducts > lower blood volume and reduced blood pressure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What are the adverse effects of thiuzide diuretics?

A

Hypokalaemia
Gout
Hyperglycaemia
Allergic reaction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What is the suffix of ACE inhibitors?

A

-pril

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What is the suffix of angiotensin receptor antagonists?

A

-sartan

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What is the suffix for beta-adrenoceptor antagonists?

A

-olol

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What is the disadvantage of tetracycline use?

A

Short-half life

Must administer 4 times/day

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

How is the disadvantage of tetracycline overcome?

A

Semi-synthetics; eg: doxycycline

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

Why was penicillin-G replaced by penicillin-V?

A

Penicillin-G acid labile
- Must be given IV
Penicillin-V acid stable
- Available orally

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What are the advantages of dicloxacillin and flucloxacillin over methicillin?

A

Less toxic

Capable of oral administration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What are the principles of selective toxicity?

A

Antibiotics should target organism but not damage host

187
Q

What antibiotic class targets the bacterial cell wall?

A

Beta-lactams

Glycopeptides

188
Q

What antibiotic class targets bacterial ribosomes?

A

Aminoglycosides

Chloramphenicol

189
Q

What antibiotic class targets bacterial nucleic acids?

A

Rifamycins

Quinolones

190
Q

What antibiotic class targets bacterial cell membranes?

A

Polymyxins

Polyenes - for fungi

191
Q

What antibiotic class targets bacterial folic acid synthesis?

A

Sulphonamides

Trimethoprims

192
Q

What is a cross-sectional study?

A

Examines sample of population selected and information obtained at 1 point/period in time
No follow up of subjects
Each subject contributes data only once

193
Q

How is data collected in cross-sectional studies?

A

Questionnaires
Examinations
Investigations
Prevalence is especially looked at

194
Q

What are the advantages of cross-sectional studies?

A

Relatively cheap

Easy

195
Q

What are the disadvantages of cross-sectional studies?

A

Often need for representative sample, as study mainly descriptive
Weak evidence of causality because no explicit data on temporal relationships

196
Q

What is a case-control study?

A

Comparison of previous exposure status between cases and controls
Looks at effect of exposure on outcome of interest

197
Q

What is the purpose of matching during a case-control study?

A

Matched on bases of confounders to reduce potential for bias

198
Q

What are the advantages of case-control studies?

A

Gives explicit knowledge about temporal relationship between exposure and outcome
Useful for studying rare outcomes

199
Q

Why is relative risk unmeasurable in case-control studies?

A

Relative risk = measure of incidence

Case-control studies retrospective > no data on incidence

200
Q

What is used as an approximation of relative risk in case-control studies?

A

Odds ratio

201
Q

What are cohort studies?

A

Longitudinal studies done with follow-up of subjects to collect incidence data
Compares outcomes between exposed and not exposed to risk factor > relative risks derived

202
Q

What are the advantages of cohort studies?

A

Explicit and detailed knowledge about temporal relationship between exposure and outcome
Can include multiple exposures and outcomes
Cohorts can be established as part of routine clinical care

203
Q

What are the disadvantages of cohort studies?

A

Difficult to study rare outcomes

Not cheap nor easy

204
Q

Why is a retrospective cohort study still considered a longitudinal study?

A

Researchers begin study at time where cohort has already been established and data available bout exposure in past

205
Q

What is bias?

A

Unintentional error, due to systematic difference between/among groups > under-/overestimation of true results

206
Q

What are the two main types of bias?

A

Selection

Information (measurement)

207
Q

What is selection bias?

A

Systematic difference in characteristics of people selected for study and those not selected

208
Q

How is selection bias minimised?

A

Representative sample
Cases and controls from same source
Maximise response
Minimise subjects lost to follow-up

209
Q

What is information bias?

A

Systematic differences in way information collected between/among groups being compared

210
Q

What is recall bias?

A

Information bias where cases more likely to recall presence of exposure due to already established prejudice about association between risk factor and outcome

211
Q

How is information bias minimised?

A

Ensuring methods for collecting information uniform between/among groups being compared

212
Q

What is confounding?

A

Influences relationship between exposure and outcome
3rd variable that
- Independently affects outcome
- Related to exposure

213
Q

What are the two universal confounders?

A

Age

Sex

214
Q

How is confounding minimised?

A
In design and execution of stages of study
- Matching by confounder
- Restriction
In analysis
- Stratification
- Multivariate analyses
215
Q

What are the three layers of the heart?

A

Epicardium
Myocardium
Endocardium

216
Q

What are the sub-layers of the epicardium?

A
Squamous epithelium
Sub-epicardial connective tissue
- Blood vessels
- Fat
- Nervous tissue
217
Q

What makes up the myocardium?

A

Muscle cells

Capillaries

218
Q

What are the sub-layers of the endocardium?

A

Endothelial layer
Subendocardial connective tisse
Conducting tissue

219
Q

Describe the cell size, location of nucleus, and fibre structure of cardiac muscle cells

A

Small cells
Central nucleus
Branching fibres
- Joined to each other by intercalated discs
Richly vascularised
WBCs between myocardial cells and capillaries

220
Q

What is the role of intercalated discs?

A

Anchors actin filaments in 1 cell’s sarcomere to sarcomere in another cells via fascia adherens

221
Q

How can intercalated discs come apart?

A

Removing Ca supply

222
Q

What are Purkinje fibres?

A

Larger, modified cardiac muscle cells with limited contractile machinery

223
Q

Where are Purkinje fibres found?

A

Bundles in subendocardium

224
Q

What is the role of the endothelium?

A

Inhibits clotting by secreting inhibitors
Secretes clotting factor into subendothelial tissue
Releases vasoactive substances

225
Q

Apart from constriction and relaxation, what is the role of smooth muscle in the tunica media of vessels?

A

Secretes connective tissue

  • Extracellular matrix
  • Collagen
  • Elastin
  • Ground substance
226
Q

Which layer is the vasa vasorum located in?

A

Adventitia

227
Q

Where are the elastic arteries generally found?

A

Closest to heat, where there’s highest blood pressure fluctations

  • Aorta
  • Carotid arteries
228
Q

What is the purpose of the many layers of elastin in elastic arteries?

A

Can stretch due to higher blood pressure
When heart relaxes, layers of elastin in media store energy > compress blood > restores energy to blood during diastole > blood pressure never drops to 0
Allows blood flow to be continuous

229
Q

What is the diastolic blood pressure due to?

A

Elastic energy applied to blood

230
Q

What is the primary role of muscular arteries?

A

Distribute blood to tissue

Regulate blood pressure through contractions of media

231
Q

How is capillary flow regulated?

A

Single smooth muscle cell sphincters

232
Q

What is the structure of capillaries?

A
Single endothelial cells rolled into tubed
Sealed to each other by tight junctions
Basal lamina = thick connective tissue
Sometimes media = pericyte
Adventitia = few collagen fibres
233
Q

How do the layers of veins differ from those of arteries?

A

Same layers
Media thinner
Adventitia thicker
have 1-way valves

234
Q

What type of vessel is the preferred site of diapedesis of leukocytes?

A

Venules

- Affected by histamines and other cytokines

235
Q

Where are the lymphatic vessels located in relation to the blood vessels?

A

On venous side

236
Q

How are lymphatics distinguished from veins?

A

Absence of RBCs

Presence of WBCs

237
Q

What are the risk factors for developing atherosclerosis?

A
Hyperlipidaemia
Hypertension
Smoking
Toxins
Haemodynamic factors
Immune reactions
Viruses
238
Q

What is the pathophysiology of atherosclerosis?

A

Endothelial injury/dysfunction > LDL entry and oxidation via ROS > cytokine production > recruitment of monocytes > phagocytosis of lipics and formation of foam cells > recruitment of lymphocytes > recruitment of smooth muscle cells from tunica media and formation of ECM > degeneration of components of plaque > lipid-rich necrotic core > initial remodelling of vessel wall preserves lumen diameter > continued endothelial dysfunction > vessel stenosis, impaired vasodilation, plaques vulnerable to rupture, local pro-thrombotic environment

239
Q

What is the definition of the metabolic syndrome?

A

Central obesity + any 2 of following

  • Raised triglycerides
  • Reduced HDL
  • Raised blood pressure
  • Raised fasting plasma glucose
240
Q

What are the complications of atherosclerosis?

A

Ischaemia
Infarction
Aneurysm

241
Q

Why can ischaemia follow atherosclerosis?

A

Fixed vessel narrowing

Acute plaque event

242
Q

Why can infarction follow atherosclerosis?

A

Occlusion by plaque/thrombus

243
Q

What are the types of aneurysms?

A

True
- Saccular
- Fusiform
False = extravasation into extravascular connective tissue

244
Q

What causes aneurysms?

A

Atherosclerosis
Congenital weakness in wall
Systemic hypertension
Infection in artery wall

245
Q

Describe the mechanism of hyaline arteriolosclerosis?

A

Age-related change caused by endothelial stress > deposition of plasma proteins and increased collage in vessel wall > arteriole wall thickens with homogenous eosinophilic hyaline > lumen narrows

246
Q

What can cause cardiac remodelling and hypertrophy?

A

Myocardial infarction
Cardiac damage
Volume overload
Pressure overload

247
Q

What effect on left ventricular mass and left ventricular wall thickness does concentric hypertrophy have?

A

Increased left ventricular mass

Increased left ventricular wall thickness

248
Q

What effect on left ventricular mass and left ventricular wall thickness does remodelling have?

A

Normal left ventricular mass

Increased left ventricular wall thickness

249
Q

What effect on left ventricular mass and left ventricular wall thickness does eccentric hypertrophy have?

A

Increased left ventricular mass

Normal relative wall thickness

250
Q

What is concentric hypertrophy?

A

Increased wall thickness without left ventricular enlargement
Often due to pressure overload
More sarcomeres in parallel

251
Q

What is eccentric hypertrophy?

A

Increased chamber size and normal relative wall thickness
Often due to volume overload
Myocyte stretching - more sarcomeres in series

252
Q

Describe LaPlace’s law

A

Wall stress = pressure pulling heart apart
- Stress counteracted by desmosomes joining myocytes
- Increasing wall thickness reduces wall stress because it increases number of desmosomes
Pressure greater determinant of wall stress than volume

253
Q

What is decompensation

A

Occurs in long-term following hypertrophy
Characterised by
- Reduced systolic function and cardiac output
- Increased left ventricular end diastolic pressure
- Eventual cardiac failure

254
Q

What are some environmental causes of concentric and eccentric hypertrophy?

A

Concentric: pressure overload due to high afterload
- Hypertension
- Aortic stenosis
Eccentric: volume overload due to high preload
- Mitral and aortic regurgitation
- Ventricular septal defect

255
Q

What are two genetic causes of ventricular hypertrophy?

A

Hypertrophic cardiomyopathy > thickened septum

Fabry’s disease

256
Q

How is left ventricular hypertrophy clinically identified?

A
Forceful apex beat
S3 and S4 heart sounds
ECG
Chest x-ray
Echocardiogram
MRI
257
Q

What are the causes of left ventricular remodelling?

A
Renin-angiotensin-aldosterone
Adrenergic nervous system
Endothelin
Cytokines
Local factors
258
Q

What are the causes of right ventricular hypertrophy?

A

Congenital
Pulmonary hypertension
Right heart valves

259
Q

Describe the pathology of hypertrophic cardiomyopathy?

A

Increased left ventricular wall thickness, especially of septum > left ventricle outflow tract obstruction > cellular hypertrophy > myocyte disarray

260
Q

What is athlete’s heart?

A

Eccentric hypertrophy with normal cardiac function

Usually regresses with deconditioning but may not regress > ventricular arrhythmia and sudden cardiac death

261
Q

What are the four determinants of cardiac output?

A

Contractility
Preload
Afterload
Heart rate

262
Q

Describe the length-tension relationship in muscle fibres

A

Increased sarcomere length > increased force of contraction

263
Q

How is left ventricular end diastolic pressure measured?

A

Measure pulmonary wedge pressure = ventricular pressure = atrial pressure

264
Q

How is right ventricular end diastolic pressure measured?

A
Non-invasive = JVP
Invasive = across tricuspid valve
265
Q

How do pulmonary artery wedge pressure and JVP reflect preload?

A

Pulmonary artery wedge pressure = left ventricular end diastolic pressure
JVP = right ventricular end diastolic pressure

266
Q

Why does cardiac failure happen?

A

Usually due to decreased cardiac output arising from systolic failure = loss of contractility

267
Q

How does cardiac failure happen?

A
Loss of myocardial muscle
- Ischaemic heart disease
- Cardiomyopathy
Pressure overload
- Aortic stenosis
- Hypertension
Volume overload
- Valve regurgitation
- Shunts
268
Q

How may cardiac failure worse by inappropriate homeostatic adaptations?

A

Increased end diastolic pressure and thus end diastolic volume to maintain same cardiac output
However, increased end diastolic volume > more fluid in left ventricle > more fluid in left atrium > more pressure in left atrium > pulmonary hypertension > pulmonary oedema

269
Q

What are the homeostatic mechanisms to combat cardiac failure?

A

RAAS stimulation > Na and water retention > peripheral/pulmonary oedema
Corresponding K loss and vasoconstriction

270
Q

Why is long term sympathetic stimulation of the heart unfavourable?

A

Vasoconstriction
Ventricular arrhythmias
Direct toxic effect

271
Q

What is the pathophysiology of diastolic heart failure?

A

Normal systolic function
Reduced left ventricular compliance due to
- Infarct
- Chronic hypertension
- Hypertrophy
Increased left ventricular end diastolic pressure required to fill left ventricle > increase in pulmonary venous pressure > pulmonary congestion/oedema

272
Q

Why is overdosing of diuretics unfavourable for cardiac function in patients with heart failure?

A

Drop in blood pressure to do compromised systolic function > cardiogenic shock

273
Q

How is peptidoglycan synthesised?

A

Synthesised from intermediates in cytoplasm on inner aspect of plasma membrane
Building block transported to exterior membrane and linked to growing peptidoglycan chain

274
Q

How does vancomycin work?

A

Binds to terminal D-Ala-D-Ala in peptidoglycan building block
Prevents cell wall synthesis

275
Q

What types of bacteria is vancomycin effective against?

A

Gram positive bacteria only, because can’t cross Gram negative outer membrane

276
Q

What is the basis of vancomycin resistance in enterococci?

A

Enterococci change terminal D-Ala to D-Lac > vancomycin can’t bind

277
Q

What is the mechanism of action of penicillin?

A

Has similar structure to D-Ala-D-Ala
Binds to and deactivates penicillin-binding proteins > no cross-linking
Defective synthesis of cell wall
Bacteria starts to break down its own cell wall

278
Q

What do beta-lactamases do?

A

Have similar structures to penicillin binding proteins
Beta-lactam antibiotic binds to beta-lactamase > hydrolysed
Unstable antibiotic > can’t fulfill role in disrupting cell wall synthesis

279
Q

What are the two mechanisms of resistance to beta-lactams?

A

Beta-lactamases

Altered penicillin-binding proteins

280
Q

What are the two types of beta-lactamase expression?

A

Expressed on plasmids
- Susceptible to clavulanic acid
Chromosomal expression
- Susceptible to ticarcillin

281
Q

Which beta-lactam is Pseudomonas aeruginosa susceptible to?

A

Ticarcillin

282
Q

What is the mechanism of beta-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA)?

A

Altered penicillin binding proteins

283
Q

Why is the combination of clavulanic acid and amoxycillin effective?

A

Clavulanic acid binds to beta-lactamase > inhibits it

Amoxycillin can now act

284
Q

How do aminoglycosides work?

A

Bind to 30S ribosomal subunit

Distort reading frame > altered proteins > early termination

285
Q

How does chloramphenicol work?

A

Binds to 50S ribosomal subunit

Interferes with peptidyl transfer

286
Q

How do macrolides work?

A

Bind to 50S ribosomal subunit

Interfere with translocation

287
Q

What are the mechanisms of resistance to aminoglycosides?

A

Efflux = drug pumped out of cell
Gram negative bacteria may modify outer membrane to reduce drug entry
Enzymatic modification > reduced entry
Ribosomal mutation > reduced drug binding

288
Q

Why is metronidazole only effective against strict anaerobes?

A

Strict anaerobes produce nitro-reductase > needed for activation of metronidazole into active form

289
Q

What is intrinsic resistance?

A

Resistant to antibiotic naturally

290
Q

What is acquired resistance?

A

Mutation

Horizontal gene transfer

291
Q

How does transfer of genes between bacteria occur?

A

Transformation
Phage-mediated transduction
Plasmid- mediated conjugation

292
Q

Why are multiresistance plasmids a major problem?

A

Potential for transfer between completely unrelated bacteria

Can lead to simultaneous acquisition of resistance to multiple antibiotics in 1 genetic event

293
Q

What is a benefit and risk of using digoxin?

A

Increases contractility but increases risk of dysrhythmias

294
Q

How does digoxin work?

A

Inhibits Na/K ATPase
Increased intracellular Na > decreased Ca exit
Increase Ca in sarcoplasmic reticulum
Increase Ca release with each action potential

295
Q

What is the drug class of digoxin?

A

Glycosides

296
Q

What are the adverse effects of using glycosides in heart failure?

A
Low therapeutic index
Affect all excitable tissues
- Gut: anorexia, nausea, diarrhoea
- CNS: drowsiness, confusin, psychosis
- Cardiac: ventricular dysrhythmias
Increased toxicity
- Hypokalaemia
- Hypercalcaemia
- Renal impairment
297
Q

How do beta1 agonists increase contractility?

A

Agonist binds beta1-adrenoceptor > in creased cAMP > phosphorylation of Ca channel > increased Ca entry

298
Q

What is dobutamine?

A

Selective beta1-adrenoceptor agonist

299
Q

What are the adverse effects associated with beta-adrenoceptor agonists?

A

Increased cardiac work

Risk of arrhythmias

300
Q

How do phosphodiesterase inhibitors work?

A

Block action of phosphodiesterase > increased cAMP > increased protein kinase activity > more open Ca channels > increased intracellular Ca > increased contractility

301
Q

What is the effect on adrenoceptors during chronic heart failure?

A

Chronic overactivation of beta1-adrenoceptors with sympathetic compensation for reduced cardiac output > reduced beta1-adrenoceptor expression and impaired beta1-adrenoceptor coupling > reduced sensitivity to beta1- adrenoceptor agonists/sympathetic drive

302
Q

What are the three main mechanisms leading to heart failure?

A

Decreased contractility due to loss of functional myocardial muscle
Pressure overload (afterload too high) due to
- Aortic stenosis
- Hypertension
Volume overload (preload too high) due to
- Valve regurgitation
- Shunts

303
Q

What are the compensatory responses to a fall in cardiac output?

A

Decreased CO > decreased BP > baroreflex > increased sympathetic outflow
- Beta-adrenoceptor activation > increased renin > increased angiotensin II > increased aldosterone > Na retention > volume expansion > increased preload
- Alpha-adrenoceptor activation + angiotensin II > vasoconstriction > increased afterload
Increased preload + afterload > increased oxygen demand > worsening function > myocardial remodelling

304
Q

Which drugs can reduce preload during congestion?

A

Venodilators
Diuretics
Aldosterone receptor antagonists
ADH receptor antagonists

305
Q

What is the drug class of nitrates?

A

Venodilators

306
Q

What is the drug class of frusemide?

A

Loop diuretics

307
Q

What is the drug class of spironolactone?

A

Aldosterone receptor antagonists

308
Q

What does spironolactone do?

A

Inhibit aldosterone action on cortical and distal tubules
Spares K
Shows increase in long term survival

309
Q

Which drugs can reduce afterload?

A
Arterial vasodilators
- Prone to reflex tachycardia
ACE inhibitors and ARBs
AT1 antagonists
Beta-adrenoceptor antagonists
310
Q

Which drugs are first line therapy for heart failure?

A

ACE inhibitors and ARBs

311
Q

In which grades of heart failure are ACE inhibitors and ARBs effective?

A

All

312
Q

What effect do ACE inhibitors and ARBs have on heart failure?

A

Improve symptoms
Delay progression of remodelling
Decrease constriction and fluid retention
Slows progression of hypertrophy

313
Q

What are the side effects of using beta-adrenoceptor antagonists for heart failure?

A
Hypotension
Fatigue
Bronchoconstriction
- Contraindication for asthma
Cold extremities
- Contraindicated in peripheral vascular disease
May cause/mask signs of hypoglycaemia
- Contraindicated for diabetes
314
Q

What is the difference between a clot and a thrombus?

A

Clot forms in static blood in vitro

Thrombus forms in vivo

315
Q

What is thrombosis?

A

Pathological formation of haemostatic plug in blood vessel in absence of blood loss

316
Q

What are the three main stages of physiological haemostasis?

A
  1. Vasoconstriction: exposed collagen on damaged vessel wall > platelets stick to collagen > become activated > release ADP and serotonin > serotonin powerful vasoconstrictor
  2. Platelet adhesion and activation: granule contents secreted > mediators synthesised > platelets aggregate and adhere
  3. Fibrin formation: thrombin cleaves fibrinogen
317
Q

What are some stimuli for platelet activation?

A

Collagen
Thrombin
Thromboxane
ADP

318
Q

How is blood coagulation controlled?

A

Enzyme inhibitors > cascade inhibition

Fibrinolysis by plasmin

319
Q

Which drugs can affect fibrin formation?

A

Procoagulant drugs
Injectable anticoagulants
Oral anticoagulants

320
Q

How does heparin work?

A

Injected

Enhances activity of antithrombin > inhibits and inactivated factor Xa and thrombin

321
Q

What version of heparin has a longer half life?

A

Low molecular weight heparin

322
Q

How does warfarin work?

A
Oral administration
Inhibits reduction of vitamin K > prevents activation of clotting factors
- II
- VII
- IX
- X
Delayed onset of action
323
Q

How is the anti-coagulant effect of heparin monitored?

A

APTT = activated partial thromboplastin time

Measure of intrinsic pathway

324
Q

Which drugs may lead to increased warfarin activity?

A
Aspirin
- Impaired platelet aggregation
NSAIDs
- Competition for plasma protein binding
Alcohol
- Competition for cytochrome p450 pathway
325
Q

How is the anti-coagulant effect of warfarin monitored?

A

PT = prothrombin time
Time for clot formation after addition of Ca and tissue factor
Measure of extrinsic pathway

326
Q

What is the INR?

A

Ratio of patient PT to normal PT

327
Q

Which drugs can affect platelet activation and adhesion?

A

ADP receptor antagonists
Thromboxane synthesis inhibitors
Glycoprotein IIb/IIIa receptor antagonists

328
Q

What is the drug class of clopidogrel?

A

ADP receptor antagonists

329
Q

How is streptokinase used?

A

Activates plasminogen
Used IV
Single use - antigenic and can activate immune system upon re-exposure because derived from bacteria

330
Q

How is alteplase used?

A

Human recombinant tPA
Not antigenic
IV infusion because has short half life

331
Q

How does aortic stenosis arise?

A

Progressive narrowing of aortic valve due to fibrosis and calcification

332
Q

How does the left ventricle respond to aortic stenosis?

A

Pressure overload of left ventricle > concentric hypetrophy > walls thicken > less compliance > diastolic dysfunction > increased left ventricular end diastolic pressure required to fill left ventricle > cardiac failure

333
Q

What are the two main types of causes of aortic regurgitation?

A
Damage of aortic leaflets
- Endocarditis
- Rheumatic fever
Dilatation of aortic root
- Marfan's syndrome
- Aortic dissection
- Collagen vascular disorders
- Syphilis
334
Q

Describe the progression of acute severe aortic regurgitation>

A

Sudden increase in left ventricular end diastolic pressure and left atrial pressure > acute pulmonary oedema > cardiogenic shock > early surgery needed to prevent death

335
Q

What are the causes of mitral regurgitation?

A

Myxomatous degeneration > mitral valve prolapse
Ruptured chordae tendineae > flail leaflet
Infective endocarditis
Myocardial infarct > ruptured papillary muscle
Rheumatic fever
Collagen vascular disease
Cardiomyopathy

336
Q

What is usually the cause of acute ischaemia?

A

Increased demand for oxygen that isn’t met

337
Q

What are the causes of infarction?

A
Arterial occlusion
- Thrombotic
- External compression in dissection
Venous occlusion
Systemic reduction in tissue perfusion
338
Q

What does the size of an infarction depend on?

A
Size of artery occluded
Duration of occlusion
Vulnerability of cells to ischaemia
Whether artery carrying oxygenated/deoxygenated blood
Nature of arterial supply
Oxygen content of blood
State of systemic circulation
339
Q

What are usually the causes of haemorrhagic infarcts?

A

Dual circulation/natural collateral circulation
Reperfusion
Venous occlusion

340
Q

What can cause endothelial dysfunction/injury?

A

Direct trauma
Atherosclerosis
Immunological/infective inflammation of endothelium/endocardim
Endocardium after MI

341
Q

What can cause hypercoagulability?

A
Post operative and post traumatic states
Genetic
Certain malignancies; eg: adenocarcinoma of pancreas
High oestrogen levels
- Peri-partum
- Oral contraceptives
Post myocardial infarction
Antiphospholipid Ab syndrome
Obesity
342
Q

What can cause changes in blood flow?

A

Turbulence; eg: aneurysm

Slowing

343
Q

What is gangrene?

A

Necrosis complicated by bacterial infection that eats dead organic matter = saprophytic

344
Q

What is primary gangrene?

A

Gas gangrene

345
Q

What is secondary gangrene?

A

Wet gangrene > complicating acute appendicitis/cholecystitis/infarction of small bowel
Dry gangrene > infarction of toes/foot/leg

346
Q

How does randomisation help to deal with confounding?

A

Aims to make treatment groups identical in all aspects other than intervention

347
Q

How is information bias dealt with?

A

Blinding - reduces preconceived ideas relating to intervention being undertaken

  • Single blind
  • Double blind
348
Q

What is intention to treat analysis?

A

Deals with selection bias
Assumes subjects remained in their randomised group, regardless of what they did in real life
Always underestimates any treatment effect

349
Q

What is survival analysis?

A

During follow up
Explicit capture of outcomes and their time of occurrence
Measures time to event
Usually plotted on the Kaplan-Meier curve

350
Q

What is the number needed to treat?

A

Number of people needed to undergo intervention in order to prevent outcome in one
Marker of efficiency of intervention

351
Q

How is the number needed to treat calculated?

A

1/absolute risk or rate reduction

352
Q

What is the number needed to treat affected by?

A

Relative effect

Underlying likelihood of outcome

353
Q

How does social gradient affect health?

A

Life expectancy shorter and disease more common down social ladder

354
Q

How does stress affect health?

A

Stressful circumstances and inability to cope damaging to health > can lead to premature death

355
Q

How does work affect health?

A

More control over work > better health

356
Q

How does unemployment affect health?

A

Job security increases health

357
Q

How does social support affect health?

A

Social support improves health

358
Q

How does food affect health?

A

Good diet and adequate food supply improve health

359
Q

What are the four stages of health transition?

A
  1. Age of pestilence and famine
  2. Age of receding pandemics
  3. Age of degenerative and man-made diseases
  4. Age of delayed degenerative diseases
360
Q

What are the most powerful class of diuretics?

A

Loop diuretics

361
Q

What is the mechanism of action of loop diuretics?

A

Acts on thick ascending limb of loop of Henle
Inhibits Na/K/2Cl carrier
Reduction in hypertonicity of blood > reduced water absorption into blood
Increased Na in distal tubule > increase osmotic pressure in tubule > reduced water reabsorption from tubule > torrential urine flow

362
Q

What are the pharmacokinetics of loop diuretics?

A

Well absorbed from gut
Plasma protein bound
Reaches site of action via secretion from blood into nephron
Duration = 3-6 hrs

363
Q

What are the adverse effects of loop diuretics?

A

K loss from distal tubule

H excretion > metabolic alkalosis

364
Q

What are the clinical uses of loop diuretics?

A
Ion/water overload in
- Acute pulmonary oedema
- Chronic heart failure
- Ascites
- Renal failure
Hypertension
365
Q

What is the mechanism of action of thiazide diuretics?

A

Acts on distal tubules to inhibit Na/Cl co-transporter

366
Q

What are the adverse effects of thiazide diuretics?

A

K loss from collecting ducts

Increased plasma uric acid > gout

367
Q

What are the clinical uses of thiazide diuretics?

A

Hypertension

Severe resistant oedema - in combination with loop diuretic

368
Q

When are potassium sparing diuretics used?

A

In combination with K losing diuretics to prevent K loss in patients with heart failure

369
Q

How does spironolactone work?

A

K sparing diuretic
Competitive antagonist of aldosterone receptor > reduced activation of Na channels and reduced stimulation of Na pump synthesis

370
Q

How do amiloride and triamterene work?

A

K sparing diuretics

Block luminal Na channels in collecting tubules and ducts > inhibition of Na reabsorption and inhibition of K secretion

371
Q

What are osmotic diuretics?

A

Pharmacologically inert
Filtered but not reabsorbed
Exert osmotic pressure

372
Q

Where do osmotic diuretics work?

A

Water permeable part of nephron

  • Proximal tubules
  • Descending loop of Henle
  • Collecting tubules
373
Q

When are osmotic diuretics used?

A

Raised intracranial pressure
Raised intraocular pressure
Prevention of acute renal failure

374
Q

How does gentamicin cause renal toxicity?

A

In apical membrane of proximal tubule
Alters generation of 2nd messengers > altered intracellular Ca > impaired mitochondrial respiration > cell injury
Nephrotoxicity can impair excretion > further renal damage

375
Q

What are the three fates of cholesterol made in the liver?

A

Ester formation and storage in liver for export in VLDL
Bile acid formation
Incorporation into membranes

376
Q

Which lipoproteins contain the most cholesterol?

A

LDL then VLDL

377
Q

What is familial hypercholesterolaemia?

A

Inherited dominant disorder
Mutation in LDL receptor gene
Failure of LDL receptors to take LDL from circulation
Elevated circulatory LDL

378
Q

Describe the different areas supplied by the coronary arteries

A

Anterior and anterior 2/3 septum = left anterior descending territory
Lateral = lateral circumflex territory
Posterior/inferior and 1/3 septum = posterior descending artery/right coronary artery territory

379
Q

What does oxygen supply to the myocardium depend on?

A

Oxygen content of blood

Myocardial blood flow

380
Q

Which section of muscle is most susceptible to ischaemia, and why?

A

Subendocardial muscle because subjected to greatest pressure, potentiallly impairing blood flow

381
Q

How is oxygen supply increased during periods of exertion?

A

In normal state, myocardium removes almost as much oxygen from blood as possible
So vasodilation to increase blood flow to meet increased oxygen demand

382
Q

Define ischaemic heart disease

A

Group of conditions in which there’s an imbalance between myocardial oxygen supply and demand > myocardial hypoxia and accumulation of waste products

383
Q

Which conditions are included under the umbrella of ischaemic heart disease?

A
Chronic coronary syndromes
- Stable angina
- Chronic cardiac failure
- Some arrhythmias
Acute coronary syndromes
- Unstable angina
- Sudden cardiac death
- Myocardial infarction
384
Q

Which part of coronary arteries does atherosclerosis primarily affect?

A

Epicardial parts

385
Q

How does atherosclerosis with stable plaque cause stable angina?

A

Chronic remodelling of vessel wall > fixed vessel stenosis and endothelial dysfunction > impaired release of vasodilators > inadequate vasodilation of smaller vessels > inability to increase flow during times of increased demand > stable angina

386
Q

What is an acute plaque event?

A

Ruptured plaque > blood exposed to subendothelial tissues > platelets activated by exposed collagen > vasoconstriction, primary haemostasis, secondary haemostasis > thrombus formation > acute narrowing of vessel lumen > acute ischaemia > acute coronary syndromes

387
Q

What is sudden cardiac death?

A

Unexpected fatal even occurring within 1 hour of beginning of symptoms/without symptoms in apparently healthy subjects/whose disease was not so severe as to predict such an outcome

388
Q

Desribe the progression of a myocardial infarction

A

ATP depletion > generation of ROS > altered Ca homeostasis > irreversible cell injury if continuing ischaemia

389
Q

What are the potential complications of a myocardial infarct after minutes to hours?

A

Damaged myocytes unstable > arrhythmia

Damaged myocytes can’t contract properly > cardiac failure

390
Q

What are the potential complications of a myocardial infarct after days?

A

Damaged myocytes unstable > arrhythmias
Damaged myocytes being destroyed > cardiac failure
Damaged wall isn’t moving normally > mural thrombus
Wall necrotic and weakened > rupture
Inflammatory mediators abound > fibrinous pericarditis

391
Q

How can the myocardial wall rupture?

A

Rupture of free ventricular wall > blood into pericardium = haemopericardium > compresses heart until it stops = cardiac tamponade
Rupture of papillary muscle > new onset murmur > mitral regurgitation > cardiac failure > acute severe cardiac failure = cardiogenic shock
Rupture of interventricular septum > new onset murmur > ventricular septal defect > cardiac failure > cardiogenic shock

392
Q

What are the potential complications of a myocardial infarct after weeks?

A

Damaged myocytes cleared, leaving islands of viable myocytes in healing tissue > arrhythmia
Damaged myocytes won’t be replaced > cardiac failure
Damged wall not moving normally > mural thrombus
Collagen deposition > wall strong again > flexible to wall stress > stretch > thin > bulge out > aneurysm > mural thrombus

393
Q

What are the potential complications of a myocardial infarct after months?

A

Arrhythmia
Cardiac failure
Aneurysm formed won’t regress - unlikely to rupture > mural thrombus

394
Q

What cellular changes occur in a myocardial infarct within 30 minutes?

A

Reversible ischaemia

395
Q

What cellular changes occur in a myocardial infarct within 12 hours?

A

Cell death and release of troponin

396
Q

What cellular changes occur in a myocardial infarct within 12-24 hours?

A

Necrosis

Start of acute inflammation

397
Q

What cellular changes occur in a myocardial infarct within 1-3 days?

A

Peak of acute inflammation

398
Q

What cellular changes occur in a myocardial infarct within 3-7 days?

A

End of acute inflammation
Start of repair by granulation tissue
Collage at 7 days

399
Q

What cellular changes occur in a myocardial infarct within 1-8 weeks?

A

Vascular granulation tissue > fibrous granulation tissue

400
Q

What cellular changes occur in a myocardial infarct after eight weeks?

A

Established fibrosis

401
Q

Outline the general treatment regime for dyslipidaemia

A

Establish fasting plasma lipid profile for diagnosis
Consider cardiovascular disease status and risk factors
Treat secondary causes
Manage modifiable risk factors

402
Q

What is the rate limiting step in cholesterol synthesis?

A

HMG-CoA > mevalonic acid by HMG-CoA reductase

403
Q

How does high cholesterol in the body lead to decreased endogenous cholesterol synthesis?

A

Negative feedback inhibits action of HMG-CoA reductase

404
Q

How does the body compensate in response to a decrease in endogenous cholesterol synthesis due to statin use?

A

Compensatory increase in hepatic LDL receptors > increased clearance of LDL from blood > decreased plasma total cholesterol and LDL > increased plasma HDL

405
Q

What is poor compliance to statins attributed to?

A

Perceived lack of efficacy due to no short term feeling of efficacy

406
Q

Why do serumu aminotransferases need to be measured every two to four months whillst on statins?

A

In case toxicity cause by drug-drug interactions

407
Q

Why might a patient on statins experience muscle pain?

A

Statin use can lead to minor increases in creatine kinase > breaks down muscle

408
Q

What are the adverse effects of statin use?

A
Mild GI symptoms
Headache
Insomnia
Dizziness
Myopathy - rare
Hepatitis, liver failure - rare
Renal failure - rare
409
Q

What are the contraindications for statin use and why?

A

Pregnancy - impair foetal myelination

Infection, pre-surgery, post-trauma - drug-drug interactions

410
Q

What is the mechanism of action of bile acid sequestrants?

A

Bind bile acid > prevent gut reabsorption of cholesterol > increase in bile excretion > increased demand for cholesterol for bile acid synthesis > upregulation of hepatic LDL receptors > removal of LDL from plasma > more cholesterol metabolism

411
Q

What are the adverse effect of bile acid resins?

A
Abdominal discomfort
Bloating
Constipation
Flatulence
Steatorrhoea
Decreases absorption of other drugs as not very specific
412
Q

What is the mechanism of action of ezetimibe?

A

Acts as sterol transporter to intestine > inhibit cholesterol absorption from intestine

413
Q

Why does ezetimibe not affect absorption of bile acids and fat soluble vitamins?

A

Extremely specific

414
Q

What is the mechanism of action of niacin?

A

Unclear

Normally used in combinations, usually after other combinations have been ineffective

415
Q

How does hiacin decrease the effects of atherosclerosis?

A

Decreases secretion of VLDL from liver
Reduces plasma LDL and triglycerides
Increases HDL
Lowers potentially atherogenic lipoprotein A

416
Q

What are the adverse effects of niacin use?

A
Vasodilation
- Flushing
Hypotension
Nausea
Vomiting
417
Q

What is the mechanism of action of fibrates?

A

Agonist at nuclear receptors > regulates gene expression to increase synthesis of lipoprotein lipase > increased breakdwon of triglycerides

418
Q

What precautions must be taken when administering fibrates?

A

Mild elevation of serum aminotransferases

Monitor at 3 month intervals

419
Q

What are the common adverse effects of fibrate use?

A

Nausea
Dry mouth
Headache
Rash

420
Q

How does orlistat act to reduce blood triglyceride levels?

A

Lipase inhibitor > triglycerides excreted undigested

421
Q

Why does most of coronary perfusion occur in diastole?

A

During systole coronary arteries compressed

422
Q

What are the aims of stable angina treatment?

A

Prevent attacks
Relieve symptoms
Prevent progression to myocardial infarct

423
Q

What is the mechanism of action of nitrates?

A

Biotransformation > releases nitric oxide > vascular relaxation

424
Q

Where do nitrates act?

A

Mainly in veins to decrease preload

Also in large arteries to decrease afterload

425
Q

What is glyceryl trinitrate (GTN)?

A

Short acting nitrate
Given sublingually
- IV for emergency

426
Q

What is isosorbide dinitrate?

A
Longer accting nitrate
Given orally 
For
- Anticipation of effort
- Prophylaxis
427
Q

What are the adverse effects of nitrate use?

A

Postural hypotension
Headache
Flushing
Reflex tachycardia

428
Q

What is the selectivity of verapamil?

A

1:1 vascular:cardiac selectivity
Can’t be used during heart failure
Can be used for angina and hypertension

429
Q

What is the selectivity of diltiazem and nifedipine?

A

Vascular selective
Used in angina and hypertension
Can also be used during heart failure

430
Q

What is the mechanism of action of calcium channel blockers in the use of ischaemic heart disease?

A

Blocks Ca entry into heart through L-type channels > decreased HR > increased filling time > increased preload > decreased contractility > decreased cardiac output > reduced oxygen demand
Vascular selective: blocks Ca entry into muscle cells of vessels > arterial dilation > reducd afterload

431
Q

What are the adverse effects of calcium channel blockers in angina?

A
Flushing
Headache
Oedema
Nifedipine: hypotension
Verapimil: bradycardia, atrioventricular block
432
Q

How do atenolol and propanolol differ?

A

Atenolol cardio-selective beta1-adrenoceptor antagonist

Propanolol non-selective beta-blocker

433
Q

How does ivabradine work?

A

Blocks If > inhibits Na into cell > decreased velocity of diastolic depolarisation

434
Q

What are the indications for ivabradine use?

A

Patients with

  • Ischaemic heart disease
  • Left ventricular dysfunction, and
  • Heart rate of >70 bpm
435
Q

What are the adverse effects of ivabradine?

A

Brightness in visula field

Conduction abnormalities

436
Q

How is variant angina treated?

A

Relieve coronary spasm with short acting nitrate

Prophylaxis with vascular selective Ca channel blocker

437
Q

How is unstable angina treated?

A

Same as with stable angina

Aspiring to prevent thrombosis

438
Q

What are the methods of antimicrobial testing?

A
Dilution methods
- Minimum inhibitory concentration
Diffusion methods
- Disc susceptibility test
E-test strips combine disc susceptibility test and minimum inhibitory concentration
439
Q

What is the rationale for using antibiotic combinations?

A

Temporary measure in ill patient
Delay emergence of resistance
Treat mixed infections
Achieve synergistic effect

440
Q

What are the mechanisms of synergy?

A

Block sequential steps of metabolic pathway
Inhibits enzymatic degradation
Enhance antimicrobial uptake by bacterial cell

441
Q

What are Jawetz’s laws of antibiotic synergy?

A

Bacteriostatic bacteriostatic = additive/indifferent
Bacteriostatic + bactericidal = antagonistic
Bactericidal + bactericidal = synergistic

442
Q

What did the Whitehall study show in relation to employment and increased risk of death from cardiovascular disease?

A

Lower employment grde associated with increased risk of death from cardiovascular disease

443
Q

What was attributed to the higher risk of mortality with a lower employment grade being because of?

A

60% - chronic stress related to lack of control within job
Lower income > decreased access to healthcare
Poorer diet
Less education
Smoking
Obesity
Less leisure time

444
Q

What are the two determinants of the psycho-biological stress response?

A

Stressors

Resistance/vulnerability factors

445
Q

Describe the formation of an X-ray image

A

Image = shadown on detector
X-ray hits detector > converts sliver-halide crystals to silver
More x-rays hit detector > blacker image

446
Q

Describe the concept of e-densities

A

How many x-rays get through an object dependent on electron density of object

447
Q

List the order of e-densities of different objects, from lowest (black) to highest (white)

A
Air
Fat
Soft tissue
Bone
Contrast agents
Metal
448
Q

What is a silhouette sign?

A

Interface only seen if tissues of differing e-densities next to each other
Will produce silhouette sign

449
Q

Why is an erect P-A chest x-ray performed under full inspiration?

A

Lung fulll of air > good silhouette for other structures

450
Q

Why are chest x-rays usually perfromed in the P-A direction?

A

Heart closer to fil and less magnified - edges of heart less blurred
Patient instructed to hug x-ray cassette so scapulae moved away from chest wall

451
Q

Why are chest x-rays usually performed with the patient in the erect position?

A

Blood flow distribution in lungs can be assessed
Vessels nearer lung base larger than vessels more superior
Easier to see pleural effusions

452
Q

In left heart failure, how do the vessels in a chest x-ray look different than normal?

A

Pulmonary vascular redistribution

- Pulmonary vessels near hilum and apex become larger and more dilated due to congestion

453
Q

Why does hypertrophy occur?

A

Increased functional demand/stimulation by hormonal or growth factors

454
Q

What happens to cellular components during atrophy?

A

Cells decrease in size and activity
Removed by autophagy
Apoptosis

455
Q

When does atrophy become irreversible?

A

Cell loss and associated fibrosis

456
Q

What is involution?

A

Physiological atrophy involving apoptosis

457
Q

Define metaplasia

A

Change from one fully differentiated mature cell type to another - better able to withstand new environmental conditions

458
Q

What is the mechanism underlying cell metaplasia?

A

Cytokine/growth factor driven reprogramming of line of differentiation

459
Q

What are the causes of disturbed valve function?

A
Congenital abnormalities
Myxomatous mitral valve
Degenerative changes due to calcifcation
Inflammatory
- Infection
- Immune mediated
460
Q

What are the possible complications of valve replacements?

A

Porcine valves susceptible to infective endocarditis
Prosthetic mechanical valves predispose to thrmobosis of valve
- Need to take anticoagulant drugs to prevent thrombosis

461
Q

Why does concentric hypertrophy develop?

A

Increased afterlad
Preserves systolic function temporarily
Ultimately thickened myocardium > impaired perfusion > ischaemia

462
Q

Why does eccentric hypertrophy develop?

A

Volume loads
Aids function up to a point
Predisposes myocardium to ischaemia

463
Q

What happens as myocardial hypertrophy worsens?

A
Decompensation
Microscopically
- Fibrosis
- Apoptosis
Dilates > increased end diastolic volume