Endocrinology Flashcards

1
Q

Name the two most common causes hyperthyroidism?

A
  • Graves’ Disease
  • Nodular Goitre (Plummer’s Disease) -> a benign tumour producing thyroxine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is Graves’ Disease?

A
  • an autoimmune disease that produces antibodies that bind to and stimulate the TSH receptor in the thyroid
  • the stimulation leads to the thyroid gland becoming smoothly enlarged (goitre)

No action

Mouse Gestures

Back

Forward

Scroll up

Scroll down

Switch to previous tab

Switch to next tab

Close all tabs except current

Close current tab

Open new tab

Close all tabs

Refresh current tab

Stop loading

Scroll to bottom

Scroll to top

Reopen closed tab

Go to home page

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

How do patients suffering from Graves’ Disease present?

A
  • patients will come complaining of being overactive with weight loss and sweating etc.
  • on palpation, the thyroid gland will feel very diffuse and it’ll be smoothly enlarged
  • a family history of hyperthyroidism
  • exophthalmos - there is another antibody that binds to the growth factor receptors behind the eye -> muscles behind the eye grow and push the eyes forward (worsened by smoking)
  • localised pretibial myxoedema (weird growth of soft tissue in the shin) - caused by another antibody
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

On further medical examination, what else would you find in a patient with Graves’ Disease?

A
  • a rapid pulse (raised metabolic rate)
  • warm
  • imaging the thyroid, by giving the patient some radioactive iodine, will show up on the scintigram, showing that the iodine is going into the thyroid gland and the whole thyroid gland is active
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is Plumber’s Disease?

A
  • a toxic nodular goitre, originating from a benign adenoma that is overactively producing thyroxine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How do patients suffering from Plumber’s Disease present?

A
  • with generally hypothyroidism symptoms
  • distinct symptoms is on palpation and examination there is a lump on one side
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

On further medical examination, what else would you find in a patient with Plumber’s Disease?

A
  • because there is too much thyroxine coming from the tumour the pituitary will stop making TSH -> normal part of the thyroid will slowly shrink and stop making thyroxine
  • the iodine thyroid scan you will just see a hot nodule and the rest of the thyroid scan will not be seen because of the negative feedback from the tumour
  • sometimes patients may have a multinodular goitre

No action

Mouse Gestures

Back

Forward

Scroll up

Scroll down

Switch to previous tab

Switch to next tab

Close all tabs except current

Close current tab

Open new tab

Close all tabs

Refresh current tab

Stop loading

Scroll to bottom

Scroll to top

Reopen closed tab

Go to home page

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

What are the effects of thyroxine on the sympathetic nervous system?

A
  • SENSITISES beta adrenoceptors to ambient levels of adrenaline and noradrenaline -> normal levels of adrenaline and noradrenaline will have much stronger effects than they should -> patients with hyperthyroidism will also have features of having too much adrenaline e.g. palpitations, tachycardia, tremor, lid lag
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Name some symptoms of having too much adrenaline?

A
  • palpitations
  • tachycardia
  • tremor
  • lid lag
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Summarise the symptoms of hyperthyroidism?

A
  • weight loss despite increased appetite
  • breathlessness
  • palpitations
  • tachycardia
  • sweating
  • heat intolerance
  • diarrhoea
  • lid lag and other sympathetic features
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is a Thyroid Storm (Thyrotoxic Crisis)?

A
  • a rare but severe complication of hyperthyroidism - MEDICAL EMERGENCY (50% mortality if untreated - die of heart failure or arrhythmia)
  • features of thyroid storm:

o hyperpyrexia > 41 degress

o tachycardia/arrhythmia – over 170 bpm

o cardiac failure

o delirium/frank psychosis

o hepatocellular dysfunction, jaundice

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

What is the cause of thyroid storm?

A
  • hyperthyroidism that has been left untreated for long enough, two or more features of a thyroid storm = high risk of death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

How is the diagnosis of a thyroid storm confirmed?

A
  • a blood test
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the treatment options for thyroid storm?

A

must be prompt and aggressive treatment

  • surgery (thyroidectomy)
  • radioiodine
  • drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Name the classes of drugs used to treat hyperthyroidism?

A

· Thionamides (anti-thyroid drugs)

· Potassium iodide

· Radioiodine

· Beta-blockers - others reduce thyroid hormone synthesis, beta blockers simply help symptoms

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

What are the uses of thionamides?

A
  • daily treatment of hyperthyroid conditions -> Graves’ or Plummer’s
  • control hyperthyroidism before thyroidectomy -> don’t want to give a general anaesthetic to someone who is tachycardic with a labile heart rate
  • following radioactive iodine treatment -> radioactive iodine takes a while to work so something else must control the thyroid hormone level before the radiotherapy starts to take effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Name the 2 main thionamides.

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

Describe the synthesis of thyroid hormone.

A
  • iodine is taken up into the follicular cells
  • under the action of thyroperoxidase with hydrogen peroxide you get iodination of tyrosine residues in the thyroglobulin
  • coupling of monoiodotyrosine and diiodotyrosine to form T3 and T4, which is taken up and released by the cells into the circulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the mechanism of thionamides

A
  • INHIBIT THYROPEROXIDASE
  • hence, they inhibit the iodination of thyroglobulin and the coupling of iodotyrosines therefore there is a net reduction in synthesis and secretion of thyroid hormones
  • also suppress antibody production in Graves’ and diodination of T4 to T3 in peripheral tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How long does it take for thionamides to have an effect?

A
  • the biochemical effects of inhibition with thionamides occurs in hours but it takes weeks before the clinical effects can be seen
  • this is because there is a lot of STORED thyroid hormone in the lumen of the thyroid follicles and anti-thyroid hormones only affect the synthesis of thyroid hormones, not stored thyroid hormone
  • NO CHANGE FOR 7-10 DAYS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the clinical consequence of the lag between giving the thyroid drug and the response?

A
  • symptoms have to managed short-term so non-selective beta blockers are given
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are the unwanted side effects of thionamides?

A
  • agranulocytosis/granulocytopenia (reduction or absence of granular leukocytes) - rare and reversible on withdrawal of the drug
  • rashes (relatively common)
  • headaches
  • nausea
  • jaundice
  • joint pain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Describe the pharmacokinetics of thionamides.

A
  • orally active
  • plasma half-life of 6-15 hours
  • crosses the placenta and is secreted in the milk
  • metabolised in the liver and excreted in the urine
  • carbimazole is a pro-drug which is converted to methimazole (active)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the significance of thionamides crossing the placenta?

A
  • thyroid disease is common in women around reproductive age so you must consider pregnancy
  • patients can conceive on thionamides but ideally on as low a dose as possible -> high doses of thionamides in a pregnant woman could cause foetal hypothyroidism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the significance of thionamides being secreted in milk?

A
  • thyroid disease is common in women around reproductive age so you must consider pregnancy and neonatal period
  • patients can breast-feed on thionamides but ideally on as low a dose as possible -> high doses of thionamides in a pregnant woman could cause foetal hypothyroidism
  • propylthiouracil crosses into breast milk LESS than carbimazole -> breast-feeding woman are put on PTU over CBZ
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

How long is it aimed to keep a patient on thionamides?

A

18 months -> 50:50 chance of relapse after stopping

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

When is iodide treatment used?

A

o Preparation of hyperthyroid patients for surgery

o Severe thyrotoxic crisis (thyroid storm)

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

What compound is most commonly used in iodide treatment?

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

What is the mechanism of action of KI?

A
  • Wolff-Chaikoff Effect = the temporary reduction in thyroid hormones following ingestion of large amounts of iodine
  • an autoregulatory phenomenon -> the thyroid rejects the ingested iodide, hence prevents the gland from sucking up loads of iodine and making too much thyroid hormone
  • KI inhibits the iodination of thyroglobulin and the generation of hydrogen peroxide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

How long does it take for KI to have an affect?

A
  • very quick, a matter of hours
  • maximum effect after 10 days of contiuous administration
  • over weeks it reduces the size and vascularity of the thyroid
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are the unwanted side effects of KI?

A
  • ALLERGIC REACTION
  • rashes
  • fever
  • angioedema
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Describe how KI is given?

A
  • given orally or Lugol’s Solution or Aqueous Iodine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is Radioiodine 131I- high dose used to treat?

A
  • Graves’
  • Plummer’s
  • Thyroid Cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is Radioiodine mechanism of action?

A
  • permanently switching off the thyroid without surgery
  • the method relies on the thyroid gland taking up iodine to make thyroid hormone so thyroid follicular cells take up the radioiodine so, it accumulates in the colloid -> emits beta particles of radiation that destroy the follicular cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What do some patients complain about as a result of radioiodine treatment?

A
  • discomfort in the neck
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What other drugs must be taken into account when prescribing someone with radioiodine?

A
  • anti-thyroid drugs must have been discontiued 7-10 days prior to radioiodine treatment so that the thyroid gland can rev up and be really active so that it sucks up a lot of radioactive iodine when it is administered to get maximal destruction of the gland
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Give the dose of radioiodine given to treat Graves’ Disease.

A
  • oral dose of approx. 500 MBq
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What is the dose of radioiodine given to patients suffering from Thyroid Cancer?

A
  • oral dose of circa 3000 MBq
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe the radioactivity of radioiodine.

A

o Radioactive half-life = 8 days

o Radioactivity negligible after 2 months (maximum effect 2-3 months)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What precautions must be taken after taking radioiodine?

A
  • avoid close contact with small children for several weeks
  • contra-indictaed in pregnancy and breast-feeding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the results of a thyroid uptake scan of Graves’ Disease?

A
  • entire gland is active and smoothly enlarged
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are the results of a thyroid uptake scan of a patient with Plummer’s?

A
  • single focus of activity and the rest is supressed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the results of a thyroid uptake scan of a patient with Thyroiditis?

A
  • inflamed thyroid gland where there is NO activity at all
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What is Viral Thyroiditis?

A
  • caused by a virusattacking the thyroid gland and it causing a fever
  • it damages the thyroid follicles and all the stored thyroxine gets released so patients present with OVERACTIVE thyroid, after a bout a month when all the thyroxine has ran out they will become hypothyroid as the damaged gland will no longer make thyroxine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are the symptoms of viral thyroiditis?

A
  • piainful dysphagia
  • hyperthyroidism
  • pyrexia
  • raised erythrocyte sedimentation rate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the treatment for viral thyroiditis?

A
  • you have to wait for all the thyroxine to have been used up before giving thyroxine to treat the hypothyroidism
  • drugs can be given to help with the symptoms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q
A

D

  • all the others will control thyroid function but the lugol’s iodine, used for 10 days before the operation, will reduce the size and vascularity of the thyroid gland
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are the 6 anterior pituitary hormones?

A
  • FSH
  • LH
  • GH
  • TSH
  • ACTH
  • prolactin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Define hypopituitarism.

A
  • decreased production of ALL anterior pituitary hormones (panhypopituitarism) or of specific hormones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

How common are congeital reason for panhypopituitarism?

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

Name a gene mutation which leads to panhypopituitarism.

A
  • PROP1 mutation - this is a transcription factor that allows the development of the pituitary gland to take place
  • is congenital
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the causes of acquired panhypopituitarism?

A

o Tumours -> hypothalamic (craniopharyngiomas) or pituitary (denomas, metastases, cysts)

o Radiation -> hypothalamic/pituitary damage (GH most vulnerable, TSH relatively resistant)

o Infection -> meningitis

o Traumatic brain injury

o Infiltrative disease -> often involves pituitary stalk (neurosarcoidosis)

o Inflammatory (hypophysitis)

o Pituitary apoplexy -> haemorrhage (or less commonly infarction)

o Peri-partum infarction (Sheehan’s syndrome)

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

What is the usually loss of secretion of hormones in panhypopituitarism?

A

o Gonadotrophins (LH and FSH)

o GH

o Thyrotrophin

o Corticotrophin

o Prolactin deficiency is uncommon/unrecognised

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

What are the symptoms of hypopituitarism?

A

o Secondary Amenorrhoea OR Oligomenorrhoea

o Impotence

o Loss of libido

o Erectile dysfunction

o Tiredness

o Waxy skin

o Loss of body hair

o Hypotension

  • no really symptoms for growth hormone in adults
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What is Sheehan’s Syndrome?

A

o develops acutely following post-partum haemorrhage resulting in PITUITARY INFARCTION

  • blood loss results in vasoconstrictor spasm of hypophysial arteries and this leads to: ischaemia of the pituitary (enlarged during pregnancy due to swelling of prolactin glands) and necrosis of the pituitary
  • develops very FAST

o more prevelant in less developed countries as hypotension post-partum is more common

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

What is the presentation of Sheehan’s Syndrome?

A
  • lethargic
  • anorexia
  • weight loss
  • failure of lactation
  • failure to resume menses post-delivery

o posterior pituitary usually not affected

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

What is pituitary apoplexy?

A
  • intra-pituitary haemorrhage or, less commonly, infarction
  • often dramatic, rapid presentation in patients with pre-existing pituitary tumours (adenomas) -> may be first presentation of a pituitary adenoma
  • often causes severe headaches and visual field defects (depression on the optic chiasm)
  • can be precipitated by anti-coagulants
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What differentiates between Sheehan’s Syndrome and pituitary apoplexy?

A
  • pituitary apoplexy isn’t specific to women
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What might be lead to if the cavermous sinus becomes involved?

A
  • diplopia (IV, VI), ptosis (III)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

How can hypopituitarism be diagnosed?

A

o Biochemical

  • basal plasma values of pituitary or target endocrine gland hormones
  • stimulated (dynamic) pituitary function test

o Radiological

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

What is the problem for testing for basal plasma values of pituitary or target endocrine gland hormones?

A
  • most things released from the hypothalamus are released in pulses -> single measurements are not useful -> low or high results could be physiological
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is a stimulated pituitary function test?

A
  • done using a combined function test -> involves the administration of various releasing hormones -> releasing hormones are administered IV
  • the same thing can be done with just one releasing hormone if you’re testing for a specific hypothyroidism -> the specific test for GH is the insulin-induced hypoglycaemia test

No action

Mouse Gestures

Back

Forward

Scroll up

Scroll down

Switch to previous tab

Switch to next tab

Close all tabs except current

Close current tab

Open new tab

Close all tabs

Refresh current tab

Stop loading

Scroll to bottom

Scroll to top

Reopen closed tab

Go to home page

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What can be revealed by a pituitary MRI?

A

o specific pituitary pathology eg haemorrhage (apoplexy), adenoma

o empty sella – thin rim of pituitary tissue

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

What is the general treatement for hypopituitarism?

A
  • hormone replacement therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is the treatment for ACTH deficiency?

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

What is the treatment for TSH deficiency?

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

What is the treatment for GH deficiency?

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

What is the treatment for LH/FSH deficiency in women?

A
  • oestrogen
  • if the patient has a uterus progesterone must be given with the oestrogen - prevents endometrium hyperplasia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the GH also known as?

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

What are the effects of a lack of somatotrophin?

A

o children -> pituitary dwarfism

o adults -> effects are unclear

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

Name the causes of short stature in children?

A

o Lack of GH

o Genetic -> Down’s syndrome, Turner’s syndrome, Prader Willi syndrome

o Malnutrition

o Emotional deprivation (potentially due stress response effecting the growth axis)

o Endocrine disorders -> Cushing’s syndrome, Hypothyroidism, GH deficiency, poorly controlled T1DM

o Systemic disease -> Cystic Fibrosis, Rheumatoid arthritis

o Malabsorption -> Coeliac disease

o Skeletal dysplasia -> Achondroplasia, osteogenesis imperfecta

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

Describe the growth axis.

A
  • GHRH is released from the hypothalamus and truggers the anterior pituitary to release GH
  • GH caues growth in target tissue as well as stimulating the production of IGF I and IGF II in the liver-> IGF I mediates growth
  • GH is controlled in the hypothalamus by GHRH and GH
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What is Laron Dwarfism?

A
  • a form of dwarfism caused by a GH receptor defect -> results in low IGF I levels because GH isn’t having an effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What is the cause of teritary hypopituitarism?

A
  • specific hypothalamic hormone defects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Give an example of tertiary hypopituitarism.

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

Name some GnRH deficiencies.

A
  • Kallmann’s Syndrome
  • Prader-Willi-Syndrome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Describe Kallmann’s Syndrome.

A
  • caused by a genetic defect where the neurones in the embryo that will go on to produce GnRH are unable to migrate to the hypothalamus -> hypothalamus LACKS GnRH neurones
  • the defect also prevents the migration of the neurones that are going to develop a sense of smell resulting in various degrees of anosmia
  • hypogonadism -> untreated suffers never go through puberty
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

How is a diagnosis of short stature made in a child?

A
  • if they fall below their percentile on the charts within their red book
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What are the likely causes of acquired GH deficiency in adults?

A
  • trauma
  • pituitary tumour
  • pituitary surgery
  • cranial radiotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

How is GH deficiency diagnosed?

A

o GH provocation tests in which plasma GH is measured at specific points before and aftwerwards

  • GHRH + ARGININE (IV) - (in combination more effective than each alone)
  • INSULIN (IV) – via hypoglycaemia
  • GLUCAGON (IM)
  • EXERCISE (e.g. 10 min step climbing; when appropriate)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What is the NICE cut-off for GH deficiency in response to hypoglycaemia?

A
  • 3mcg/L
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is the treatment for GH deficiency?

A

o hormone replacement therapy

  • human recombinant GH
  • daily, subcutaneous injection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the signs and symptoms of GH deficiency in adults?

A

o impaired ‘psychological well being’ and reduced quality of life -> only proved symptom

  • reduced lean mass
  • increased adiposity
  • increased waist:hip ratio
  • reduced muscle strength & bulk à reduced exercise performance
  • decreased plasma HDL-cholesterol & raised LDL-cholesterol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

State the potential benefits of GH therapy in adults.

A

o improved psychological well being and quality of life

  • improved body composition -> decreased waist circumference, less visceral fat
  • improved muscle strength and exercise capacity
  • more favourable lipid profile - higher HDL-cholesterol, lower LDL-cholesterol
  • increased bone mineral density
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What are the potential Risks of GH Therapy in Adults?

A

o increased risk of cardiovascular accidents -> due to its growth promoting effects, an excess of GH can cause cardiomegaly

o increased soft tissue growth, leading to e.g. cardiomegaly

o expensive

o increased susceptibility to cancer was thought but proved wrong

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

Describe Hyperpituitarism.

A

o symptoms associated with excess production of adenohypophysial hormones

  • can be due to isolated tumours but can be ectopic in origin (eg vasopressin in lung tumours)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What is hyperpituitatarism associated with?

A
  • visuial field defects
  • often accompanied with other defects particularly cranial nerve defects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Exactly, what crosses at the optic chiasm?

A
  • fibres from the inner (nasal) part of the retinae cross
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What kind of visual impairment is caused by an enlarged pituitary gland?

A

o bitemporal heminaopia

  • you lose the temporal/peripheral vision
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

How are bitemporal hemianopia assessed?

A

o perimetry

  • press button when the patient can see the flashing light which builds up a visual field
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What is the difference between Cushing’s Disease and Cushing’s Syndrome?

A
  • the syndrome is simply to much ACTH whereas the disease is due to a pituitary adenoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Give two physiological and one pathological reason for hyperprolactinaemia.

A

o physiological = pregnancy and breastfeeding

o pathological = prolactinoma (often microadenomas<10nm diameter)

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

In terms of the gonadal axis, what does high prolactin do?

A
  • it suppresses GnRH pulsatility
  • can cause secondary hypogonadism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are the presenting complaints/problems with hyperprolactinaemia in women?

A
  • galactorrhoea (milk production when not breastfeeding)
  • secondary amenorrhoea or oligomenorrhoea
  • loss of libido
  • infertility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the presenting complaints/problems with hyperprolactinaemia in men?

A
  • loss of libido
  • erectile dusfunction
  • infertility
  • galactorrhoea can occur but is very uncommon
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What supresses prolactin release?

A
  • dopamine binding to D2 receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

How is hyperprolactinamia treated?

A
  • 1st line = dopamine receptor agonists therefore inhibiting prolactin and reduce the tumours size
  • include bromocriptine and cabergoline via oral administration

o unusual that it is medical and not surgical treatment

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

What are the side effects of dopamine receptor agonists?

A
  • nausea and vomiting
  • postural hypotension
  • dskinesias
  • depression
  • pathological gambling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What does excess GH causes?

A

o in children = gigantism

o in adults = acromegaly

  • in adults, the growth plates of the long bones have fused so there is no longer a possibility of an increase in height but you still get other effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What is the major cause of excess GH?

A
  • usually benign growth hormone secreting pituitary adenoma
  • maybe benign but can be deadly due to pressure it can provide on cranium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Describe the signs and symptoms of acromegaly. Go onto state the issues that arise if acromegaly is left untreated.

A
  • incidious onset -> gradual changes over many years

o increased morbidity and mortality

  • > death: CVD = 60%
  • > respiratory complications = 25%
  • > cancer = 15%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What grows in acromegaly?

A

o periosteal bone

o cartilage

o fibrous tissue

o connective tissue

o internal organs (cardiomegaly, splenomegaly, hepatomegaly etc.)

o prognathism

o enlarged supraorbital ridges

o enlarged soft tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

State the clinical features of acromegaly.

A
  • EXCESSIVE SWEATING (HYPERHIDROSIS)
  • HEADACHES
  • enlargement of supraorbital ridges, nose, hands and feet, thickening of lips and general coarseness of features
  • enlarged tongue (macroglossia)
  • mandible grows causing protrusion of lower jaw (prognathism)
  • carpal tunnel syndrome (median nerve compression)
  • barrel chest
  • kyphosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What are the metabolic effects of acromegaly?

A

excess GH -> increased endogenous glucose production and decreased muscle uptake -> increased insulin production -> increased insulin resisitance -> impaired glucose tolerance (50% of patients) -> diabetes mellitus

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

State 4 major clinical complications linked to acromegaly.

A

o obstructive sleep apnoea -> bone and soft-tissue changes surrounding the upper airway lead to narrowing and subsequent collapse during sleep

o hypertension

o cardiomyopathy -> hypertension, DM, direct toxic effects of excess GH on myocardium

o increased risk of cancer -> colonic polyps, regular screening with colonoscopy

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

What other hormone if often high in acromegaly?

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

State and explain the ‘gold standard’ test for acromegaly.

A
  • oral glucose tolerance

o paradoxical rise of GH following oral gluscose load

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

What is the treatment for acromegaly.

A

o SURGERY is the main treatment - transphenoidal hypophysectomy

o Radiotherapy - problem is that over a long period of time the patient ends up hypopituitary as a result

o Chemotherapy/Drugs -> somatostatin analogues or dopamine Agonists

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

Name a somatostatin analogue.

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

Name two dopamine agonsits.

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

What is the clincal use of octreotide?

A
  • short-term treatment before pituitary surgery -> reduces the size of the tumour to make surgery easier
  • treatment of other neuroendocrine tumours e.g. carcinoid tumours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

How are somatostain analogues administed?

A
  • subcutaneous or intramuscular
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the side effecst of somatostatin analogues?

A

o GI tract disturbances (somatostatin is produced by the small intestine) -> gallstones

o initial reduction in insulin secretion - transient hyperglycaemia -> inhibits the production of insulin by beta cells leading to transient hyperglycaemia

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

What does the posterior pituitary look like on MRI?

A
  • is the bright spot at the back of pituitray gland
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What two hormones are produced in the neurohypophysis/posterior pituitary?

A
  • vasopressin (ADH)
  • oxytocin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What is the role of oxytocin?

A
  • constriction of myometrium at parturition
  • milk ejection reflex
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

How is vasopressin regulated?

A
  • osmoreceptors in the organum vasculosum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What are the two types of diabetes insipidus?

A

o central/cranial -> absense or lack of circulating vasopressin

o nephrogenic -> end-organ (kindeys) resistence to vasopressin

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

What are the causes of cranial diabetes insipidus?

A

o acquired (more common) -> damage to neurohypophysial system

  • traumatic brain injury
  • pituitary surgery
  • pituitary tumours, craniopharyngioma
  • metastasis to the pituitary gland - e.g. breast
  • granulomatous infiltration of median eminence - e.g. TB, sarcoidosis

o Congenital – rare

  • usually receptor gene mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What are the causes of nephrogenic diabetes insipidus?

A
  • familial (rare)
  • drugs -> LITHIUM (used to treat bi-polar disorder), dimethyl chlortetracycline (DMCT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What are the signs and symptoms of diabetes insipidus?

A
  • polyuria
  • very dilute urine (hypo-osmolar)
  • thirst and increased drinking (polydipsia)
  • dehydration (and consequences) if fluid intake is not maintained
  • possible disruption of sleep with associated problems
  • possible electrolyte imbalance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What is the cycle of diabetes insipidus?

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

What happens to the usual cycle of diabetes insipidus if the patient doesnt have access to water?

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

Name a disease which presents with similar symptoms to DI?

A
  • psychogenic polydipsia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What is the cycle of patients suffering from psychogenic polydipsia?

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

How can DI and psychogenic polydipsia be distinguished between?

A

o normal person has a tightly maintained osmolality (270-290 mOsm/kg H2O)

o diabetes insipidus = high osmolality

o psychogenic polydipsia = low osmolality

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

What is the ‘gold standard’ test to diagnose diabetes insipidus?

A

o fluid deprivation test

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

What are the biochemical features of DI?

A
  • hypernatraemia
  • raised urea
  • increased plasma osmolality
  • dilute (hypo-osmolar) urine - ie low urine osmolality
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

State the biochemical features of psychogenic polydipsia.

A
  • mild hyponatraemia -> excess water intake
  • low plasma osmolality
  • dilute (hypo-osmolar) urine -> ie low urine osmolality
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What is the treatment for cranial DI?

A
  • V2 receptor agonists -> desmopressin (DDAVP)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What is the clinical use of terlipressin?

A
  • is V1 receptor agonist -> used to treat GI bleeds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

How is desmopressin administrated?

A
  • nasally or orally
  • produces a prompt sustained decrease in urine volume and increase in urine osmolarity -> has a very long effect so you don’t need to take desmopressin too often
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

Apart from cranial DI, what else can desmopressin be used to treat?

A
  • nocturnal eneuresis (bedwetting)
  • haemophilia - you need V2 receptor stimulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What are the side effects of desmopressin?

A
  • fluid retention and hyponatraemia
  • abdominal pain
  • headaches
  • nausea
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What is the treatemnt for nephrogenic diabetes insipidus?

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

Define SIADH.

A
  • plasma vasopressin concentration is INAPPROPRIATE for the existing plasma osmolarity -> too much ADH for current osmolarity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

Show the pathway of SIADH.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What are the signs and symptoms of SIADH?

A

o Signs of SIADH:

  • raised urine osmolarity
  • decreased urine volume (initially)
  • hyponatraemia - decrease in plasma sodium concentration due to increased water reabsorption

o Symptoms of SIADH:

  • often asymptomatic
  • when Na+ concentration falls < 120mM = generalised weakness, nausea and poor mental function
  • when Na+ conc. falls <110mM = confusion, coma and death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are the causes of SIADH?

A
  • tumours (ectopic secretion)
  • neurohypophysial malfunction (e.g. meningitis, cerebrovascular disease)
  • pulmonary disease (e.g. pneumonia)
  • malignancy (in particular small cell lung cancer)
  • endocrine disease (e.g. Addison’s disease)
  • physiological - it can happen under quite normal circumstances where AVP release is stimulated by non-osmotic stimuli (e.g. hypovolaemia, pain, surgery)
  • drugs (e.g. chlorpropamide)
  • idiopathic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What is the treatment of SIADH?

A
  • treat the cause (e.g. tumour)
  • if someone is already hyponatraemic, you need to deal with that as quickly as possible: -> immediate = fluid restriction and longer-term = drugs that prevent vasopressin action in the kidneys (e.g. lithium, dimethyl chlortetracycline (DMCT), V2 receptor antagonists)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are Vaptans?

A
  • non-peptide vasopressin analogues
  • tolvaptan - V2 receptor antagonist

o can be used for SIADH but are very expensive so are rarely used

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What is another disease (not SIADH) that vaptans could be used for)

A
  • congestive heart failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What drugs affect ADH secretion?

A
  • increase = nicotine
  • decrease = alcohol and glucocorticoids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What are the symptoms of primary hypothyroidism?

A

o Tongue gets thick

o Speech slows down

o Deepening of the voice

o Basal Metabolic Rate falls

o Bradycardia

o General weakness

o Depression

o Cold intolerance

o Weight gain and reduced appetite

o Constipation

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

What happens to TSH in primary hypothyroidism?

A
  • it goes up to try and stimulate the thyroid
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What enzyme converts T4 to T3?

A
  • deiodinase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What percentage of hormones are made in the thyroid?

A
  • 80% is deiodination of T4
  • 20% is direct thyroidal secretion of T3
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

Describe the mechanism of thyroid hormones.

A
  • thyroxine enters the target cell and is converted to T3 by deiodinase (can also be T3 that enters directly)
  • T3 moves to the nucleus and binds to the thyroid hormone receptor and then heterodimerises with a Retinoid X Receptor
  • this complex then binds to the Thyroid Response Element that causes a change in gene expression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What is used in thyroid hormone replacement therapy?

A
  • the usual thyroxine replacement is levothyroxine sodium or direct thyroxine (T4)
  • rarely liothyroxine sodium is used as a T3 replacement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What is levothyroxine sodium used to treat?

A
  • primary hypothyroidism - autoimmune or iatrogenic (post-thyroidectomy or post-radioactive iodine)
  • secondary hypothyridsim (pituitary tumour, post-pituitary surgery/radiotherapy)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What is used as reference to thyroxine dose in primary hypothyroidism?

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

What is used as a reference for thyroxine dose in secondary hypothyroidism?

A
  • isn’t any TSH (or is very low) so middle of T4 reference range is used
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is the clincal use of liothyronine?

A
  • to treat MYXOEDEMA COMA - a very rare complication of hypothyroidism
  • IV liothyronine (T3) is given in this situation because the onset of action is faster than T4
  • then when the patient has recovered from the coma oral thyroxine replacement is given
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

As well as being expensive, name another reason why T3 hormone replacement therapy is not favoured.

A
  • T3 is very potent -> very difficult to get the dose right
  • high T3 switches off TSH and patients show thyrotoxicosis symptoms = palpatations, tremors and anxiety
  • combined thyroid hormone replacement does exsist but again isn’t favoured
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

How does T3 and T4 circulate in the blood plasma?

A
  • 99.97% of T4 and 99.7% of T3 are bound to thyroxine binding globulin (TBG) (not to be confused with thyroglobulin in the colloid of follicular cells)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What factors affect plasma protein production?

A
  • increase in pregnancy and with prolonged treatment with oestrogens and phenothiazides

decreased with liver disease or if you are severely malnourished

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

Name two drug classes that can compete for binding sites of TBG.

A
  • phenytoin
  • salicylates (e.g. aspirin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the most common cause of hypothyroidism?

A
  • autoimmune -> 5% of people suffer from this in their life
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are the adverse effects of thyroid hormone over-replacement?

A

o usually associated with a low/suppressed TSH

o Skeletal -> increased bone turnover, reduction in bone mineral density therefore risk of osteoporosis

o Cardiac -> tachycardia, risk of Dysrrhythmia (particularly atrial fibrilation)

o Metabolism - increased energy expenditure, weight loss

o Increase b-adrenergic sensitivity -> tremors, nervousness

ARE ALL SYMPTOMS OF THYROTOXICOSIS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q
A
  • D
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q
A
  • D
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What is Cushing’s Syndrome?

A
  • too much cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What are the clinical features of Cushing’s Syndrome?

A
  • centripetal obesity
  • moon face
  • buffalo hump
  • proximal myopathy
  • hypertension
  • hypokalaemia
  • red striae
  • thin skin and bruising
  • osteoporosis
  • diabetes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What are the causes of Cushing’s Syndrome?

A

o exogenous

  • taking too many steroids

o endogenous

  • pituitary dependent cushing’s disease (pituitary produces too much ACTH causing adrenal growth therefore too much cortisol)
  • ectopic ACTH from lung cancer
  • adrenal adenoma secreting cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What is the pattern of cortisol release?

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

Compare the release of cortisol throughout the day between a normal person and someone with Cushing’s Syndrome.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What are the methods of diagnosing Cushing’s Syndrome?

A
  • 24h urine collection for urinary free cortisol
  • blood diurnal cortisol levels
  • low dose dexamethasone suppression test
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

Describe the low dose dexamethasone suppression test.

A
  • 0.5mg of dexamethasone (artificial steroid) every 6hrs for 48hrs
  • normally this suppress cortisol to 0
  • in any cause of Cushing’s Syndrome this will fail to suppress it to 0

o GOLD STANDARD

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

Barring surgery, what treatments are available for Cushing’s Syndrome?

A
  • inhibitors of sterid biosynthesis -> metyrapne, ketoconazole
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

Barring surgery, what treatments are available for Conn’s Syndrome?

A
  • mineralocorticoid receptor (MR) antagonists -> spironolactone, epleronone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What is the mechanism of metyrapone?

A
  • inhibition of 11beta-hydroxylase therefore blocking steroid synthesis in the zona fasciculata
  • causes ACTH to rise as no cortisol is made so no negative feedback
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What are the clinical scenarios in which metyrapone is used?

A

o Control of Cushing’s syndrome prior to surgery

  • cushing’s patients are not good surgical candidates because they are predisposed to infection and have very thin skin
  • metyrapone is used to improve the patient’s symptoms and promotes better post-op recovery (better wound healing, less infection)
  • oral metyrapone is dosed according to cortisol production (aim for mean serum cholesterol of 150-300 nmol/L)

o Control of Cushing’s symptoms after radiotherapy

  • radiotherapy is usually quite slow to work so you would give metyrapone after radiotherapy until the effects of the radiotherapy start to come about
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

State two biological/mechanical negative aspects about metyrapone.

A

o accumulation of 11-deoxycorticosterone which has mineralcorticoid properties -> sodium retention and potassium secretion -> salt retention -> hypertension

o all precursors are channeled towards sex steroid synthesis -> increase in adrenal androgens -> hirsutism and acne (more unpleasant for women is general)

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

What are the side effects of metyrapone?

A
  • nausea, vomiting, dizziness
  • sedation
  • hypoadrenalism
  • hypertension on long-term administration
  • hirsuitism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

How does ketoconazole function?

A
  • inhibits Cytochrome P450 side chain cleavage enzymes meaning that glucocorticoid, mineralocorticoid and sex steroid production is blocked
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What are the uses of ketoconazole?

A
  • treatment and control of symptoms prior to surgery
  • an anti-fungal (not licensed anymore due to its reduction on cortisol)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What are the side effects of ketoconazole?

A
  • nausea, vomiting
  • abdominal pain
  • alopecia
  • gynaecomastia, oligospermia, impotence, decreased libido -> due to reduction of sex steroid production
  • ventricular tachycardias
  • hepatotoxicity -> can be fatal - patients are monitored with regular liver function tests
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

Where in the adrenal gland is aldosterone made?

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

What is Conn’s Syndrome?

A
  • too much aldosterone
  • due to a benign adrenal cortical tumour in the zona glomerulosa
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What are the features of Conn’s Syndrome?

A
  • hypertension and hypokalaemia (low potassium)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

How is Conn’s Syndrome (aka primary hyperaldosteronism) diagnosed?

A
  • high aldosterone and low renin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What is spironolactones mechanism of action?

A
  • is converted into several metabolites including canrenone which competitively antagonises MRs -> blocks the action of aldosterone (sodium reabsorption and potassium secretion in the kidney tubules)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

When is spironolactone used?

A
  • prior to surgery to make the patient a better candidate
  • people who have bilateral adrenal hyperplasia -> can’t take both adrenals out as not enough cortisol or aldosterone would be produced
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

Name the side effects of spironolactone.

A

o spironolactone is a very non-specific drug so there are lots of side-effects

  • a progesterone receptor agonist so causes menstrual irregularities
  • an androgen receptor antagonist so it can cause gynaecomastia in men -> clinically can’t be used in men for this reason (must use much more commonly used epleronone)
  • GI tract irritation
  • renal and hepatic disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What makes epleronone a better drug than spironolactone?

A
  • has the same mechanism with same affinity for MRs while being more specific so doesn’t interfere with progesterone or androgen receptors so has less side effects
  • FAR MORE COMMONLY USED
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What is Phaeochromocytoma?

A
  • tumours of the adrenal MEDULLA which secrete catecholamines (adrenaline and noradrenaline)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What are the effects of phaeochromocytoma?

A
  • adrenaline has a rapid effect -> from time to time (more likely after trauma) the tumour will release a lot of adrenaline -> massive rise in bp (e.g. 300/150) -> to the sort of level that cause sudden strokes or MIs - MAIN EFFECT
  • is a sudden onset of panic, you feel anxious, tachycardia and severe hypertension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What are the clinical features of phaeochromocytoma?

A
  • hypertension in a young person
  • episodic severe hypertension (after abdominal exam - press on adrenal causing release of NA and A)
  • more common in certain inherited conditions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What makes the diagnoses of phaeochromocytoma a medical emergency?

A
  • severe hypertension -> MI or Stroke
  • high adrenaline -> Ventricular Fibrillation (VF) -> Sudden Cardiac Death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What is the treatment/management of phaeochromocytoma?

A
  • first is alpha blockade - patients may need intravenous fluid as alpha blockade commences
  • second is beta blockade added to prevent tachycardia
  • once adrenalines effects have been nullified surgery to remove the gland
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

State some key statistics about the positioning and status of phaeochromocytoma tumours.

A
  • 10% extra-adrenal (sympathetic chain) and 90% inside the adrenal
  • 10% are malignant
  • 10%are bilateral making 90% are curable by operation
  • phaeochromocytoma is EXTREMELY RARE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What hormones are present in the hypothalamo-pituitary-adrenal axis?

A
  • CRH = corticotrophin releasing hormone
  • ACTH = adrenocorticotrophic hormone (corticotrophin)
  • aldosternone, cortisol, adrenaline and noradrenaline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

Where does all steroid comes from?

A
  • cholesterol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What is made in the zona glomerulosa?

A
  • aldosterone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

What is made in the zona fasciculata?

A
  • cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

What is produced in the adrenal medulla?

A
  • adrenaline and noradrenaline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

Describe the pathway that leads to aldosterone and cortisol production?

A
  • after each number adding hyrdoxylase gives you the enzyme
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

Globally, what is the most common cause of adrenal failure?

A
  • TB Addison’s Disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

In the UK, what is the most common cause of adrenocortical failure?

A
  • Autoimmune Addison’s Disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

Barring Addison’s Disease, name a cause for adrenocortical failure?

A
  • congenital adrenal hyperplasia -> caused by enzyme deficiency so hormones aren’t made properly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

What are the consequences/symptoms of adrenal failure?

A
  • fall in blood pressure -> no aldosterone
  • loss of salt in urine -> unable to retain salt because of the lack of aldosterone -> salt loss in urine and potassium plasma
  • fall in glucose -> due to glucocorticoid deficiency
  • high ACTH (no -ve feedback) resulting in increased pigmentation with some vitilgo-> ACTH is cleaved from POMC -> the remaining part is MSH (melanocyte stimulating hormone)

o eventual death due to severe hypotension -> if this happens suddenly it is called an Addisonian Crisis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What tests can be done to diagnose Addison’s?

A
  • low 9am cortisol and high ACTH at any time in blood tests
  • unresponsive/reduced amount of cortisol produced to a big dose of synthetic ACTH (synacthen)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What is the results of a typical Addison’s patient to tests for Addison’s?

A
  • 9am cortisol = 100 (normal = 270-900)
  • 9.30, post IM injection of synacthen = 150 (normal = >600)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

What is the commonest cause of congenital adrenal hyperplasia?

A
  • 21-hydroxylase deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

How long will a baby with complete 21-hydroxylase deficiency survive without treatment?

A
  • less than 24 hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

What happens to the hormone balance in someone with complete 21-hydroxylase deficiency?

A
  • no aldosterone and cortisol
  • lots of sex steroids, particularly testosterone, because all pro-hormones are channelled down that pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What is the age of presentation of congenital adrenal hyperplasia?

A
  • as a neonate with salt losing Addisonian crisis - floppy baby
  • before birth if a previous child had suffers from CAH
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What is an important feature to notice immediately after birth due to congenital adrenal hyperplasia?

A
  • ambiguous genitalia in girls due to the excess testosterone -> virilisation - fusion of labia into a scrotum like structure
  • if this is noticed they are keep in hospital to prevent arrest
  • can be prevented by giving the mother some steroid in pregnancy if there is a known risk of 21-hydroxylase deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What occurs in partial 21-hydrxylase deficiency?

A
  • low levels of cortisol and aldosterone but crucial enough to survive so they present at any age
  • excess sex steroid -> often misdiagnosed as PCOS or other similar disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What are the main problems with partial 21-hydroxylase deficiency?

A
  • boys = precocious/early false puberty
  • girls = hirsutism, virilisation and acne
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

What are the consequences of 11-beta-hydroxylase deficiency?

A
  • acts as an aldosterone receptor agonist so behaves as if there is too much aldosterone -> hypertension and hypokalaemia
  • sex hormones are also in excess so virilisation, hirsutism etc
  • lack of cortisol and aldosterone -> aldosterone issues dont present due to reasons above
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

What happens to the hormone balance in 17-hydroxylase deficiency?

A
  • low cortisol and sex steroids
  • 11-deoxycorticosterone and aldosterone are high
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

What are the presentations of 17-hydroxylase deficiency?

A
  • never go through puberty -> usually present around pubertal age
  • hypertension without addisonian crisis because they have aldosterone
  • borderline hypoglycaemia
  • lots of infections because you need cortisol to cope with the stress of infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

What is made in the zona reticularis?

A
  • androgens and oestrogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

Which adrenal hormone is not under ACTH control? State what controls its release.

A
  • aldosterone -> release is stimulated by renin in the renin-angiotensin system
  • hyperkalaemia, hyponatraemia, drop in renal blood flow and beta-1 adrenoceptor stimulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

Compare glucocorticoid and minerocorticoid receptors?

A
  • GR = wide distribution, selective for glucocorticoids, low affinity for cortisol
  • MR = descrete distribution to the kidney, don’t destingiush between aldosterone and cortisol, high affinity to cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

How come cortisol doesn’t usually activate MR receptors despite being able to do so?

A
  • 11b-hydroxysteroid dehydrogenase deactivates cortiol into cortisone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

Why is hypokalaemia seen in Cushing’s Syndrome?

A
  • the 11b-hydroxysteroid dehydrogenase system becomes overwhelmed so potassium is excreted due to activation of MRs
  • the same mechanism cause hypertension by absorbing sodiuma nd water
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

Describe the structure of hydrocortisone.

A
  • cortisol and hydrocortisone have the same structure, it’s just that cortisol is endogenous and hydrocortisone is synthetic - can be used to treat Addison’s disease/crisis
  • at high doses it can cause MR activation because it overwhelms the 11b-hydroxysteroid dehydrogenase system just like cortisol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

Describe prednisolone.

A
  • a glucocortcoid with weak mineralocortioid activity
  • tends to be used as an immunosuprressive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

Describe dexamethasone.

A
  • very POTENT glucocorticoid -> used clinically for things like brain metastases where there is a lot of oedema -> used as an acute anti-oedema agent
  • NO mineralocorticoid effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

Describe fludrocortisone.

A
  • an aldosterone analogue -> used as an aldosterone substitute in Addison’s disease
  • NOT a glucocorticoid

-

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

What is the method of administration for corticosteroids?

A
  • all can be given orally
  • in acute situations they are given parenterally (IV or IM)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

Compare the biological activity of the corticosteroids.

A
  • binding to plasma proteins (corticosteroid binding globulin and albumin)
  • hydrocortisone is about 90-95% bound
  • prednisolone is less bound and dexamethasone and fludrocortisone are even less bound
  • fludrocortisone is only bound to albumin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

State and explain the difference in treatment between patients with primary and secondary adrenocortical failure.

A
  • primary = hydrocortisone and fludrocortisone
  • secondary = hydrocortisone -> is a porblem with the pituitary and not adrenal so aldosterone isn’t affect as renin-angiotensin system is fine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What is the treatment for Addisonian crisis?

A
  • FIRSTLY = 0.9% NaCl for rehydration
  • then high dose of hydrocortisone (IV or IM)
  • 5% dextrose if hypoglycaemic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

How is congenital adrenal hyperplasia treated?

A
  • cortisol replacement -> hydrocortisone or dexamethasone
  • replace aldosterone -> fludrocortisone
  • suppress ACTH -> reduced adrenal androgen production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

How is therapy for CAH monitored?

A

o17a-hydroxyprogesterone levels

o clinical assessments of patients complaintst:

  • cushingoid - glucocorticoid dose too HIGH
  • hirsuitism/acne - glucocorticoid dose too LOW (so ACTH is high)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

Name some times in which hydrocortisone/cortisol mimicing drugs dosage should be increased.

A
  • when the patient is vunerable to stress
  • minor illness = x2 normal dose
  • surgery = IV or Im hydrocortisone in the build up to the generla anaesthetic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q
A
  • C
231
Q

Describe the hypothalamo-pituitary-testes axis.

A
232
Q

What are the 3 phases of the menstrual cycle?

A
  • Follicular Phase
  • Ovulation
  • Luteal Phase
233
Q

Describe the hypothalamo-pituitary-ovarian axis in the follicular phase.

A
  • LH stimulates production of oestradiol and progesterone in the ovaries
  • FSH stimulates follicular development and inhibin
  • by around day 10, the leading follicle develops into a Graffian Follicle
  • oestrogen initially has negatively inhibits LH and FSH secretion
  • so in the follicular phase their HPG axis is basically the same as men
234
Q

Describe the hypothalamo-pituitary-ovarian axis in the luteal phase.

A
  • once the oestrogen levels reach a certain point, it switches from negative feedback to positive feedback
  • it increases GnRH release and increases LH sensitivity to GnRH -> leads to a mid-cycle LH surge -> triggers ovulation from the leading follicle
  • if implantation does NOT occur, the endometrium is shed (menstruation), but if implantation does occur you have pregnancy
235
Q

Define infertility in the clinical environment.

A
  • inability to conceive after 1 year of regular uprotected sex
236
Q

How common is infertility and in what percentage of causes are the abnormalities in males and females?

A
  • 1/6 couples are affected
  • males = 30% - females = 45% - unknown = 25%
237
Q

What happens to the hormone balance in someone with primary gonadal failure?

A
  • high GnRH
  • high LH and FSH
  • low testosterone/oestradiol
238
Q

What happens to the hormone balance in someone with secondary gonadal failure?

A
  • low FSH and LH
  • low testosterone/oestradiol
  • could also be a low GnRH if it hypothalamic
239
Q

State 5 clinical features of male hypogonadism.

A

· Loss of libido

· Impotence

· Small testes

· Decreased muscle bulk

· Osteoporosis (testosterone has an anabolic action in the bone)

240
Q

What are the causes of male hypogonadism?

A
  • hypothalamic-pituitary disease
  • primary gonadal disease
  • hyperprolactinaemia
  • adrogen receptor deficiency
241
Q

Name some hypothalamic-pituitary diseases which lead to infertility.

A
  • Hypopituitarism -> pituitary is not working properly -> secondary gonadal failure
  • Kallman’s Syndrome (anosmia (lack of smell) and low GnRH) -> GnRH neurones migrate forwards with olfactory neurones hence link
  • Illness/underweight -> due to low levels of leptin -> body knows you’re underweight -> it’s not an appropriate time to reproduce)
242
Q

State some causes of primary gonadal disease.

A
  • congenital: Klinefelter’s syndrome
  • acquired: testicular torsion, chemotherapy
243
Q

What tests can be ran to investigate male hypogonadism?

A
  • LH, FSH and testosterone blood tests -> if all low then pituitary (look at an MRI)
  • prolactin -> check for hyperprolactinaemia
  • sperm count and motility
  • chromosomal analysis -> Klinefelter’s (XXY)
244
Q

Compare the terms azoospermia and oligospermia.

A
  • Azoospermia = absence of sperm in ejaculate
  • Oligospermia = reduced numbers of sperm in ejaculate
245
Q

What treatments are available for male hypogonadism?

A

o replacement testosterone for all patients -> increase their muscle bulk and protect against osteoporosis

o for fertility (if hypothalamic/pituitary disease) -> subcutaneous gonadotrophins (LH and FSH) to stimulate testosterone release (don’t want to switch off pituitary functions so can’t give straight testosterone because that will have negative feedback) -> induces spermatogenesis

o dopamine agonist for hyperprolactinaemia -> main hypothalamic influence over prolactin release and it has a negative effect on prolactin release

246
Q

What are the endogenous sites of androgen production?

A

· Interstitial Leydig cells of the testes

· Adrenal cortex (males and females)

· Ovaries

· Placenta

· Tumours

247
Q

What are the main actions of testosterone?

A

· Development of the male genital tract

· Maintains fertility in adulthood

· Control of secondary sexual characteristics

· Anabolic effects (muscle, bone)

248
Q

How does testosterone circulate in the body?

A

o 98% is protein bound

249
Q

What happens to testosterone in certain tissues?

A

o in different tissues you get testosterone being converted to other things

  • 5 a-reductase converts testosterone to dihydrotestosterone (DHT), in muscle and bone, which then acts on androgen receptors (AR)
  • aromatase, found in brain and adipose tissue, can convert testosterone to 17b-Oestradiol (E2) which acts via the oestrogen receptors (ER) -> mechanism which may explain obesity causing infertility
  • DHT and E2 receptors act via nuclear receptors -> are intracellular -> have to go into the nucleus to have an effect
250
Q

Name 4 clinical uses of testosterone.

A

o testosterone in adulthood will increase:

  • lean body mass
  • muscle size and strength
  • bone formation and bone mass (in young men)
  • libido and potency

o it will NOT restore fertility -> requires treatment with gonadotrophins to restore normal spermatogenesis

251
Q

What are the 3 categories of infertility in women?

A

· Amenorrhoea

· Polycystic Ovarian Syndrome (PCOS)

· Hyperprolactinaemia

252
Q

Define primary amenorrhoea.

A
  • failure to develop spontaneous menstruation by the age of 16 years -> likely to be an abnormal puberty -> could be congenital
253
Q

Define secondary amenorrhoea.

A
  • absence of menstruation for 3 months in a woman who has previously had cycles
254
Q

Define oligomenorrhoea.

A
  • irregularly long cycles
255
Q

What are the main causes of amenorrhoea?

A

o PREGNANCY

o Lactation -> progesterone suppresses menstration

o Ovarian Failure:

  • premature ovarian insufficiency (early menopause)
  • ovariectomy
  • chemotherapy
  • ovarian dysgenesis (Turner’s Syndrome (45 X)) - lacking an X chromosome
256
Q

What are the features of Turner’s Syndrome?

A
  • thick neck tissue with further swelling (cystic hygroma)
  • heart and kidney issues
  • short stature
  • cubitus valgus (where the forearm is angled away from the body to a greater degree than normal when fully extended)
  • gonadal dysgenesis (defective development)
257
Q

Name some of the rarer causes of amenorrhoea.

A

o Hypothalamic/pituitary disease

o Kallmann’s syndrome

o Low BMI

o Post-pill amenorrhoea (downregulates the hypothalamus and pituitary) -> if you use the pill for a long time then periods don’t come back for around 12 months after cessation of use -> why it is advised that you stop using the pill every 4 years

o Hyperprolactinaemia

o Androgen excess: gonadal tumour

258
Q

What tests can be conducted to diagnose/find the cause of amenorrhoea?

A
  • Pregnancy Test
  • LH, FSH and Oestradiol -> hard due to changes in natural cycle
  • Day 21 Progesterone -> in the second half of the menstrual cycle the progesterone rises over a prolonged period of time -> there should be a rise in progesterone around this time to show that they are ovulating
  • Prolactin -> hyperprolactinaemia
  • Thyroid function test -> hyper- and hypothyroidism can cause problems with periods
  • Androgen blood test (testosterone, androstenedione, DHEAS)
  • Chromosomal Analysis (Turner’s)
  • Ultrasound Scan Ovaries/Uterus
259
Q

Describe the treatment for amenorrhoea.

A
  • Treat the cause (e.g. low weight)
  • Primary Ovarian Failure = Hormone Replacement Therapy
  • Hypothalamic/pituitary disease -> HRT for oestrogen replacement but for FERTILITY = gonadotrophins (LH and FSH) -> part of IVF treatment
260
Q

What female infertility is associated with an increased risk of CVD and diabetes?

A
  • PCOS
261
Q

What is the criteria for PCOS diagnosis?

A

o Need TWO of the following:

  • polycystic ovaries on the ultrasound scan
  • oligoovulation/anovulation
  • clinical/biochemical androgen excess
262
Q

What are the clinical features of PCOS?

A

· Hirsutism – male pattern hair growth

· Menstrual cycle disturbance

· Increased BMI makes the symptoms worse

263
Q

What is the treatment for PCOS?

A
  • Metformin -> insulin sensitiser used in type II diabetes
  • Clomiphene -> anti-oestrogenic effect in the hypothalamo-pituitary axis -> binds to oestrogen receptors in the hypothalamus thereby blocking the negative feedback -> results in an increase in GnRH and gonadotrophin secretion -> kick-starts the HPG axis for short periods for fertility
  • Gonadotrophin therapy as part of IVF treatment
264
Q

Describe the control of prolactin secretion.

A
  • dopamine has a negative effect on prolactin release, is the main hypothalamic hormone controlling prolactin release
  • TRH has a mild stimulatory effect
  • prolactin stimulates the production of milk in lactating women
  • if it becomes dysregulated it will switch off gonadal function via LH actions on the ovaries and testes -> natural contraceptive just after birth
  • also reduces GnRH pulsatility so that it is released basally all the time rather than in regular pulses
265
Q

What are the cause of hyperprolactinaemia, highlight the main ones?

A
  • DOPAMINE ANTAGONISTS -> anti-emetics (metoclopramide) or anti-psychotics (phenothiazines) **
  • PROLACTINOMA **
  • COMPRESSION ON THE STALK DUE TO A PITUITARY ADENOMA **
  • hypothyroidism
  • PCOS
  • eestrogens (oral contraceptive pill), pregnancy, lactation
  • idiopathic
266
Q

What are the clinical features of hyperprolactinaemia?

A
  • galactorrhoea (discharge unrelated to breastfeeding)
  • reduced GnRH secretion?LH action leads to hypogonadism
  • if its due to prolactinoma -> headaches and visual field defects
267
Q

Describe the treatment for hyperprolactinaemia.

A

o Treat the cause - e.g. stop the drugs if that’s what’s causing it if it is possible (e.g. anti-psychotics can’t always be stopped, it’s a balancing act)

o Dopamine Agonists -> bromocriptine or cabergoline -> also causes a decrease in the size of the tumour if it is being caused by a prolactinoma

o Pituitary surgery but is rarely needed (because drugs usually work)

268
Q
A
  • 3
269
Q
A
  • 1 -> prolactin switches off GnRH pulsitivity so FSH and LH would be low as well as oestradiol
270
Q

Define menopause.

A
  • permanent cessation of menstraution due to a lack of ovarian follicular activity
271
Q

Define climacteric.

A
  • the periiod of transition in which menstraution become irregular
272
Q

What is the average age of menopause?

A
  • 51 -> (45-55)
  • before 40 is deemed to be premature
273
Q

State the symptoms of the menopause.

A
  • hot flushes
  • urogenital atrophy and dyspareunia (difficult or painful sexual intercourse) -> due to vaginal atrophy
  • sleep disturbance
  • depression
  • decreased libido
  • joint pain

o symptoms usually diminish/disappear with time

274
Q

What are the hormonal chages that occur during meopause?

A
  • levels of oestradiol and inhibin B fall due to follicular atresia -> less negative feedback so gonadotrophin levels rise
  • a 55-year old women is expected to have high LH and FSH
275
Q

What are the two major complication of the menopause?

A

o osteoporosis - caused by oestrogen deficiency -> oestrogen is an anabolic hormone -> in osteoporosis there is less matrix and that predisposes these patients with oestrogen deficiency to osteoporotic fractures

  • 10-fold increase in the risk of fracture in a post-menopausal woman

o Cardiovascular Disease - oestrogen is protective against CVD before menopause

  • women have the same risk as men after the age of 70
276
Q

What is given as HRT therapy?

A
  • usually oestrogen + progesterone -> can’t just give oestrogen as is increases the risk of endometrial carcinoma (progesterone inhibits the proliferation)
  • if the patient has had a hysterectomy then oestrogen only can be given
277
Q

How can oestrogen be prepared?

A

o Oral oestradiol

o Oral conjugated equine oestrogen

o Transdermal (patch) oestradiol

o Intravaginal

278
Q

How often is HRT given?

A
  • Cyclical -> oestradiol every day and for the last 12-14 days you take some progesterone
  • Continuous Combined -> a little bit of oestrogen and a little bit of progesterone every day
279
Q

When is HRT prescribed?

A
  • for disabling hot flushes
280
Q

How well is oestrogen absorbed?

A
  • very well absorbed but it has a low bioavailabitility due to extensive first pass metabolism
281
Q

How is the issue of oestrogens low bioavailability overcome?

A
  • oestrone sulphate (conjugated oestrogen)
  • ethinyl oestradiol: semi-synthetic oestrogen -> form that’s used in oral contraceptives

o provides protection from first pass metabolism

282
Q

What are the side effects of HRT?

A
  • Breast Cancer
  • Venous Thromboembolism (VTE)
  • Stroke
  • Gallstones

o absolute risk of complications for healthy symptomatic postmenopausal women in their 50s taking HRT for five years is very low but there is a relative risk

o MUST consider the circumstance of the patient e.g. family history of breast cancer

283
Q

Explain the link between HRT and coronary heart disease.

A
  • there is an increased risk of CHD with HRT
  • younger women had no problem, they had no increased risk but those who were in their 60s and started HRT, had an increased risk of CHD
  • it is about the timing of exposure -> older patients who are started on HRT have a higher risk of CHD due to baseline disease

Back

284
Q

What are the benefits of oestrogen and how does it change with HRT?

A
  • lipid profile and endothelial function however, synthetic progestins negates these beneficial effects
285
Q

Describe Tibolone.

A
  • synthetic prohormone which has oestrogenic, progestogenic and weak androgenic effects
  • reduces the risk of fracture
  • increase the risk of STROKE (RR: 2.2) and possibly breast cancer
286
Q

Describe Raloxifene.

A

o Selective Oestrogen Receptor Modulator (SERM)

o it is tissue selective:

  • bone - it has oestrogenic effects = reduces the risk of fracture
  • breast and uterus - it has anti-oestrogenic effects = this reduces the risk of breast cancer

o however, raloxifene is associated with an increased risk of fatal stroke and VTE

o HRT used to be prescribed to women as a first line treatment of osteoporosis but it is NOT any more because you have other treatments like oral bisphosphonates which are not associated with a risk of breast cancer -> however, if someone has menopausal symptoms then HRT is the treatment of choice

287
Q

Describe Tamoxifen.

A
  • anti-oestrogenic on breast tissue

  • used to treat oestrogen-dependent breast tumours and metastatic breast cancers
288
Q

What are the causes of premature ovarian insufficiency (early menopause)?

A
  • occurs to 1% of women

o Autoimmune

o Surgery

o Chemotherapy

o Radiation

289
Q

Describe the combined oral contraceptive pill?

A
  • oestrogen in the oral contraceptive is often ethinyl oestradiol
  • progestogen in the pill is levonorgestrel or norethisterone
  • take it for 21 days and stop for 7 days
290
Q

What is the mechanism of the combined oral contraceptive

A
  • suppresses the hypothalamus and pituitary (both E and P do this)
  • progesterone also thickens cervical mucous (this makes it more difficult for the sperm to pass through)
291
Q

When is progesterone only contraception given?

A
  • when oestrogens are contra-indicated - this is if there is a risk of THROMBOSIS -> oestrogen has pro-coagulant effects so progesterone is used instead
  • less effective than the combined contraceptive but if there are contra-indications (such as in a smoker) then the progesterone only contraceptive is the best option
292
Q

What is very important about administration of progesterone only contraception?

A
  • must be taken at the same time every day due to a short half-life
293
Q

What is a long acting progesterone only contraception?

A
  • mirena coil
294
Q

Name 3 emergency contraceptives.

A
  • copper IUD
  • levonorgestrel
  • ulipristal
295
Q

What is the mechanism of the copper IUD?

A
  • affects sperm viability and function -> inhibits fetilisation
296
Q

When is copper IUD the prefferred option?

A
  • in the overweight as its effectiveness isn’t compromised
297
Q

What is levonorgestrel?

A
  • a high progesterone dose
298
Q

How does ulipristal function?

A

o Anti-progestin activity

o Delay ovulation by as much as 5 days

o Impairs implantation

299
Q

How long after coiteus can emergency contraception be taken?

A
  • copper IUD = 5-7 days
  • levonorgestrel = 72 hours
  • ulipristal = 5 days
300
Q

How many sperm reach the ovum?

A
  • 1 in a million manages the journey of 100,000 times the length of a sperm
301
Q

How is oestrogen made in men?

A
  • testosterone is converted to oestrogen by aromatase
302
Q

How is most tubular fluid reabsorbed?

A
  • within the rete testis and early epididymis under control of oestrogen
303
Q

What elements of the epididymal fluid is under the influence of androgens?

A
  • energy for the journey
  • coating of the surface of the spermatozoon
304
Q

Which location within the male reproductive tract has the highest concentration of spermatoza?

A
  • vas deferens concentration of spermatozoa is higher than later on in the reproductive tract once other fluids have been added
305
Q

Where is cut when performing a vasectomy?

A
  • vas deferens towards the bottom end
306
Q

How many spermatoza are in the average ejaculate?

A
  • 15-120x106/ml (average has 2-5 ml of seminal fluid
307
Q

What are the elements of ejaculate?

A
  • spermatoza
  • seminal fluid
  • leucocyte
  • potentially viruses -> hepatitis B, HIV etc
308
Q

Where does seminal fluid come from?

A
  • mainly = accessory sex glands = prostate, bulbourethral gland and the seminiferous tubules
309
Q

Where does capacitation of sperm occur?

A
  • achieving fertilisation capacity takes place in the ionic and proteolytic environment of the fallopian tube
310
Q

What changes occur in the capacitation of sperm?

A
  • loss of glycoprotein coat
  • change in surface membrane characteristics
  • develop whiplash movement of tail
311
Q

What is the capacitation of sperm dependent on?

A
  • oestrogen
  • calcium
312
Q

Describe the acrosome reaction.

A
  • spermatozoon binds to the ZP3 glycoprotein on the zona pellucida -> once bound to ZP3, progesterone stimulates calcium influx into the spermatozoon
  • results in the calcium-dependent acrosome reaction
  • enables an exposed spermatozoon recognition site to bind to a second glycoprotein (ZP2) -> once ZP2 has bound, the acrosome releases its enzymes allowing penetration of the zona pellucida so that the head of the spermatozoon can enter the ovum

Back

313
Q

Where does fertilisation occur?

A
  • fallopian tube
314
Q

What are the results of fertilisation?

A

o leads immediately into zonal reaction

  • cortical granules release molecules which degrade the zona pellucida (including ZP2 and ZP3) -> prevents further binding of other sperm
  • also calcium dependent

o explusion of the second polar body

315
Q

Describe the development of a conceptus?

A
  • conceptus continues to divide as it moves down the fallopian tube to the uterus (3-4 days)
  • until implantation, the developing conceptus receives its nutrients from uterine secretions
  • cell division continues you will have a ball of cells with the outside cells receiving the nutrients but the inner cells will be receiving less and less nutrients
  • the free-living phase will last about 9-10 days
316
Q

In what stage of the menstraul cycle does development of the conceptus occur?

A
  • luteal phase -> oestrogen and progesterone will be high
317
Q

What follows the formation of a conceptus?

A
  • compactation into 8-16 cell morula
  • followed by blastocyst formation
318
Q

What does a blastocyst become?

A

o Inner cell mass - becomes the embryo

o Trophectoderm - becomes the chorion (which becomes the placenta)

319
Q

What assists the transfer of the blastocyst into the uterus ?

A
  • increasing progesterone : oestrogen ratio in luteal phase
320
Q

What are the two stages of implantation?

A
  • attachment phase -> outer trophoblast cells make contact with the uterine surface epithelium -> establises a system for getting the nutrients from the mother to all the cells within the embryo
  • decidualisation phase -> deeper implantation into the underlying uterine stromal tissue occuring within a few hours
321
Q

What is the process of implantation dependent on?

A
  • progesterone domination in the presence of oestrogen
322
Q

What two molecules are particularly important in the attachment phase?

A

o Leukaemia Inhibitory Factor (LIF) - from endometrial secretory glands -> stimulates attachment of blastocyst to the endometrial cells

o Interleukin-11 (IL-11) - from endometrial cells -> released into the uterine fluid

323
Q

Describe what changes occur due to decidualisation.

A

o endometrial changes due to progesterone

  • glandular epithelial secretion
  • glycogen accumulation in stromal cell cytoplasm
  • growth of capillaries
  • increased vascular permeability
324
Q

What factors are required for decidualisation?

A
  • mainly IL-11
  • histamine
  • certain prostaglandins
  • TGFb - promotes angiogenesis
325
Q

What hormonal changes occur during pregnancy?

A
  • hCG (produced in the placenta) is vital for the first 6 weeks at least as its purpose is to replace LH which will be low due to high oestrogen and progesterone -> in a normal menstrual cycle, the oestrogen and progesterone levels will eventually fall and you’ll get menstruation
  • to maintain the stimulation of oestrogen and progesterone, hCG is being produced by the trophoblast cells -> hCG binds to LH receptors on the corpus luteum hence replacing the effects of LH
  • after about 5 weeks or so, the placenta will have taken over the production of hormones so the ovaries are no longer necessary
  • oestrogen and progesterone levels increase throughout pregnancy and right until the end, progesterone remains the DOMINANT influence
326
Q

Describe oestrogen production in pregnancy.

A
  • mother provides the precursors which tends to be pregnenolone which leads to progesterone -> progesterone is then going to lead to steroid production by the foetus (in the foetal adrenals)
  • maternal AND foetal adrenals produce a precursor (which is an androgen) - dehydroepiandrosterone sulfate (DHEAS)
  • 16a-hydroxy-DHEAS is then taken up by the placenta to produce OESTRIOL
  • DHEAS is also used by the placenta to produce small amounts of oestrone and oestradiol
327
Q

What is the main oestrogen in pregnancy?

A
  • oestriol
  • molecule for molecule, weaker than oestradiol but it is produced in large amounts during pregnancy
  • source of oestriol is placenta
328
Q

Describe a hormonal test that can be done to check the general health of a feotus.

A
  • looking at the ratio of oestriol : oestradiol and oestrone or the ratio of oestriol : total oestrogens and you see a change
329
Q

What changes in maternal hormones occur during pregnancy?

A
  • hGH (human growth hormone) decreases as the placental hGH-variant increases towards term
  • kiss peptin also increases 1000s of times to act as a natural contraception by down regulating the axis
330
Q

Describe the endocrine control of lactation.

A
  • suckling stimulates neural pathways to the hypothalamus and on to the pituitary
  • the neurohypophysis secretes oxytocin and the adenohypophysis secretes prolactin
  • prolactin = promotion of milk synthesis
  • oxytocin = promotoin of milk ejection
331
Q

Describe the hormonal control of parturition.

A
  • parturition is all about contraction of actin and myosin filaments which requires CALCIUM -> to initiate contraction you need to increase the intracellular calcium concentration
  • oestrogen stimulates production of prostaglandins by the endometrial cells -> prostaglandins stimulates the production and release of calcium into the cytoplasm from intracellular stores

o Main Point: OESTROGEN tends to INCREASE the chance of contraction

  • progesterone has the OPPOSITE EFFECT TO OESTROGEN -> inhibits prostaglandin synthesis and oestrogen receptors -> keeps the effects of oestrogen under control
  • once the foetus reaches a certain size, the production of steroids is switched from the production of progesterone to the production of oestrogen (oestrogen dominates)
  • at parturition, oxytocin will be released which will bind to its receptor and open calcium channels allowing calcium ions to move in from outside

Back

332
Q

Summarise calcium homeostasis.

A

o interactive system between PTH and vitamin D

o as serum calcium falls, you get an increase in PTH (parathyroid hormone)

  • PTH mobilises calcium from bone to increase plasma calcium
  • direct effect on the kidneys to increase reabsorption of calcium in the kidneys
  • stimulates an increase in plasma calcium indirectly via the activation of vitamin D (calcidiol from liver to calcitriol/active vitamin D)

o calcitriol then has two main effects to increase serum calcium:

  • increased calcium absorption in the intestines
  • increased calcium mobilisation in bone
333
Q

What is the mechanism of action of calcitriol?

A
  • act on oesteoblasts to cause them to release RANKL -> RANKL activates osteoclasts
334
Q

Name two hormones that inhibit Na+ and PO43- reabsorption in the kidneys.

A
  • PTH
  • FGF23
335
Q

What are the actions of FGF23?

A
  • inhibit Na+ and PO43- transporter in the kidneys
  • negative feedback on calcitriol -> less PO43- reabsorption from the gut
336
Q

Which enzyme activates 25 OH-D3 into calcitriol (1,25 (OH)2D3)?

A
  • renal 1 alpha-hydroxylase
  • stimualted by PTH
337
Q

What are the actions of calcitriol?

A
  • Ca2+ absorption in the gut
  • Ca2+ maintenance in the bone
  • increased renal Ca2+ reasbsorption
  • ve feedback on PTH
338
Q

Name some causes of Vitamin D deficiency.

A
  • diet
  • lack of sunlight
  • gastrointestinal malabsorptive states -> liver disease, coeliac, IBD
  • renal failure
  • receptor defects (autosomal recessive)
339
Q

What are the resulting symptoms of Vitamin D deficiency?

A

o Softening of bone - you are unable to bear the weight of the rest of the body so you get the characteristic bowing of the legs

o Bone deformities

o Bone pain

o Severe proximal myopathy

340
Q

What are the signs and symptoms of hypocalcaemia?

A
  • convulsions
  • arrhythmias
  • tetany
  • parasthesia (hands, mouth, feet, lips)

o CATs go Numb

341
Q

What is the general trend caused by hypocalcaemia?

A
  • sensitises excitable tissues -> muscle cramps/tetany, tingling
342
Q

What two signs are checked for if hypocalcaemia is suspected?

A
  • Chvostek’s
  • Trousseau’s
343
Q

What is Chvostek’s sign?

A
  • tap the facial nerve just below the zygomatic arch
  • POSITIVE response = twitching of the facial muscles
  • indicates neuromuscular irritability due to hypocalcaemia
344
Q

What is Trousseau’s sign?

A
  • inflation of the blood pressure cuff for several minutes induces carpopedal spasm (aka neuromuscular irritability due to hypocalcaemia)
  • very painful so isn’t done for long
345
Q

Name some causes of hypocalcaemia.

A
  • Vitamin D deficiency
  • hypoparathyroidism (low PTH) -> surgical, auto-immune, magnesium deficiency
  • PTH resistance (e.g. pseudohypoparathyroidism)
  • renal failure -> impaired 1a-hydroxylase -> decreased production of calcitriol
346
Q

What are the signs and symptoms of hypercalcaemia?

A
  • stones - renal effects -> polyuria + thirst can go onto cause nephrocalcinosis (deposition of calcium in the kidneys), renal colic, chronic renal failure
  • abdominal moans - GI effects -> anorexia, nausea, dyspepsia, constipation, pancreatitis
  • psychic groans - CNS effects -> fatigue, depression, impaired concentration, altered mentation, coma (the last three only tend to occur when calcium concentration is > 3 mmol/L)

o Stones, Moans and Groans

347
Q

State some causes of hypercalcaemia.

A

The first two causes are responsible for 90% of cases of hypercalcaemia

  • PRIMARY HYPERPARATHYROIDISM -> e.g. due to a parathyroid adenoma which becomes too busy and produces excessive amounts of PTH
  • MALIGNANCY - TUMOURS/METASTASES -> bone metastases where increased bone turnover causes an increase in serum calcium plus tumours can also produce PTH-related peptide
  • conditions with a high bone turnover -> hyperthyroidism, Paget’s Disease
  • Vitamin D excess (rare)
348
Q

What is the diagnostic approach to hypercalcaemia?

A
  • normally, there is negative feedback on PTH by calcium so if serum calcium rises, PTH will decrease
  • in primary hyperparathyroidism a parathyroid adenoma secretes PTH autonomously and is not responding to negative feedback

o Calcium - HIGH

o PTH - HIGH (unsuppressed)

  • malignancy is a nother major cause but serum results show low PTH with high calcium -> check for bony metastases
349
Q

What is Vitamin D deficiency reffered to as?

A

o In children = Rickets

o In adults = Osteomalacia

350
Q

What diagnostic results would be found in the serum of a patient with Vitamin D deficiency?

A
  • plasma 25-hydroxycholecalciferol = usually LOW
  • plasma calcium = LOW
  • PTH = HIGH -> secondary hyperparathyroidism that can make plasma calcium appear normal
  • plasma phosphate = LOW
351
Q

What is the treatment for Vitamin D deficiency?

A

o In patients with normal renal function -> 25-hydroxy vitamin D (inactive) - either as ergocalciferol (D2) or cholecalciferol (D3)

  • patient can convert this to calcitriol via 1a hydroxylase

o In patients with renal failure -> active Vitamin D (alpacalcidol)

  • don’t have their own 1a hydroxylase activity, so they can’t activate 25-hydroxy vitamin D preparations
352
Q

What are causes of Vitamin D excess?

A

o excessive treatment with active metabolites of vitamin D -> only due to the potency of alfacalidol -> lots and lots of Vitamin D is required to be intoxicating

o Granulomatous Disease - e.g. sarcoidosis, leprosy and tuberculosis

  • macrophages in the granuloma tissue can convert 25-hydroxycholecalciferol to the active metabolite 1,25-dihydroxycholecalciferol
353
Q

Define oesteoblast.

A
  • synthesise osteoid and participate in mineralisation/calcification of osteoid (bone formation)
354
Q

Define osteoclast.

A
  • release lysosomal enzymes which breakdown bone (bone resorption)
355
Q

What is RANK ligand?

A
  • an important molecule that activates osteoclasts from precursor form -> more mature osteoclasts = more bone resorption
356
Q

What is the mechanism of RANKL?

A
  • RANKL expressed on osteoblast surface
  • RANKL binds to RANK-R to stimulate osteoclast formation and activity
  • osteoblasts express receptors for PTH and calcitriol -> regulate balance between bone formation & resorption
357
Q

Describe bone structure.

A
  • cortical (hard) bone -> outer area
  • trabecular (spongy or trabecular) bone -> inner area

o both formed in a lamellar pattern = collagen fibrils laid down in alternating orientations, mechanically strong

358
Q

What is meant by woven bone?

A
  • bone with disorganised collagen fibrils -> far weaker than healthy bone
359
Q

What is the general problem on bones caused by Vitamin D deficiency?

A
  • inadequate mineralisation of newly formed bone matrix (osteoid) -> bone remains newly formed and therefore soft
360
Q

How does rickets affect bones?

A
  • CHILDREN

o affects cartilage of epiphysial growth plates and bone

o skeletal abnormalities (bowed legs) and pain, growth retardation, increased fracture risk

361
Q

How does osteomalacia affect bones?

A
  • ADULTS

o after epiphyseal closure

o skeletal pain, increased fracture risk, proximal myopathy

362
Q

Describe the kind of fractures that patients who are Vitamin D deficient suffer from.

A
  • stress fractures -> normal stresses on abnormal bone cause insufficiency fractures, particularly in Looser Zones -> these include the pelvis and tibia which leads to a Wide Waddling gait
363
Q

What are the effects of renal failure on bone disease?

A
  • decreased renal function = decrease in the production of calcitriol and phosphate excretion so plasma phosphate increases
  • the decrease in calcitriol production will cause a decrease in calcium absorption from the intestines while phosphate plasma concentration rises both leading to hypocalcaemia
  • hypocalcaemia will stimulate PTH release -> PTH will break down bone matrix in order to try and restore the blood calcium concentration to the normal level -> hypocalcaemia decreases bone mineralisation
  • combination of the increased bone resorption and decreased bone mineralisation will lead to osteitis fibrosa cystica
  • imbalance between calcium and phosphate you can get some of it forming the main salts which can be deposited in extra-skeletal tissue causing vascular calcification
364
Q

What are Brown Tumours?

A
  • radiolucent bone lesions due to osteoclastic bone resorption due to high PTH -> very suspectible to fractures
  • feature of osteitis fibrosa cystica
365
Q

What is the treatment for osteitis fibrosa cystica?

A
  • hyperphosphataemia -> low phosphate diet and phosphate binders to reduce GI phosphate absorption
  • alphacalcidol -> e.g. calcitriol analogues
  • parathyroidectomy in tertiary hyperparathyroidism
366
Q

Define osteoporosis - include the clinical cut off point?

A
  • a condition of reduced bone mass (fewer trabeculae) and a distortion of bone microarchitecture which predisposes to fracture after minimal trauma
  • clinically having a bone mineral density (BMD) that is 2.5 standard deviations (SD) or more below the average value for young healthy adults
367
Q

How is bone mineral density measured?

A
  • in a DEXA -> dual energy x-ray absorptiometry
  • measures in femoral neck and lumbar spine
368
Q

Name some conditions that predispose you to osteoporosis.

A

o Post-menopausal Oestrogen Deficiency

o Age-related deficiency in bone homeostasis (men and women)

o Hypogonadism in young men and women

o Endocrine conditions -> cushing’s syndrome, hyperthyroidism, primary hyperparathyroidism

o Iatrogenic -> prolonged use of glucocorticoids, heparin

369
Q

What treatment options are available to patients suffering form osteoporosis?

A
  • Oestrogen (HRT)/Selective Oestrogen Receptor Modulators
  • Bisphosphonates
  • Denusomab
  • Teriparatide
370
Q

How does oestrogen work to treat osteoporosis in post-menopausal women?

A
  • anti-resoptive effect on bone -> prevents bone loss
371
Q

Why is everyone post-menopausal women not given HRT as a preventative measure against osteoporosis?

A

o increased risk of breast cancer

o venous thromboembolism

o women with an intact uterus need additional progestogen to prevent endometrial hyperplasia/cancer

372
Q

Why can selective oestrogen receptor modulators?

A

o tissue-selective ER ANTAGONISTS/anti-oestrogens -> e.g. TAMOXIFEN

  • has oestrogenic activity in the bone -> however, it has oestrogenic effects on the endometrium which limits its use in osteoporosis
  • tissue-selective ER AGONISTS -> e.g. RALOXIFENE
  • has selectivity oestrogenic activity in bone and anti-oestrogenic activity in breast and uterus
  • isn’t as widely used due to risk of venous thromboembolism and stroke

No action

Mouse Gestures

Back

Forward

Scroll up

Scroll down

Switch to previous tab

Switch to next tab

Close all tabs except current

Close current tab

Open new tab

Close all tabs

Refresh current tab

Stop loading

Scroll to bottom

Scroll to top

Reopen closed tab

Go to home page

373
Q

What is the main first-line treatment for osteoporosis?

A
  • bisphosphonates
374
Q

What is the mechanism of bisphosphonates in osteoporosis treatment?

A
  • binds avidly to hydroxyapatite and is ingested by osteoclasts -> impairs the ability of osteoclasts to resorb bone and decreases maturation of osteoclasts from their precursors
  • possibly promotes osteoclast apoptosis

o NET RESULT = REDUCED BONE TURNOVER

375
Q

What are the clinical uses of bisphosphonates?

A

o osteoporosis - first line treatment

o malignancy -> particularly helpful when associated to hypercalcaemia or to reduced bone pain from metastases

o paget’s disease -> reduces bony pain

o severe hypercalcaemic emergency -> after IV saline to REHYDRATE

376
Q

What are the side effects of bisphophonates?

A
  • oesphagitis if given orally -> may require switch from oral to IV preparations
  • flu-like symptoms -> often this is limited to the first dose
  • osteonecrosis of the jaw -> greatest risk in cancer patients receiving IV bisphosphonates
  • atypical fractures -> might reflect over-suppression of bone remodelling in prolonged bisphosphonate use
377
Q

What is the mechanism of denosumab?

A
  • is a human monoclonal antibody
  • binds to RANKL and inhibits osteoclast formation and activity -> inhibits osteoclast-mediated bone resorption
378
Q

How is denosumab administrated?

A
  • subcutaneously every 6 or 12 months
379
Q

What is second-line treatment for osteoporosis?

A
  • denosumab
380
Q

How does teriparatide work as an osteoporosis treatment?

A
  • is a recombinant PTH (of 34 AA)
  • increases bone formation and resorption but FORMATION OUTWEIGHS RESORPTION
381
Q

Define Paget’s Disease.

A
  • very active (increased), localised but disorganised bone metabolism - usually slowly progressive
382
Q

Describe what occurs in Paget’s Disease.

A
  • excessive bone resorption occurs due to osteoclastic overactivity -> this is followed by compensatory increase in bone formation (osteoblasts)
  • causes new bone formation = WOVEN bone -> structurally disorganised and mechanically weaker than normal adult lamellar bone -> leads to bone frailty, hypertrophy and deformity
383
Q

What characterises Paget’s disease?

A
  • abnormal, large osteoclasts in excessive numbers
  • a disease of the elderlt -> only appears after 50-60
  • men and women are affected equally
  • common in the UK -> maybe genetic
384
Q

What bones are most commonly affected in Paget’s disease?

A

· Pelvis

· Femur

· Spine

· Skull (associated with hearing loss)

· Tibia

385
Q

What are the clinical features of Paget’s disease?

A
  • bone pain
  • increased vascularity -> warmth over affected bones
  • deafness -> cochlear involvement
  • radiculopathy -> due to nerve compression
  • multiple fractures
  • increased bone width
386
Q

How is Paget’s disease diagnosed?

A

o High Plasma Alkaline Phosphatase (ALP) - calcium is normal

o Radiology demonstrating variable features -> loss of trabecular (spongy/cancellous) bone, increased density, deformity

o Radioisotope (Technetium) scanning can be performed to indicate the areas of involvement

387
Q

State what treatments are available for Paget’s disease.

A
  • bisphosphonates – very helpful for reducing bony pain and disease activity
  • simple analgesia
388
Q

What is the arcuate nucleus?

A
  • key area of the brain (within the hypothalamus) involved in food intake regulation
  • is a circumventricular organ - meaning that it has an incomplete blood-brain barrier thus allowing access to peripheral hormones -> allows the arcuate nucleus to integrate peripheral and central feeding inputs
389
Q

What are the two neuronal populations of the arcuate nucleus?

A

o Stimulatory -> NPY + Agrp neurons

o Inhibitory -> POMC neurons

390
Q

What are the two key hypothalamic nuclei in appetite regulation?

A
  • arcuate nucleus
  • paraventricular nucleus
391
Q

Describe the melanocortin system.

A
  • under normal conditions, the POMC, from arcuate nucleus, will be broken down to a-MSH which is an endogenous agonist of the MC4R (paraventricular nucleus) -> the reason why you aren’t hungry all the time is because of this a-MSH tone
  • you trigger hunger by the release of Agrp from the arcuate nucleus which is an endogenous antagonist of MC4R
  • when you need to eat, you increase Agrp activity, block the inhibitory signal of a-MSH and stimulates food intake
392
Q

What CNS mutation occur in humans that affects appetite?

A
  • POMC and MC4 receptor
  • NO NPY or Agrp
393
Q

Describe the ob/ob mouse.

A
394
Q

What is missing in the ob/ob mouse?

A
  • Ob genes codes for leptin
395
Q

Where is leptin released from?

A
  • white adipose tissue -> tells brain how much fat storage there is and therefore regulates eating
396
Q

Name diseases caused by mutations which lead to obesity.

A
  • leptin deficiency
  • Prader-Willi Syndrome
397
Q

What happens if leptin is administrated to a patient who suffers from leptin deficiency?

A
  • decreased food intake
  • increased thermogenesis/energy expenditure
398
Q

What is the mechanism of leptin?

A
  • activates POMC and inhibits NPY/Agrp neurons
399
Q

Why can’t leptin be used as a weight control drug?

A
  • most fat people have high leptin but a lot are leptin resistant
400
Q

What is proportional to insulin levels?

A
  • body fat
401
Q

What are the different effects of insulin?

A

o Chronically - reduce body fat

o Acutely - if you have a big glucose load, you should suppress having more sugar

402
Q

What is the largest endocrine gland/organ in humans?

A
  • the GI tract
  • releases over 20 hormones
403
Q

What does CCK do?

A
  • causes contraction of the gall bladder
404
Q

What is ghrelin also known as?

A
  • the hunger hormone
405
Q

Where is ghrelin released from?

A
  • the stomach
406
Q

Describe the structure of ghrelin.

A
  • 28 amino acids long
  • fatty acid is found on the 3rd amino acid
407
Q

What enzyme converts ghrelin into its active form?

A
  • ghrelin O-acyltransferase (GOAT)
408
Q

How does ghrelin affect appetite?

A
  • increases appetite -> stimulates Agrp/NPY neurones and inhibits POMC neurones
409
Q

Where is PYY secreted from?

A
  • L-cells in the distal small intestine
410
Q

How does PYY 3-36 affect appetite?

A
  • PYY 3-36 is released post-prandially and is dependent on the size of the meal -> decreases appetite -> inhibits NPY release and stimulates POMC
  • has the opposie effect of ghrelin
411
Q

Where is GLP-1 secreted from?

A
  • L-cells in the distal small intestine
412
Q

What is GLP-1?

A
  • a gut hormone (from L-cells) coded for by the pre-propglucagon gene releazed post-prandially
  • has incretin effects -> increases insulin levels
  • reduces food intake
413
Q

What are the clinical uses of GLP-1 based drugs?

A
  • GLP-1 based drug is important in the treatment of diabetes mellitus
  • GLP-1 only stimulates glucose-induced insulin release which is good because it means that it only stimulates insulin release when it’s needed -> isn’t going to cause hypoglycaemia
414
Q

How long is GLP-1 active for?

A
  • a very short amount of time -> quickly deactivated by DP-4
  • drugs have to be altered so they have a longer half-life
415
Q

What can saxenda be used to treat?

A
  • licensed by FDA and EMEA as a diabetes and obesity treatment
416
Q

Describe how saxenda works.

A
  • long-acting GLP-1 receptor agonist (liraglutide) with a tweak structure -> it is more resistant to degradation which also stops it from being cleared from the circulation giving it a much longer half-life
417
Q

Name some co-morbidities associated with obesity.

A
418
Q

What is the thrifty gene hypothesis?

A
419
Q

What is the adaptive drift hypothesis?

A
  • there used to be a normal distribution of body weight
  • ones that were too light will die/not be able to reproduce
  • ones that were too heavy would get eaten
  • eventually, we got better at defending against predators and so excess bodyweight became a neutral change
420
Q

State some ambiguity that occurs when diagnosing diabetes.

A
  • autoimmune (Type I) diabetes leading to insulin deficiency can present after decades of life -> this is Latent Autoimmune Diabetes in Adults (LADA)
  • T2DM can present in childhood (linked to an increasing prevalence of childhood obesity)
  • people with T2DM can present with diabetic ketoacidosis (though this is more common in T1DM)
  • monogenic diabetes can present phenotypically as Type 1 or Type 2 diabetes (e.g. MODY, mitochondrial diabetes)
  • diabetes can present if someone has a problem with the pancreas -> e.g. pancreatitis
  • diabetes can also present in endocrine diseases
421
Q

Name the three endocrine diseases which are particularly linked to diabetes?

A

o Phaeochromocytoma

o Cushing’s Syndrome

o Acromegaly

  • all of these cause hyperglycaemia
422
Q

What is the current classification of diabetes?

A
423
Q

What is the pathogenesis of T1DM?

A
  • various environmental and genetic triggers and regulators which come into play and lead to destruction of beta cells
  • eventually, auto-antibodies are produced against beta cells -> patients will then go on to lose their first phase insulin
424
Q

What is measured as a marker for insulin?

A
  • C peptide
425
Q

What does the relapsing-remitting theory of T1DM suggest?

A
  • over time the beta cells reduce, then stabilise then reduce again -> a theory suggests that this is due to an imbalance between effector T cells and T-regulatory cells
  • as time passes the effector T cells increase in number and the T-regulatory cells decrease
  • much like the mechanism of MS
426
Q

What is the importance to T1DM autoimmune basis?

A
  • increased prevalence of other autoimmune disease -> most commonly B12 deficiency and coeliac disease as well as others e.g. rheumatoid arthritis, thyroid disease, fertility problems
  • risk of autoimmunity in relatives
427
Q

How can the autoimmune basis of T1DM be useful clinically?

A
  • auto-antibodies can be clinically useful (check the progress of the disease and to confirm the autoimmune basis of the diabetes)
  • immune modulation offers the possibility of novel treatments
428
Q

What allele has a susceptibility to T1DM?

A
  • HLA-DR on chromosome 6
  • various DR alleles -> particularly DR3 and DR4
  • DR2 has a protective effect
429
Q

What markers can be measured to clinically define someone as having T1DM?

A

o two most significant markers

  • islet cell antibodies (ICA) - group O human pancreas
  • glutamic acid decarboxylase antibodies (GADA)

o other two antibodies:

  • insulin antibodies (IAA)
  • insulinoma-associated-2 autoantibodies (IA-2A)-receptor like family
430
Q

What are the symptoms of T1DM?

A
  • polyuria
  • nocturia -> passing lots of urine at night
  • polydipsia
  • blurring of vision
  • thrush -> due to increased risk of infections
  • weight loss
  • fatigue
431
Q

What are the signs of T1DM?

A
  • dehydration
  • cachexia
  • hyperventilation -> due to metabolic acidosis -> try to blow off CO2 -> is called Kussmaul breathing
  • smell of ketones
  • glycosuria
  • ketonuria
432
Q

What does insulin do?

A

o insulin has a negative effect on:

  • hepatic glucose output
  • protein breakdown in the muscle
  • glycerol being taken out from the fatty tissue into the periphery

o insulin has a positive effect on:

  • glucose being taken up by the muscle
433
Q

What is the general trend in patients who are insulin deficient?

A
  • lots of glucose goes into the circulation and isn’t utilised/taken up by tissues
434
Q

Name some hormones that increase hepatic glucose output.

A
  • catecholamines
  • cortisol
  • glucagon
  • growth hormone
435
Q

What is the mechanism of diabetic ketoacidosis?

A
  • glucose isn’t taken up into cells, due to insulin deficiency, and utilised so a lot of our energy comes from fatty acids -> lipids in the adipocytes is broken down
  • normally, you get glycerol coming out of the adipocytes and going to the liver but in the case of insulin deficiency, you get FATTY ACIDS coming out of the adipocytes into the circulation -> fatty acids then go to the liver where they are converted to ketones (this process is normally inhibited by insulin)

o Key Point: as you are deficient in insulin, you get a lot more ketone bodies being produced by the liver

436
Q

What does a large amount of ketones in the body define?

A
  • insulin deficiency - T1DM
437
Q

What are the aims of treatment of T1DM?

A
  • reduce early mortality
  • avoid acute metabolic decompensation
  • prevent long-term complications

o TYPE 1 DIABETICS REQUIRE EXOGENOUS INSULIN TO PRESERVE LIFE

438
Q

State some long-term complications of T1DM.

A

o Retinopathy

o Nephropathy

o Neuropathy

o Vascular Disease

439
Q

What diet recommendations are suggested to patients with T1DM?

A
  • reduce calories as FAT
  • reduce calories as REFINED CARBOHYDRATES
  • increase calories as COMPLEX CARBOHYDRATES
  • increase SOLUBLE FIBRE
  • balance distribution of food over the course of a day with regular meals and snacks
440
Q

Describe insulin treatment for those who are insulin deficient.

A

o With Meals

  • short acting insulin
  • human insulin
  • insulin analogues

o Background Insulin - 50% of insulin requirement is a basal form

  • long acting
  • non-C bound to zinc or protamine
  • insulin analogues
441
Q

Describe insulin pumps.

A
  • provides continuous insulin delivery as well as allowing patients to give dose after eating food (replaces subcutaneous method -> pre-programmed basal rates and a bolus for meals
  • does NOT measure blood glucose, so it can’t form a feedback loop and completely replace the beta cell function
442
Q

For patients that insulin replacement that fails to control severe hypoglycaemic attacks, what other treatment are available?

A

o Islet Cell Transplants

  • islet cells are harvested, isolated and injected into the liver
  • a high risk of rejection, so patients must be on immunosuppressants for life
443
Q

How can treatment of T1DM be monitored?

A
  • capillary monitoring
  • HbA1c
444
Q

Describe capillary monitoring.

A
  • capillary glucose levels can be measured by pricking the finger tips -> is reflective of venous blood glucose but isn’t as accurate
  • patients can titrate their insulin does based on the capillary glucose reading
445
Q

Describe HbA1c.

A
  • used for long-term blood glucose control is monitored by measuring HbA1c
  • more glucose that is present in the blood, the more the haemoglobin is glycosylated -> there is a good correlation between plasma glucose over a reasonably long period of time and HbA1c
446
Q

For how long does HbA1c show a marker for?

A
  • 3 months -> RBC lifespan is 120 days
447
Q

What is the target HbA1c for T1DM patients?

A
  • <7%
448
Q

What is linked to lower HbA1c?

A
  • lower risk of complications -> particularly microvascular complications
449
Q

When does HbA1c not work?

A
  • the measurement relies on haemoglobin -> anything causing an increased turnover of haemoglobin (e.g. haemolytic anaemia or haemoglobinopathy) means the HbA1c may not be accurate
450
Q

What are the acute complications of T1DM?

A

o Main Consequence = HYPERGLYCAEMIA

  • reduced tissue glucose utilisation
  • increased hepatic glucose production

o METABOLIC ACIDOSIS

  • circulating acetoacetate and hydroxybutyrate -> results from increased ketone body production in the liver
  • osmotic dehydration and poor tissue perfusion -> hypotension
451
Q

Are hypos completely avoidable in T1DM?

A
  • no -> they are inevitable as a result of treating T1DM
  • is a major source of anxiety in patients and relative
452
Q

Define hypoglycaemia.

A
  • a plasma glucose <3.6mmol/L
453
Q

Define severe hypoglycaemia.

A
  • any hypoglycaemic event that requires another person to treat it
454
Q

At what glucose level are mental processes impaired?

A
  • <3mmol/L
455
Q

At what glucose level does consciousness become impaired?

A
  • <2mmol/L
456
Q

What are the consequences of severe hypoglycaemia?

A
  • arrhythmia and sudden death
  • possible long-term effects on the brain
457
Q

What are the dangers of recurrent hypos?

A
  • loss of warning signs -> hypoglycaemia unawareness -> associated with poorly controlled diabetes
458
Q

Why do diabetic patients become hypoglycaemic?

A

o Unaccustomed exercise

o Missed meals

o Inadequate snacks

o Alcohol (if they have too much then they can become unaware of the hypoglycaemia)

o Inappropriate insulin regime

459
Q

What are the signs and symptoms of hypoglycaemia?

A

o due to increased autonomic activation

  • palpitations/tachycardia
  • tremor
  • sweating
  • pallor/cold extremities
  • anxiety

o due to imparied CNS function

  • drowsiness
  • confusion
  • altered behaviour
  • focal neurology
  • coma
460
Q

How is hypoglycaemia treated?

A

o if the patient is conscious -> feed the patient

  • glucose -> rapidly absorbed as solution or tablet
  • after some recovery complex carbs -> maintains blood glucose after initial treatment

o if unconsciousnessly impaired -> parenternal

  • IV dextrose
  • 1mg glucagon IM
  • avoid concentrated solutions if possible
461
Q

What defines diabetes?

A
  • a fasting blood glucose > 7 mmol/L
462
Q

What is impaired fasting glucose?

A
  • between 6 and 7 mmol/L -> between normal and diabetes
  • has some complications but not those to microvascular
463
Q

What factors influence the pathophysiology of T2DM?

A
  • genetics
  • intra-uterine environment
  • adult environment
464
Q

What is the genetics behind maturity onset diabetes of the young (MODY)?

A
  • autosomal dominant
  • several hereditary forms
  • strong family history
465
Q

Is MODY associated with obesity?

A
  • no
466
Q

What are some possible mechanisms of MODY?

A
  • ineffective pancreatic beta cell insulin production
  • mutations of transcription factor genes and glucokinase genes
467
Q

What is the treatment of MODY?

A
  • specfic to the the type of MODY -> solve that problem and therefore help the MODY
468
Q

Name some risk factors to T2DM.

A
  • genetics - studies show its almost autosomal dominant
  • obesity
  • intra-uterine growth restriction
469
Q

When is microvascular disease found?

A
  • diabetes with hyperglycaemia
470
Q

What does insulin resistance cause?

A
  • dyslipidaemia and stimulation of the mitogenic pathway -> causes smooth muscle hypertrophy and hypertension -> macrovascular disease
471
Q

What is the relationship between age and insulin resistance and secretion?

A

o we all make less and less insulin as we grow older

o at the same time, we become more and more insulin resistant

o insulin resistance and insulin secretion lines will intersect -> beyond this point we won’t be able to make enough insulin for our insulin resistance

  • in caucasians, this intersection normally occurs around 110 years of age, however in other ethnic groups it intersects much sooner
472
Q

What occurs in the stages of insulin secretion?

A
  • 1st phase -> stored insulin is released immediately
  • 2nd phase -> over time more insulin is produced and released
473
Q

Describe the deteriotation of insulin secretion with progression of diabetes.

A
  • patients developing diabetes will still have some insulin production but they will lose their 1st phase insulin response -> they can make insulin eventually but it takes a longer time for them to do it -> can be got around by eating complex carbs which release the glucose more slowly thus reducing the need for a first phase insulin response
  • over time insulin will stop being made and both 1st and 2nd phase will be lost -> insulin deficiency sets in
474
Q

How is insulin serum levels maintained after a period of not eating?

A
  • hepatic glucose output maintains blood glucose at 4 mmol/L
475
Q

How does insulin effects hepatic glucose output?

A

o reduces hepatic glucose output

  • after eating, insulin stops HGO because you don’t need this output from the liver once you’ve just eaten while driving the glucose into muscle and adipose tissue

o both of these effects are absent in T2DM -> there is insufficient insulin to inhibit hepatic glucose output and insufficient insulin to move glucose into muscle and fat

476
Q

Describe the effects of insulin resistance on metabolism.

A
  • adipocytes are full of triglycerides which can be broken down to glycerol and non-esterified fatty acids (NEFA)
  • insulin would stop this breakdown of triglycerides because there is no need to break down fat stores after you’ve had a meal -> glycerol and NEFA travel to the liver
  • in the liver, glycerol can be used to make glucose - gluconeogenesis and can also be released from the liver via glycogenolysis -> insulin resistance means that there is increased hepatic glucose output and decreased glucose uptake into tissues
  • the fatty acids go to the liver, are chopped up into 2-carbon segments and CANNOT be used to make glucose -> instead used to make very low density lipoprotein triglycerides which are ATHEROGENIC -> contributes to the atherogenic profile of insulin resistant subjects -> patients can have atheroma years before high blood glucose
477
Q

What is waist circumference a predictive marker for and why?

A

o ischaemic heart disease

  • breakdown of triglycerides are a particular markers for omental adipocytes
478
Q

What is the link betwen diabetes and obesity?

A
  • more than a percipitant -> fatty acids and adipocytokines are important -> OBESITY APPEARS TO BE PART OF THE MECHANISM OF T2DM
  • adipocytokines modulate the insulin resistance
  • central or omental obesity are the biggest problems
  • losing wight is a beneficial treatment
479
Q

How is the gut microbiome linked to obesity, insulin resistance and T2DM?

A
  • may be important through signalling to the host -> various lipopolysaccharides are fermented by the gut bacteria to short chain fatty acids -> can enter the circulation and modulate bile acids and therefore modulate host metabolsim
  • also appear to be important in inflammation and are involved in adipocytokine pathways
  • microbiota transplants are being investigated as a treatment for obesity
480
Q

What is the most common side effect of diabetes treatment?

A
  • weight gain
481
Q

Which is the only diabetes treatment that causes weight reduction?

A
  • metformin
482
Q

What are the presentations of T2DM?

A
  • osmotic symptoms
  • infections
  • screening test
  • upon presentation of a complication -> acute such as hyperosmolar coma or chronic such as IHD and retinopathy
483
Q

What is the basic management of T2DM?

A
  • education
  • diet
  • pharmacological treatment
  • complication screening
484
Q

Why treat T2DM?

A
  • can help the symptoms of T2DM
  • reduces the chance of acute metabolic complications - though these are unlikely in T2DM
  • reduce the chance of long-term complications; good evidence base
  • education -> people generally only treat things if they have symptoms so it’s important to educate them about the consequences of poor treatment
485
Q

What diet should take place in patients with T2DM?

A
  • control total calories/increase exercise (weight)
  • reduce refined carbohydrate (less sugar)
  • increase complex carbohydrate (more rice etc)
  • reduce fat as proportion of calories (less IR)
  • increase unsaturated fat as proportion of fat (IHD)
  • increase soluble fibre (longer to absorb CHO)
  • address salt (BP risk)
486
Q

What is monitored in T2DM treatment?

A
  • weight
  • glycaemia
  • blood pressure
  • dyslipidaemia
487
Q

What pharmalogically treatments are available for T2DM?

A
  • metformin -> used to treat insulin resistance in the liver
  • orlistat -> pancreatic lipase inhibitor -> stops the break down of fat thus restricting fat absorption in the gut
  • sulphonylureas -> makes the existing pancreas secrete more insulin
  • alpha glucosidase inhibitor -> delays glucose absorption
  • thiazolidinediones -> acts on the adipocytes and addresses insulin resistance peripherally in fat and muscle
  • SGLT2 inhibitors -> increases glucose excretion by the kidneys
488
Q

What is metformin?

A
  • a biguanide (class of drugs used as oral anti-hyperglycaemic drugs) -> an insulin sensitiser
  • used in more or less everyone with T2DM, in particular, overweight patients with T2DM where diet alone has not succeeded
489
Q

What are the effects of metformin?

A

o reduces insulin resistance

  • reduces hepatic glucose output
  • increase peripheral glucose disposal

o side effects are uncommon but are GI related

490
Q

When can metformin not be used?

A
  • in patients with -> severe liver failure, severe cardiac failure, mild or worse renal failure
491
Q

What is the mechanism of sulphonylureas in diabetes treatment?

A
  • block the ATP sensitive potassium channel and cause the influx of calcium which leads to INSULIN SECRETION
492
Q

What is glibenclamide?

A
  • is a sulphonylurea and insulin secretagogue
493
Q

What are the side effects of glibenclamide?

A
  • hypoglycaemia
  • weight gain -> therefore preferred in lean suffers of T2DM
494
Q

What is acarbose?

A
  • an alpha glucosidase inhibitor
495
Q

What is the mechanism of alpha glucosidase inhibitors?

A
  • prolongs the absorption of oligosaccharides -> allows insulin secretion to cope following the loss of the first phase insulin response
  • as effective as metformin in early diabetes
496
Q

What is the major side effect of acarbose/alpha glucosidase inhibitors?

A
  • flatulence -> sugar reaches the large intestine where it is fermented
497
Q

What is the thiazolidinedione that is available to be prescribed?

A
  • pioglitazone
498
Q

How do thiazolidinediones work as treatment for T2DM?

A
  • peroxisome proliferator-activated receptor (PPAR-g) agonists -> are insulin sensitiser - mainly peripheral -> improvement in glycaemia and lipids
499
Q

What are the side effects of thiazolidinedione?

A
  • peripheral weight gain -> modification to adipocyte differentiation
  • older types -> hepatitis and heart failure
500
Q

What is the incretin effect?

A
  • the modulation of insulin secretion by L-cells in the gut -> produces products, such as GLP-1, that increases insulin secretion
501
Q

What hormone can be utilised as a treatment for T2DM?

A
  • GLP-1
  • gut hormone produced by L-cells that stimulates insulin secretion (incretin effect) and suppresses glucagon
  • increases satiety -> helps with obesity
502
Q

What is the issue with GLP-1 and how is this overcome?

A
  • it has a short half-life due to degeneration from dipeptidyl peptidase-4 (DPP-4)
  • gliptins are DPP-4 inhibitors so increase the half-life of GLP-1
503
Q

What other aspects, excluding blood sugar, should be controlled in T2DM?

A
  • weight
  • blood pressure -> 90% of T2DM patients -> clear benefits
  • diabetic dyslipidaemia (high cholesterol, triglycerides, HDL-cholesterol) -> clear benefits to treatment
504
Q

What is the most affective treatment at prevent the progression of diabetes?

A
  • intensive lifestyle with good diet and exercise
505
Q

What are the sites of microvascular diabetic complications?

A
  • retinala rteris -> diabetic retinopathy
  • glomerular arterioles -> nephropathy
  • vaso vasorum - tiny arteries that supply nerves neuropathy
506
Q

State some factors that increase the risk of microvascular complications.

A
  • severity of hyperglycaemia
  • hypertension
  • genetics
  • hyperglucaemic memory -> poorly controlled diabetes, even for a short-time, before good controll increase the risk
  • tissue damage through originally reversible and later irreversible alterations in proteins
507
Q

What is the mechanism of glucose damage to the microvascular?

A
508
Q

What is the main cause of blindness in people of working age, in the UK?

A
  • diabetic retinopathy
509
Q

What does a normal retina look like?

A
  • optic disc at the nasal part of the eye and the macula more laterally (involved in colour vision and acuity)
  • various vessels that come out into the back of the retina and they usually have a very regular arrangement
510
Q

What are the 4 retinopathies?

A
  • Background Diabetic Retinopathy
  • Pre-proliferative Retinopathy
  • Proliferative Retinopathy
  • Maculopathy
511
Q

What are the features of background diabetic retinopathy?

A
  • hard exudates -> appear as cheesy, yellow space in the retina -> due to leakage of lipid content
  • microaneurysms -> can rupture and cause blot haemorrhages
512
Q

What are the features of pre-proliferative diabetic retinopathy?

A
  • cotton wool spots - aka soft exudates -> show retinal ischaemia
  • show severall haemorrhages
513
Q

Describe proliferative retinopathy.

A
  • involves the formation of new vessels in response to retinal ischaemia
  • if the vessels form in the region of the macula they cause problems with acuity and colour vision
  • the newly formed vessels are generally more fragile -> can bleed at anytime
514
Q

Describe maculopathy.

A
  • hard exudates near the macula -> same disease a background diabetic retinopathy barring the location of the hard exudates
  • can threaten direct vision
515
Q

What is the management of background diabetic retinopathy?

A
  • improve control of blood glucose
  • warn the patient taht the warning signs are present
516
Q

What is the treatment for pre-proliferative diabetic retinopathy?

A
  • pan-retinal photocoagulation -> laser the retina and therefore stop the vessels bleeding
517
Q

What is the treatment for proliferative diabetic retinopathy?

A
  • pan-retinal photocoagulation -> laser the retina and stop the vessels from bleeding
518
Q

What is the treatment for maculopathy?

A
  • GRID of photocoagulation -> no need for pan-retinal photocoagulation, can be more targetted
519
Q

What are the features of diabetic nephropathy?

A
  • hypertension
  • progessively increasing proteinuria
  • progressively deteriorating kidney function
  • classic histological features
520
Q

What events likelihood of occuring increases with diabetes and chronic kidney disease?

A
  • cardiovascular events
521
Q

What are the features of nephropathy?

A
  • mesangial expansion
  • basement membrane thickening
  • glomerulosclerosis (hardening of the capillaries) -> becomes less flexible and harder -> absorption of nutrients changes -> more pressure going through the kidneys -> changes in blood pressure control

o essentially, in diabetic nephropathy there is overproduction of matrix leading to mesangial expansion and to basement membrane thickening

  • as it progresses, you get sclerosis of the glomerulus and secondary effects on the tubulointerstitium -> several stimuli for these processes including the effects of prolonged exposure to high glucose or glycosylated proteins in at risk patients
  • rise in the pressure within the glomerular capillaries can stimulate expansion of the matrix -> angiotensin stimulates pathways that result in overproduction of matrix -> angiotensin can also cause the constriction of the efferent arterioles, thus increasing transglomerular capillary pressure
522
Q

How is nephropathy diagnosed?

A
  • albumin in the urine
523
Q

What factor affect the chance of diabetic nephropathy in T2DM?

A
  • age at development of disease will affect the epidemiology
  • racial factors
  • age at presentation
  • death due to cardiovascular morbidity/macrovascular disease -> kills them before development of nephropathy
524
Q

What are the clinical features of diabetic nephropathy?

A
  • progressive proteinuria
  • increased blood pressure
  • deranged renal function
525
Q

How is proteinuria measured?

A
  • urine dipsticks
526
Q

What are the normal and nephropathy ranges of protein in urine?

A
  • normal = <30mg/24hrs
  • nephrotic = >3,000mg/24hrs
527
Q

What are the strategies in diabetic nephropathy intervention?

A
  • diabetic control
  • blood pressure control -> slows down the deterioration in kidney function -> usually use ACE inhibitors -> reduce the rate of decline of creatinine thus reducing the rate of deterioration of kidney function
  • stopping smoking
528
Q

What is the most common cause of neuropathy?

A
  • diabetes -> therefore it is the most common cause of amputation
529
Q

What does neuropathy result from?

A
  • blockade of the vasa vasorum - small blood vessels which supply the nerves
530
Q

What types of neuropathy are associated with diabetes?

A

o Peripheral polyneuropathy

o Mononeuropathy

o Mononeuritis multiplex

o Radiculopathy

o Autonomic neuropathy

o Diabetic amyotrophy

  • at Parties, Marilyn Manson Regularly Abuses Drugs
531
Q

What are the clinical features of peripheral neuropathy?

A
  • loss of sensation
  • loss of ankle jerks
  • loss of vibrational sense
  • multiple fractures in the feet -> can’t feel in correct walking -> can lead to Charcot’s joints and permenant damage
532
Q

Which areas are most commonly effected in peripheral neuropathy?

A
  • hands and feet -> longest nerves supply the feet and then the hands
533
Q

Peipheral neuropathy is most common in which groups of patients?

A
  • tall patients
  • patients with poorly controlled glucose
534
Q

How can you test for peripherla neuropathy?

A
  • monofilament examination
535
Q

What are the features of mononeuropathy?

A
  • usually a sudden motor loss - usually in the longest nerves -> wrist or foot drop
  • can cause cranial nerve palsy -> double vision as a consequence
536
Q

What is third nerve palsy?

A
  • the loss of motor function in an eye -> the eye is usually down and out because of the unopposed action of lateral rectus and superior oblique
537
Q

What are the two different 3rd nerve palsy, which one occurs in diabetes and explain why it occurs in diabetes?

A

o pupil sparing third nerve palsy -> diabetes

  • pupil responds to light as parasympathetic fibres are on the outside so don’t lose blood supply

o third nervy palsy caused by aneurysm

  • pupil is fixed dilated as the aneurysm presses on the parasympathetic fibres first
538
Q

What is mononeuritis multiplex?

A
  • a random combination of peripheral nerve lesions
539
Q

What is radiculopathy?

A
  • pain over spinal nerves -> usually affects the dermatomes on the abdomen and/or chest wall
540
Q

Which diabetic neuropathy is the hardest to treat?

A
  • autonomic
541
Q

What is autonomic neuropathy?

A
  • loss of sympathetic and parasympathetic nerves to the GI tract, bladder and cardiovascular system
542
Q

What are the effects of autonomic neuropathy on the GI tract?

A

o difficulty swallowing

o delayed gastric emptying

o constipation/nocturnal diarrhoea

543
Q

What systems are affected by autonomic neuropathy?

A
  • GI
  • urinary system
  • CVS
544
Q

What are the effects of autonomic neuropathy on the urinary system?

A
  • bladder dysfunction
545
Q

What are the effects of autonomic neuropathy on the CVS?

A
  • postural hypotension
  • cardiac autonomic supply -> cause of sudden death in diabetes patients
546
Q

How can you check for autnomic neuropathy?

A

o measure changes in heart rate in response to the Valsalva manouevre -> patients blows into a tube -> this normally causes a change in heart rate but it doesn’t in diabetics

547
Q

What is the biggest danger in peripheral neuropathy?

A
  • a suffers steps on something like a rusty nail and doesnt know it is there for days
548
Q

Describe the development of atheroma.

A
  • small accumulations of extracellular lipid will lead to atheroma which then leads to fibroatheroma -> could lead to a complicated lesion which can ulcerate, exposing the fat underneath and this can either thrombose entirely or send emboli further down the circulation
  • initially, this process begins with accumulation of lipid but progresses to involve smooth muscle -> this can lead to smooth muscle hypertrophy, fibrosis and calcification
549
Q

What are the macrovascular complications of diabetes?

A
  • ischaemic heart disease
  • cerebrovascular
  • renal artery stenosis
  • peripheral vascular disease
550
Q

What is the link to insulin resistance and life expectancy, explaining the link?

A

o the more insulin resistant you are, the shorter the lifespan

551
Q

What is the likelihood that someone with diabetes, who hasn’t had a MI, will have a MI?

A
  • the same as those who don’t have diabetes but have had a MI
552
Q

What is the Framingham risk score?

A
  • a gender-specific algorithm used to estimate the 10-year cardiovascular risk of an individual
553
Q

What is the main cause of morbidity and mortality in diabetes?

A
  • ischaemic heart disease -> same mechanism as without diabetes
554
Q

What are the difference between diabetic and non-diabetic when it comes to cerebrovascular disease?

A
  • earlier with diabetes
  • more widespread -> lots of small strokes as well as larger ones
555
Q

What is the major problem with peripheral vascular disease?

A
  • contributes to diabetic foot problems with neuropathy
556
Q

Does treating blood glucose offset the increased risk of CVD?

A
  • intensive glucose control does improve coronary heart disease risk but not significantly -> does not translate to much of a change in mortality
557
Q

Which race has an increased risk of CHD?

A
  • for a given cholesterol, South Asians have an increased risk of CHD mortality
558
Q

What are the risk factors for macrovascular disease?

A
  • low birth weight increases the chance of macrovascular disease
559
Q

What two complications predispose diabetic to foot disease?

A
  • neuropathy
  • peripheral vascular disease
560
Q

Describe the monofilament test.

A
  • the filament bends you have applied 10g of pressure
  • a normal foot will be able to feel the 10g
  • normally, the ball of the foot is checked because that is the most common site at which ulcers are found -> 50% of new foot disease starts at the ball of the foot
561
Q

What is the pathway to foot ulceration?

A
562
Q

What is th consequence of motor neuropathy to the feet?

A
  • motor neuropathy can cause an imbalance between the extensors and the long plantar flexors causing an abnormal shape in the foot -> means that increased pressure is being applied on the ball of the foot and the knuckles of the toes
563
Q

Name a problem that could occur withd iabetes that can make tendons less flexible?

A
  • sugar binding to haemoglobin (HbA1c)
564
Q

What are the consequences of autonomic neuropathy to the foot?

A
  • abnormal blood flow in the foot -> could cause an increased blood flow at the wrong time so that there is an increase in pulse pressure in the foot
  • reduced sweating/oil release in the foot which normally protects it from minor disease
565
Q

What are the symptoms of neuropathic foot?

A
  • numb
  • warm and dry
  • palpable foot pulses
  • ulcers at points of high pressure -> ball of the foot
566
Q

What are the symptoms of ischaemic foot?

A
  • cold
  • pulseless
  • ulcers at the foots margins -> toes and heel
567
Q

What are the symptoms of neuro-ischaemic foot?

A
  • numb
  • cold and dry
  • pulseless
  • ulcers at the points of high pressure and the margins of the foot
568
Q

What group of patients are most at risk to neuro-ischaemic foot?

A
  • diabetics who smoke
569
Q

Nme some strategies of managing/preventing diabetic foot ulceration?

A
  • prevent hyperglycaemia
  • prevent hypertension
  • prevent dyslipidaemia
  • stop smoking
  • education
  • control diabetes
  • inspect feet daily
  • have feet measured when buying shoes
  • buy shoes with laces and square toe box
  • inspect inside of shoes for foreign objects attend chiropodist
  • cut nails straight across
  • care with heat
  • never walk barefoot
570
Q

What are the treatment of foot ulceration

A

o relief of pressure -> bed rest (increases risk of risk of DVT, heel ulceration) so may choose to redistribe the pressurevia a total contact cast

o antibiotics -> possibly long term if it has affected bone

o debridement

o revascularization -> angioplasty, arterial bypass surgery

o amputation if it is severly infected -> could be toe, foot or below the knee

571
Q

What is this?

A
  • a Charcot’s Foot
572
Q

Describe Charcot’s Foot.

A
  • bones in the feet don’t have their normal articulations due to loss of joint position in diabetes -> are articulated in such a way that it would be excruciatingly painful in a normal person but in diabetics it is totally painless
  • abnormal shape of the foot means that there is extreme risk of ulceration
573
Q

How is it possible to distinguish betwen osteomyelitis (infection of the bone) and Charcot’s foot?

A
  • MRI