CDL302 Flashcards

1
Q

define aetiology

A

the cause, set of causes or manner of causation of a disease

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

define pathogenesis

A

the origination and development of a disease

the cellular events and relations and other pathologic mechanisms occurring in the development of disease

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

what is a respiratory disease?

A

any disease that affects the airways therefore affecting gas exchange in the air sacs

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

in asthmatic patients, what component in the airways is affected and how?

A

the level of mucus in the airway of asthmatic patients is increased

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

how many diseases are there that can affect the airways?

A

over 40

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

how can respiratory diseases be divided?

A

they can be divided in 4 ways

  • obstructive conditions
  • restrictive conditions
  • infectious, environmental and other diseases
  • vascular diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

what is the difference between obstructive and restrictive lung diseases?

A
  • obstruction leads to a reduction in air flow whereas restriction leads to a reduction in lung volume
  • obstructive conditions make it hard to exhale as air remains in the lungs even after full expiration whereas in restrictive conditions, it is hard to inhale as you can’t fully expand the lungs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

what is the difference between the cause of obstructive and restrictive lung diseases?

A
obstructive = due to inflammation, excess mucus, airway narrowing (smooth muscle tightening)
restrictive = due to lung scarring, fibrosis and extra-parenchymal problems
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

what are the symptoms of obstructive and restrictive ling conditions?

A

they both have similar symptoms:

-difficulty breathing - particularly under exertion i.e. trying to do exercise

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

what are some examples of obstructive conditions?

A

COPD, asthma, bronchitis etc

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

what are some examples of restrictive lung diseases?

A

interstitial lung disease, sarcoidosis etc

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

are conditions affecting the lungs life threatening?

A

many are mild and can be self limiting e.g. cold whereas others are life threatening i.e. pneumonia
the self limiting conditions can become life threatening if they have underlying chronic conditions e.g. COPD

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

how much air do we inhale a day?

A

11,000 litres

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

what is the problem with the fact that we inhale so much air a day?

A

there are inhalation exposures i.e. allergens (pollen), microbes, aerosolised toxins (tobacco smoke,air pollutants) that can contact our lungs

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

what kind of people are especially vulnerable to inhalation exposures to our lungs?

A

spatially (geographically), temporally (age), circumstances (morbidly obese, nutrition, economics, race, gender etc)

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

what are some genetic influences on how the lungs are affected?

A

usually rare diseases due to a single genetic defect

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

what are monogenic diseases?

A

caused by mutation in a single gene e.g. sickle cell disease, cystic fibrosis etc

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

what are polygenic diseases?

A

depend on the simultaneous presence of several genes, not inherited as simply a single-gene disease. e.g. hypertension, CHD, type 2 diabetes
they are associated with the effects of multiple genes in combination with lifestyles and environmental factors

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

are lung diseases usually due to genetics or the environment?

A

they are usually not single entities

so are due to a gene-environment interaction

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

are respiratory diseases caused by genetic factors or environmental factors or both
with examples

A

genes and the environment can independently cause a respiratory disease but can also combine their effects

  • CF = monogenic
  • asthma, lung cancer and COPD = polygenic
  • carbon monoxide poisoning = environmental
  • alpha 1 anti-trypsin deficiency = monogenic but ca be influenced by the environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

is alpha-1 trypsin deficiency caused by genes or the environment?

A

it is monogenic however the environment can have an effect on it

  • main genetic cause of COPD
  • if you have this deficiency, you will develop features of COPD but if you smoke, this massively potentates the progression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

what is the difference between monogenic and polygenic diseases?

A
monogenic = rare diseases attributable to genetic variations with large effects. inherited in classic mendelian fashion
polygenic = complex diseases controlled by 2 or more genes at different loci or different chromosomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

what are the genes that are seen in both monogenic and polygenic diseases?

A

TGFB1, TNFA and ADAM33

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

is smoking a purely environmental cause of lung disease?

A

no because 90% of COPD patients are smokers and 10-20% get COPD. this shows that it can’t be just environmental

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

how can genetic be useful in the study of lung diseases?

A
diagnosis
estimation of risk
understanding pathological mechanisms
ene therapy
guidance for new therapies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

what are the cells of the immune system? give examples of each

A
  • lymphocytes (T, B and NKC)
  • phagocytic cells(monocytes and macrophages)
  • granulocytic cells (neutrophils, eosinophils and basophils)
  • dendritic cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

what are some barriers that pathogens encounter before reaching the immune system?

A

there are anatomical and physiological barriers that the pathogens encounter
in the lungs, there are cells that line the airways and produce surfactants, there is ciliary clearance, a low stomach pH and lysosomes in the tears and saliva

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

what is the function of the epithelial cells that lien the airways in the lungs?

A

they have a barrier function and are also able to detect and respond to pathogens

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

what are the cellular and humoral protections of the innate immune system>?

A

cellular: NKC, dendritic cells, macrophages, eosinophils, neutrophils, mast cells etc
humoral: part of the acute phase response (LPs binding proteins)

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

what are the cellular and humoral protections of the adaptive immune system?

A

cellular: T cells and B cells
humoral: antibodies

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

is there any interaction between the innate and adaptive immune system?

A

yes, they communicate a lot

the adaptive immune systems progressed from the innate system

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

what are the cells that sit in the cusp between the innate and adaptive immunity systems?

A

NKT cells and dendritic cells sit on the cusp of the 2 systems

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

describe the innate immune system

A

it is important in not getting us sick initially
involves a rapid host defence against invading pathogens
it is evolutionarily conserved
involved in the recognition of invading pathogens and activation of anti-microbial response
is emerging as a critical regulator of human inflammatory disease
implicated in the development of some diseases e.g. asthma

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

what is the significance in the fact that the innate immune system never changes?

A

this means that it will always respond to a pathogen in the same way i.e. if you inhale a bacteria today and again the same bacteria in 6 years, it will respond in the same way and take the same amount of time. this is because the quicker second response is by the adaptive immune system

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

how does the innate immunity deal with the fact that pathogens are able to evolve much quicker than our cells are?

A

our genetic attributes are much slower than the pathogens ones and this means that we would struggle to keep up. so, in innate immunity, they recognise very conserved structures within bacteria and also across different sets of pathogens.
they have also evolved to recognise some of the biological consequences of infection

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

what does the action of the innate immune system rely on?

A

they rely on a limited number of genetically predetermined receptors called pattern recognition receptors
these are highly conserved structures that are expressed by a large group of pathogens
they are common biological consequences of infection

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

what are the 3 broad strategies that the innate immune system utilises?

A

2 of the 3 strategies utilise pattern recognition receptors

  1. microbal non-self = pathogen associated molecular patterns (PAMPS) e.g. TLRs, RLRs, NLRs and collecting family
  2. consequences of damage/injury = damage associated molecular patterns (DAMPS) e.g TLRs, RLRs, NLRs and RAGE family
  3. missing self = MHC class 1 specific inhibitory receptors - these cells are on the cell surface and inordinacy health, they would be presenting host cells to the immune system and NK cells recognise these as host cells and cause an inhibitory signal so that they don’t attack themselves
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

how does the innate immune system function using PAMPs

A

it recognises the PAMPs that are found on pathogens but not usually on host cells
PRRs either act to directly bind PAMPs or interacting with other receptors that are bound to PAMPs
bacteria, fungi and viruses are all recognised by their PAMPs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

what is the good thing about using a pathogens PAMPs to recognise it?

A

they are not easily mutated and they are very crucial to the cells function and so this would be something easy to recognise a pathogen by

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

how does the immune system function using DAMPs?

A

they are endogenous molecules that are created to alert the host to tissue injury and to initiate repair
can be intracellularly or extracellularly
intracellular: molecules released by cell necrosis/activation following injury
in the extracellular metric, molecule fragments are released/up-regulated in response to tissue injury
these prime the immune system to be ready to attack

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

what kind of cells are released by DAMPs intracellularly?

A

proteins = heatshock proteins (TLR2 and 4), high mobility box1 protein (TLR2 and 4)
self nucleic acids: mRNA (TLR3), ssRNA (TLR7 and 8), DNA (TLR9), IgG-chromatin complexes (TLR9)

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

what kind of cells are released by DAMPs extracellularly?

A

proteins: fibrinogen (TLR4), fibronectin (TLR4), tenascin-c (TLR4)
proteoglycans and glycosaminoglycans: billycan (TLR2&4), version (TLR2), hyaluronic acid fragments (TLR2&4)

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

how could DAMPs cause pathogenesis?

A

harmful stimuli e.g. pathogens, injury, heat, auto-antigens, tumours and necrotic cells can all trigger damage leading to release DAMPs.
this will activate TLRs and lead to the release of pro-inflammatory mediators which then can feedback and cause an increase tissue damage.
if this loop is allowed to propagate and lead to the release of more and more DAMPs, it can have a detrimental effect and cause pathogenesis

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

what kind of diseases are associated with high levels of DAMPs?

A

associated with many inflammatory and autoimmune diseases swell as atherosclerosis and cancer
- although endogenous signals can be good (to prime the host if your under attack immunologically), if they are allowed to propagate and not checked it can do more harm

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

how many TLR receptors are in humans?

A

10

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

how can TLRs be divided?

A

can be broadly divided into those who recognise bacteria which are on the cells surface and those who recognise viruses which are contained within endosomes

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

what is the difference between TLRs on the cell surface and those in endosomes?

A

TLRs in endosomes generally can’t recognise host molecules where as those on the cell surface recognise any kind of DNA or RNA

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

give 2 examples of TLRs - 1 in humans and 1 in a model organism

A

TLR10 - cell surface, ligand undetermined in humans

TLR11-13 = expressed in mice

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

what can TLR4 recognise and what can it do?

A

TLR4 can recognise bacterias e.g. lipopolysaccharide and mannan , also Repiratory Sinsitual Virus and a lot of host proteins including Tenascin-C
– this will trigger an inflammatory response to all of these different ligands

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

how are TLRs different to normal receptors?

A

they are very different to normal receptors as they have the ability to recognise a wide range of ligands
they sometimes use co-receptors to do this e.g. TLR2 can work with TLR1 and 6 which is a way to expand the repertoire of proteins that they can see/recognise

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

describe TLR signalling

- using TRL4 as e.g.

A

the TIR domains of TLRs interact with TIR domains of the adaptor proteins
TLR4-TRIF signalling = initiated within endosomes
(in bacteria): phosphorylation cascades activate NF-kB and MAPKs allowing entry to the nucleus to drive expression of cytokine genes - predominantly in response to bacteria
(in viruses): phosphorylation of IRFs lead to localisation of the nucleus to drive expression of type 1 interferon (IFN) genes to help control viral infections
-in response to TRIF, IRF activation happens
-cross talk an occur i.e. TRIF activating NF-KB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

which TIRs can interact with MYD88?

A

all TIRs except TLR3.

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

what are the TIR adaptors and how many are there?

A

there are 4 main TIR adaptors: MYD88, TRIF, TIRAP/MAL & TRAM

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

what does TLR4 need to bind to Myd88?

A

it needs a bridge i.e TRF1 as it can’t directly bind to Myd88

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

what is the difference between TLRs and NLRs?

A

NLRs detect bacteria from the cytosol

TLRs detect bacteria from the endosomes and cell surface

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

how many NLR genes are present in humans and in mice?

A

22 in humans

34 in mice

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

what is the role of NLRs?

A

they are involved in sensing intracellular bacterial pathogens and DAMPs, and in the regulation of inflammatory and cells death responses

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

what are NLRs characterised by?

A

by the presence of conserved nucleotide-binding and oligodimerisation domain (NOD) motif that activates the signing complex

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

how many structural subfamilies of NLR are there and what are they?

A
there are 5
NRLA - acidic domain
NRLB - BIR domain
NLRC - CARD domain (NOD1, NOD2)
NRLP - Pyrin domain (NRLP3)
NRLPX - no known homology
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

how many functional subfamilies of NRLs are there?

A

there are 2 functional NRLs

  • NRL C(NOD1 and NOD2) - inflammatory and antimicrobial signalling
  • NRLP3, NRLP6 and NAIPs - inflammasomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

what do NOD1 and NOD 2 sense?

A

NOD1 and 2 sense bacterial molecules produced during the synthesis, degradation and remodelling of peptidoglycan (PGN) - which is a major component of bacterial cell walls
NOD1 recognises mess-diaminopimelic acid(mess-DAP) containing PGN fragments (MAINLY GRAM NEGATIVE)
NOD2 senses muramyl dipeptide (MDP) found in the PGN of nearly all GRAM POSITIVE and GRAM-NEGATIVE organisms

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

what does the NOD family result in the singling of?

A

NOD family results in the signalling of NFK-6, MAPKs and the release of pro-inflammatory cytokines

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

how are inflammasomes different from NODs?

A

they are different as they also can recognise the microbial products in the cytoplasm and also cellular/metabolic stress which can trigger activation of the inflamasomes which are involved in cleaving/activating cytokines e.g. IL-1B and IL-18. this is an additional control mechanism

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

what are RLRs and what can they detect? what members are there?

A

RLRs = rig like receptors
there are 3 members that detect viral RNA in the cytoplasm
RIG-1, MDA-5 and LGP2

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

describe RIG-1

A

RIG-1 = retanoic acid inducible gene 1
short dsRNA (up to 1kb)
s’ triphosphate (5’ppp) cas (ss or ds RNA)
influenza A and RSV

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

describe MDA-5

A

melanoma differentiated gene 5
long ds RNA (over 2kb) with no end specificity
replication intermediates
rhinovirus

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

describe LGP2

A

laboratory of genetics and physiology 2

very high affinity for any dsRNA

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

describe RLR signalling

A

ligand binding induces conformational changes and oligomerisation of RLRs to activate the signalling partner IPS-1 on mitochondrial membranes
IPS-1 activates signalling cascades leading to activation of IRFs and NF-kB an the expression of interferon and cytokine genes
LGP2 functions as a positive regulator in RIG-1 mediated and MDA5 mediated virus recognition

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

what cells are involved in the innate response to infection? what are there roles?

A

cytokines, chemokine and others (e.g. acute phase response, adhesion molecules)
these have 2 roles: immediate defence and direct adaptive immunity

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

how many cytokines are involved in the inflammatory response? what are the main functions of the cytokines and how do they work?

A

5 main cytokines
1 of the main functions is to recruit the inflammatory cells to the site of infection.
could be through effects on the vascular endothelium to make it more leaky so that the cells can get out
could upregulate adhesion molecules on these cells allowing them to bind to the endothelium and pass through into the area where the pathogen is

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

what is the role of adhesion molecules?

A

they are induced on circulating immune cells and endothelial cells
they work to co-ordinate movement of cells into infected tissues where phagocytosis and killing takes place

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

what can be triggered during the systemic effects?

A

the acute phase

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

describe the acute phase response in the liver

A

cytokines can activate hepatocytes in the liver to generate acute phase proteins

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

describe the acute phase response in the hypothalamus

A

they are endogenous pathogens and so act on the hypothalamus to raise the body temperature (fever)

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

describe the acute phase response in dendritic cells

A

TNF-alpha stimulates migration to lymph nodes and maturation

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

why advantage is there to inflammation causing a fever?

A

because bacteria and viruses find it more difficult to replicate at a high temperature whereas our adaptive immune system works much better at these high temperatures

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

give an example of a condition which can occur where the acute phase response system goes wrong
- stats

A

sepsis is a condition that occurs when the acute phase response goes wrong
affects ~200000 people in the UK every year and ~40,000 people die
many of these are avoidable but this condition can be very hard to diagnose
its usually detected in the latter stages, making it difficult to stop

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

how does sepsis develop and how does it cause damage to organs?

A

starts with an infection from insect bites/cuts etc
immune response responds to this in a normal way
for some reason, wither the body can’t control the spread of infection around the body or because the immune system is in overdrive leads to sepsis. this can cause damage to organs

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

what does the release of interferons lead to the activation of in the anti-viral response to infection?

A

release interferons will then act on interferon receptors (IFNalpha and IFNbeta) activating the STAT pathway
has 3 main effects on our cells

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

what are the 3 main effects on our cells caused by the activation of STAT pathway in the anti viral response to infection

A

1 = induce resistant to viral replication.
activates genes that cause the destruction of mRNA
inhibits the translocation of viral proteins and some host proteins
interferons tend to communicate to neighbouring cells that viral attack is underway thereby priming them for an attack
2 = increases MHC class 1 expression and antigen presenting of viral proteins
facilitates recognition and susceptibility to cytotoxic t cells
3 = activates NK cells to kill virus infected cells
as well as activating dendritic cells and macrophages + include chemokine to recruit lymphocytes

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

how can you target an over active immune system?

A

1 way to target to detrimental effects on over active immune system is to inhibit the response using antagonists

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

what are the 2 main strategies of immunomodulation

A

antagonists: block binding of ligands/protein ligands complexes to receptors/ interfere with intracellular adaptor molecules of common signalling pathways
antagonists developed to date = small molecules, proteins, oligonucleotides, antibodies and peptides
agonists: adjuvant effect (used with a primary treatment) promoting protective responses e.g. interferes with anti viral, vaccines

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

what are side effects of antagonists and agonists as strategies of immunomodulation?

A

Antagonists - side effects = potentially repress protective mechanisms
Agonists - side effects = potentially enhance inflammation

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

what are the respiratory tracts mace of?

A

cartilaginous rings

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

what part of the body is the respiratory tract attached to?

A

joined to the middle ear cavity and this is why when you get a cold, your ear is sometimes blocked

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

what are the functions of the lungs split into?

A

respiratory and non-respiratory

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

what are the respiratory functions of the lungs?

A

ventilation and gas exchange CO2, O2, pH, humidifying

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

what are the non-respiratory functions of the lungs?

A
  • making, activating and inactivating of vasoactive substances, hormones and neuropeptides
  • lung defence: compliment activation of host defence proteins, recruitment of leukocytes, cytokines and growth factors
  • speech, vomiting, defacation and child birth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

how are lungs a major biosynthetic organ?

A

they are highly vascularised

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

why are respiratory infections so rare in comparison to the amount of air we inhale?

A

we inhale ~10,000 litres of air every day and lots of pathogens could potentially come in and infect our lungs but our host defence system can prevent pathogens from coming in and establishing an infection

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

what is epithelial tissue composed by?

A

cells that line the cavities and surfaces of structures thouhout the body

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

what tissue are glands formed by?

A

epithelial cells

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

what are connective tissue and epithelial tissue layers separated by?

A

basement membrane

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

what is the primary role of respiratory epithelium?

A

to line the airways and moisten them. also to protect them by acting as a barrier to prevent potential pathogens or foreigners from infecting the lungs

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

how does the respiratory epithelium work to prevent pathogens and foreign particles etc from entering the lungs?

A

by work of the mucociliary escalator

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

what are the chemical epithelial barriers?

A
  1. anti-proteases: SLP-1, lysozyme and phospholipase A
    - these are to protect the lungs from proteases that could be left behind after an inflammatory response etc
  2. anti-fungals
  3. anti-microbial peptides
  4. surfactant A and D - opsonise pathogens for enhanced phagocytosis
    - interact with the surface of pathogens and then link the phagocytosis mechanism within the lungs and they mark these pathogens for uptake by professional phagocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

give an example of a non specific defence mechanism in the lungs

A

bacterial flora - provide a low level induction system that keep the defence system primed

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

what evidence is there to show that different parts of the respiratory tract have different functions?

A

different parts of the tract have different airway epithelium i.e. bronchioles = ticker than in the alveoli
as you go down the tract, cells get more cuboidal/columnar and alveoli gets more squamous

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

are mice and human epithelia similar? what are some differences?

A

they are similar
human airways are more cartilaginous and have a greater number of mucosal glands
the epithelial cell population is about the same

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

what is the name of the junction where bronchioles move into alveolar gas exchange regions?

A

the bronchioalveolar duct

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

what are neuroendocrine cells?

A

basal cells which are progenitor cells for repopulation of airways following injury in the trachea and bronchi

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

what is a pericellar lining fluid? where is it

A

there is cilia with pericellar lining fluid above it which is thin watery fluid that the airways secrete
on top of this is a mucin layer

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

what cell types are there in the submucosal gland?

A

there are 2 types of secretory cells.

  • goblet cells which produce mucus
  • serous cells which produce a different series of proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

what cells are specialised to help with the gas exchange in the alveolar epithelium?

A

type 1 and type 2 cells in the alveoli are very thin cells that have a huge surface area with a nucleus that sticks out. they allow reasonably for gas exchange between the capillaries and internal surface of the alveolar structure
-surfactant = a lipid based material that serves to lower the surface tension of the respiratory tract

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

what is mucus made of and what secretes it?

A

airway mucus is a viscoelastic gel containing water,carbohydrates, proteins and lipids.
it is the secretory product of the mucous cells (goblet cels and submucosal glands)

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

what is the gel - brush hypothesis?

A

it describes the mucus on top of cilia which brushes it

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

what is the mucus gel layer made by?

A

created by huge proteins- MucSAC/SB

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

where is mucus transported to and how is this done?

A

mucus is transported from the lower respiratory tract into the pharynx by air flow and mucociliaryy clearance

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

what does mucus protect the epithelium from?

A

mucus protects the epithelium from foreign material and from fluid loss

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

how does the mucociliary escalator work?

A
  • encapsulate foreign particles then the cilia beat in organised waves
    the cilia beat in directional (synchronised) waves to move the mucus up the airways
    this happens all the time in the body to prevent being clogged up
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

is the mucus clearance via the escalator a period thing or contents?

A

happens constantly - its innate

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

what is a cough?

A

an expulsive reflex that protects the lungs and respiratory passages from foreign bodies

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

what are the causes of coughs?

A

irritants - smokes, fumes, dusts etc
diseased conditions e.g. COPD, tumours etc
infections (influenza)

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

what is a sneeze?

A

an involuntary expulsion of air containing irritants from the nose

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

what are the causes of sneezes?

A

irritation of nasal mucosa

excess fluid in airways

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

what kind of cells do infections target and what is the consequence of this?

A

they target airway epithelial cells

a conséquence of this is damage

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

in most cases, what happens to airway epithelium following an insult?

A

a complete repair

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

what is the cycle that happens after epithelial cells suffer insult?

A

there is a cycle of differentiation, damage, spreading an d migration, proliferation then differentiation again

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

why are epithelial cells able to change their phenotype?

A

they have a degree functional plasticity which is what allows it to change its phenotype

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

what are the principle progenitor cells in the epithelium?

A

basal cells

there is no evidence that in different regions, basal cells have been able to form different cells

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

what happens if there is an epithelial abnormality in the lungs?

A

pulmonary disease

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

what underpins many obstructive long diseases?

A

abnormal epithelia responses to insult/injury

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

why are mucus plugs/inflammation associated with severe disease?

A

when there is mucus and inflammatory cells blocking the airways, mucus plugs can completely obstruct the airways. this can be coughed up by a patent during an asthmatic attack

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

does cancer or respiratory diseases kill more people?

A

respiratory diseases kill more people than all cancers combined

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

what does the failure of adaptive immunity cause?

A

Failure of adaptive immunity leads to recurrent respiratory infections, specific to the cell affected

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

what are the internal and external defences in the innate immune system?

A

internal:

  • phagocytic cells
  • antimicrobials
  • inflammatory cells
  • natural killer cells

external:

  • skin
  • mucous membrane
  • secretions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

what are the defences are in adaptive immunity?

A
humoral response (antibodies)
cell mediated response (cytotoxic lymphocytes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

what is the difference between an antigen and a pathogen?

A

an antigen is a molecule capable of inducing an immune response
a pathogen is a molecule (bacteria or fungi) that are covered in a myriad of unique receptors)

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

what are the key properties of the adaptive immune system?

A

specific - ability to mount a specific immune response to a huge range of antigens including pathogens
self-recognising - recognition of self antigens and eliminate of attractive clones
memory - development of immunological memory and so is able to remember long lived B and T cells

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

how do lymphocytes recognise antigens?

A
  • T cells and B cells express different unique antigen receptors
  • The variable region determines the receptor specificity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

how many different antigen receptors are there? why is this a problem for DNA?

A

~10^18

DNA can’t encode all those receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

describe how there is diversity and specificity in adaptive immunity

A

• Early in lymph development, diversity comes from DNA recombination
• Generation of pathogen specific variable regions in lymphocyte receptors is the basis of diversity and specificity
• Much diversity is generated early in development via DNA rearrangements (VDJ recombination)
o VDJ lays the blueprint of DNA
• Exposure to relevant antigen, triggers replication and somatic hyper-mutation generating further diversity

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

what is affinity maturation?

A

Selection for higher affinity

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

how do we get millions of unique t cells?

A

• Multiple variable and diverse joining regions
• More looping mechanisms which are excised
o Very random excision of regions then they are put back together
• Random recombination events occur between about 50 Variable (V) 27 Diversity (D) and 6 Joining (J) segments in both heavy and light chains
• Recombination (splicing) is not precise (nucleotides inserted or deleted) greatly increasing diversity
• Further mutations occur on pathogen exposure (somatic hypermutation)
• with selection of higher affinity clones (affinity maturation)

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

in adaptive immunity, what regions does recombination happen (V,D,J)

A

50(V), 27(D), 6(J)

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

what is immune tolerance?

what are the different types of immune tolerance and what can they cause?

A

¥ A state of unresponsiveness of the immune system to substances or tissue that have the capacity to elicit an immune response
¥ Tolerance can be to self, but also to the fetus in pregnancy, or it can develop to pathogens (chronic infections) or cancers

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

what happens if there is a failure to establish tolerance?

A

¥ Failure to establish tolerance leads to autoimmune disease, where our immune system attacks our own body. An example is rheumatoid arthritis

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

what are the 2 types of tolerance?

A

central and peripheral

  • Central tolerance – way to distinguish cells that are self to those that are non self – most in early life
  • Peripheral tolerance – way to prevent over-reactivity of the immune system to environmental entities — continues throughout life
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

where do the 2 types of immunological tolerance occur? and what happens?

A

Central tolerance
In the thymus, lymphocytes that react with self-antigens are deleted or develop into suppressor ‘Tregs’

Peripheral tolerance
In lymph nodes, autoreactive clones escaping central tolerance are deleted or suppressed by Tregs

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

what are t-reg cells?

A

• T cells that react to an autoantigen (antigen from normal tissues which are targeted by humoral or cell mediated immune system) become t-reg cells
o They migrate to migratory tissue and are established there and then mediate peripheral tolerance

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

what is the advantage of immunological memory?

A

¥ Allows rapid immunological response on subsequent exposure

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

in what aspects are memory b cells better than naive cells?

A

¥ Following activation, a small proportion of high affinity B and T cells differentiate into long-lived memory cells, residing in lymph nodes or tissues
¥

Memory B cells are distinguished from naive B cells by an increased lifespan, faster and stronger response to stimulation and expression of high affinity immunoglobulin (IgG)

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

describe the process of antigen presentation

A
  • APCs that have interacted with Th are licensed to activate cytotoxic T cells (CD8 +ve) kill virus-infected or tumour cells
  • Cytotoxic T cells form pores in their target cells which ‘explode’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

how do t helper cells help other immune cells?

A
  • Following APC-Th cell interaction, governed by MHC and co-stimulatory receptor interactions, cytokines are produced
  • Cytokines activate different T cell transcription factors leading to differentiation into different Th cell subsets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

describe the interaction between T helper cells and B cells

A
  • B cells can interact with antigen directly
  • In some cases, they can produce antibody without ‘help’ (T-independent)
  • Mostly B cells need input from Th cells (Th2 and Tfh cells)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

what do activated B cells become?

A

plasma cells

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

what happens in isotype switching?

A

¥ In isotype switching (class switch recombination or CSR) the ‘variable’ antibody region is unchanged but becomes attached to a different constant region, altering its properties.

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

what do naive B cells express initially and what does this change to upon maturation?

A

initially express IgM and IgD

after maturation - IgM = switched to IgG, IgA or IgE

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

what are IgM, IgG, IgA, IgE and IgD for?

A

¥ IgM Produced early in immune response

¥ IgA Mucosal immunity (gut and lung)
¥ IgG >80% circulating antibodies are IgG

¥ IgD Found on the surface of all B cells

¥ IgE Binds mast cells, mediates allergic reactions

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

what does the variable region and constant region of antibodies bind to?

A

• Variable region binds target antigen and constant region interacts with effectors

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

what are IgM ad IgG especially good at ?

A

• IgM are good at neutralising and agglutinating, IgG are good at opsonising and fixing complement

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

How can we manipulate the adaptive immune response therapeutically?
for vaccination, immune suppression,MAB?
also in infection, inflammatory diseases and malignant disease?

A

¥ Vaccination – augment the adaptive response
¥ Immune suppression – reduce the adaptive response
¥ Monoclonal antibodies (’biologic therapy’)
¥ Varied applications including
¥ Inflammatory disease (suppress immune system)
¥ Infection (additive to immune system)
¥ Malignant disease (augment immune system)

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

what are pathogens derived from? and what can they do?

A

¥ Vaccines are derived from pathogens or toxins (e.g. dead or attenuated bacteria, capsular polysaccharide, viral proteins)
¥ Vaccines can prevent infection or reduce morbidity in individuals, and also generate ‘herd immunity’ - need to vaccinate 75-95% (disease-specific)

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

how can vaccinations be administered?

A

¥ Can be administered by injection, nasally, orally etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

describe the problem with the influenza vaccination

A

¥ Annual outbreaks of influenza A lead to massive strains on health services and many deaths
¥ Major antigens are neuraminidase (N) and hemagluttinin (H)
¥ Mutations (antigenic drift – all flu viruses) mean protection is reduced, and genetic re-assortment with viruses affecting other species (antigenic shift – Influenza A only) may lead to pandemics
¥ Influenza A is classified by its expression of N and H e.g. H1N1, H3N1 etc. As influenza A affects other species, antigenic shift can occur
¥ Influenza B causes less severe disease and is less variable (antigenic drift only)

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

how is an influenza vaccine manufactured?

A
  • WHO selects ‘likely’ flu strains for next year in February (2 flu A and 1 flu B). Don’t always guess correctly.
  • Manufacture low pathogenicity virus to express the relevant H and N
  • Mass produce virus in eggs and kill virus to generate vaccine
  • Targets – elderly, chest diseases, diabetes. Immune responses may be insufficient Patients who can’t make antibodies derive limited benefit
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

what are the typed of immunosuppressants?

describe each

A

¥ Corticosteroids – modulate transcription and suppress both innate and adaptive responses. Multiple side effects
¥ Antimetabolites – suppress DNA synthesis – mainly T cells
¥ Methotrexate inhibits dihydrofolate reductase and prevents synthesis of folic acid
¥ Purine analogues e.g. azathiaprine
¥ Mycophenolate – inhibits guanosine synthesis
¥ Calcineurin inhibitors (cyclosporin, tacrolimus) – prevent G0-G1 cell cycle progression – T cells
¥ Monoclonal antibodies - wide range of targets and applications e.g. rituximab (antiCD20) targets B cells

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

how are MABs made? how has this process become advanced?

A

¥ Clonal antibodies engineered to specific targets for scientific and medical applications
¥ Fuse myeloma cells with spleen cells from immunised mice (Kohler and Milstein)
¥ Can now be made by recombinant technology using viruses or yeast and made chimeric and even ‘humanised’ to reduce allergic reactions

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

how do tumour cells evade checkpoint receptors?

A

¥ Presentation of tumour antigen with MRC and co-stimulation should lead to an activated T cell response
¥ T cells express inhibitory checkpoints receptors e.g PD-1 (Programmed Death 1). Tumours may express ligands eg PD-L1 to engage these checkpoints, inhibiting the immune response

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

what has been proposed as a way to stop tumour cells from evading checkpoints?

A

¥ Monoclonal antibodies have been engineered to prevent this, enhancing recognition and immune responses to tumour cells

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

what is asthma?

A

A chronic inflammatory disorder of the airways causing recurrent episodes of wheezing, breathlessness, chest tightness, and cough. These episodes are usually associated with widespread but variable airflow obstruction that is often reversible either spontaneously or with treatment.”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

what kind of cells does asthma involve?

A

both innate and immune cells

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

what are the different causes of asthma?

A

o airway inflammation
o bronchial hyper-responsiveness
o recurrent reversible airway obstruction

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

what is the burden of asthma in the UK?

A

♣ 5.4m in the UK receiving treatment. 1.1m children (1:11), and 4.3m adults (1:12)
♣ 1,246 asthma deaths in 2012 (358 males, 888 females)
- 21 aged 0-14 years
- 204 aged 15-64 years
- 1,021 aged >65 years
♣ 1 person dies from asthma every 7 hours
♣ 60% of asthmatics report significant persistent symptoms or symptom burden
♣ 31%said asthma symptoms prevent them doing things in spare time
♣ 65% of asthmatics report severe attacks (exacerbations)
- excessive mucus clogs up airways in asthma patent

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

why is it difficult to treat asthma with drugs? what is used instead?

A
  • difficult to treat severe asthma attack with drugs because the airways would be clogged up and so taking drugs wouldn’t work as they can’t get access
  • use systemic drugs to open up the airways instead
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

how much does asthma cost the NHS per annum - what is the breakdown of the costs?

A

♣ £1.1bn per annum costs to the NHS
- 6.3m primary care consultations (74% costs)
- 93,000 hospital in-patient episodes
- 1800 intensive-care unit episodes
- 36,800 disability living allowance claims (13%)
♣ 1m lost working days per annum

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

why is asthma becoming more common?

A

♣ Epidemiological data over the last 50 years shows a steep increase in allergic conditions e.g. asthma and immune disorders i.e. Multiple Sclerosis
♣ During this time, there has been a decline in infectious diseases i.e. measles mumps etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

why has the occurrence of infectious diseases gone down?

A

Could be due to successful drugs/therapies for these infectious diseases

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

what is the 2 theories that explain why asthma happens?

A
  • Biodiversity hypothesis shows that proposes that reduced contact of people with natural environmental features and biodiversity, including environmental microbiota, leads to inadequate stimulation of immunoregulatory circuits.
  • Hygiene theory describes the protective influence of microbial exposures in early life on the development of allergy and asthma,
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

what is the link between th1 phenotype and getting asthma?

A

• Exposure to microbial components early in life skews immune response
Th2 Th1
o At birth children have a th2 immunology
o Exposure to microbial infections drive it to a more th1 phenotype which protects it
• Decrease in prototypical Th1-stimulating infections e.g. measles, mumps, tuberculosis

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

why is it suggested that a decline in biodiversity and better hygeine can drive asthma? what is the family link to this?

A

• Declining biodiversity, urbanisation and associated changes to diet and lifestyle
♣ Decrease in infection in early childhood
♣ Elder siblings would be passing on all germs and infections to child earlier than when they would get it so youngest in family would be less likely to get asthma etc
♣ The number of children per family has reduced so this is less of the case
♣ Better hygiene standards and mean that its less common to transfer germs
♣ Children that grew up on farms are more exposed to more microbes and unpasteurised milk etc.

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

what are the risk factors for developing asthma?

A
♣	Development of asthma and triggering exasabtions
o	Allergens
o	Genetics 
o	Food additives
o	Smoking 
o	Repiratory infecions
o	obesity
o	Medication
o	Occupational exposures
o	Pollution
♣	Involved in triggering exasterbations
o	Temperature change
o	Exercise
o	Gastric reflux
o	Stress and emotions
o	Cold air
o	Strong odours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

what is the genetic link to asthma occurrence?

A
♣	Heterogeneous & genetically complex (polygenic)
♣	>100 genes identified
♣	Close relative with atopic disease
♣	1 parent = 25% risk
♣	2 parents = 50% risk
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

what have twin studies shown about the link of asthma to genetics?

A

♣ twin studies used to compare between people with either 100% identical genetic makeup (monozygotic) or 50% of the same genetic makeup (dizygotic)
♣ Twin studies - genetic factors account for:
♣ ~70% variation in disease susceptibility
♣ ~24% variation in overall asthma symptom severity
♣ High heritability does not preclude important contribution of environmental factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

what is epigenetics?

A

♣ Epigenetics - the transcriptional dynamic alterations leading to changes in gene expression

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

what genes have been most linked to asthma?

A

Chemokines expressed in epithelial cell layer

Spink5 and flag involved in tight junction formation

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

what are the physiological aspects inked to asthma?

A

Physiological (e.g. obesity and diet)
-overweight’ higher asthma risk’
nitrites = involved in preserving meat e.g. bacon and they have been linked to asthma
Vitamin D – show to reduce colds etc. the cold virus = key trigger of many asthma exacerbations

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

what are the environmental disease is linked to asthma?

A

• Respiratory Virus Infections (Respiratory Syncytial Virus –RSV)

  • 33.8m RSV infections worldwide per annum in children <5 years
  • Small proportion develop severe disease
  • Severe disease = fourfold incidence of asthma
  • Major trigger of exacerbations
  • High number of children can develop bronchitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

what environemntal factor links to asthma?

A

♣ Tobacco smoke
- Pregnancy
- Parental smoking in childhood
- Genetics (ORMDL3; PCDH1)
♣ All these mean there is an increased risk of asthma in childhood and adulthood
♣ Can cause epigenetic changes
♣ These genes are more closely related to children whose parents smoke

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

what are the phenotypes associated with asthma?

A

• Asthma has evolved from a term describing a single disease to one encompassing multiple subgroups or ‘phenotypes’
• Asthma used to be associated with only th2
• There are other phenotypes that are eosinophilic
• Non-eosinophilic – associated with neutrophils
o It tends to be corticosteroid refractive – don’t respond to inhalers

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

describe the eosinophilic and non-eosinophlic phenotypes of asthma

A

♣ Th2 (eosinophilic)
- Early- and later-onset disease over a range of severities
- Associated with allergy (atopy – the tendency to develop IgE mediated reactions to common aeroallergens)
- Non-allergic variants (late onset, exercise-induced)
♣ Non-Th2 (non-eosinophilic)
- Obesity, smoking, neutrophilic
- Associated with more severe disease

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

describe eosinophilic asthma including the genes that are involved and what it causes

A

♣ Eosinophilic asthma
o Environmental allergens
o Dendritic cells which are APC
o Takeup allergen, process it and present tem in lymph nodes oto t cells. This drives a th2 phenotype of the th2cells
o Allergic info = driven by the t clls
o Cytokines: il4,5 and 13 =key processes
o Il4 – eosinophilic
o Il4- and 13 – effects on epithelial layer and smooth muscle cells
o Mast cells =key as they nind the ag specific IgE leading to degranulation
o Tryase – proinflammatory
o X – pro fibrotic
o Release bronchoconstriction
o Causes SM to contract leading to bronchio-hyperactivity

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

descibe non-eosinophilic asthma including the genes involved

A
•	Non-eosinophilic asthma
o	More theoretical so less I known
o	Key triggers – pathgens
o	Toll like receptors
o	Smoke etc.
o	Act on epithelial cells and macrophages releasing il8
o	Neutrophils cause airway 
o	T cells have a key role
o	Th1 and th17 cells – drive epithelial cell to produce il8
o	Release if Gamma – act on mast cells etc
o	Known defects –
o	Interferons
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

what is the main pathology of asthma?

A

Decrease in diameter of the airways

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

what is a condition linked to the airway inflammation caused in asthma? are they linked?

A

Bronchial hyper-responsiveness (BHR)
• Hallmark of asthma – degree of BHR correlates with asthma severity
• Caused by airway inflammation
• A dynamic and reversible process
• Inappropriate and excessive contraction ‘bronchoconstriction’ of smooth muscle
♣ In response to allergen, rapid decrease in lung function within 15 minutes of lung insult
♣ Reversed to normal baseline soon (ACUTE)
♣ In 50% patients, time after that allergic insult, they get a late asthmatic insult 6-24 hours after initial insult
♣ It’s much more severe and prolonged than the initial insult

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

describe the acute and late airway allergen response in bronichial hyper-responsiveness condition

A

♣ AAR = triggered by binding of allergen to membrane bound IgE on mast cells
o Release of mediators (histamines, cysteinyl leukotrienes, PGs)
o bronchoconstriction
♣ LAR = due to influx of inflammatory cells (especially eosinophils)
o Influx of inflammatory cells, mainly eosinophils
o Airway oedema
o Bronchoconstriction
♣ If people have a modest asthma attack and don’t use their inhalers, could get the prolonged attack later which would be much more damaging to the airways

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

what is airway remodelling?

A

• Airway Remodeling - The development of specific structural changes in the airway wall in asthma accompanying long-standing and severe airway inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

what happens to the airway smooth muscle in airway remodelling?

A

o Hypertrophy and hyperplasia (proliferation) of airway smooth muscle cells in response to growth factors (PDGF, endothelin-1) released from inflammatory or epithelial cells = further narrowing of airway lumen
o Release of inflammatory mediators = maintained inflammation
o Irreversible ‘fixed’ airflow obstruction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

what happens to the blood vessels in airway remodelling?

A

♣ Blood vessels
o Increased mucosal blood flow
o Angiogenesis in response to growth factors (VEGF)
o Microvascular leakage from post-capillary venules = oedema & plasma exudation into lumen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

what causes mucus hyprsecretion in airway remodelling?

A

• Mucus hypersecretion
o Hyperplasia of submucosal glands & increased epithelial goblet cells
o Mucus plugs that occlude asthmatic airways – especially fatal asthma
o IL-13 & neutrophil elastase are potent inducers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

what causes fibrosis in airway remodelling?

A

o Aberrant repair to persistent epithelial injury
o Basement membrane thickened due to sub-epithelial fibrosis
o Type III and IV collagen depositions below basement membrane
♣ Triggered by myofibroblasts
♣ Mucus gland = much larger
♣ Thickened SM
♣ Lymphocytes in the airway
o Release of pro-fibrotic mediators (TGFβ) from epithelial cells - associated with eosinophil infiltration
o Persistence of fibrosis = asthma would be irreversible due to the airway narrowing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

what is a potential treatment for asthma to treat airway remodelling?

A

o Bronchial thermoplasty = potential treatment for asthmatic patients – burning SM off the airways which could benefit patients

  • Epithelial layer shedding
  • Goblet cell hyperplasia
  • Collagen deposition beneath basement membrane
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

what are treatments for asthma?

A

♣ Usually controlled using inhaled corticosteroids (ICS) and bronchodilators - long-acting β2-agonists (LABA)
♣ Use both in combination

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

why do some patients not properly adhere to inhaled corticosteroids as an asthma treatment and what does this cause?

A

♣ Could be due to poor adherence of treatment
♣ Compliance increases when a patient has exacerbations
♣ the younger you are, the less likely you are to comply
-could lead to the asthma being more severe
♣ poor communication with healthcare professional –
♣ i.e. not using inhalers right etc.

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

what is the biggest problem of severe asthma to the economy and why can it be hard to treat?

A

♣ Severe asthma accounts for 5-10% patients, but consumes >50% medical expenditure
♣ Different phenotypes of asthma with different patterns of inflammation may benefit from specific therapeutic targeting
♣ different phenotypes require different medications

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

what is COPD and what does it usually result from?

A

A chronic and progressive disease that is characterized by the development of airflow limitation that is not fully reversible and by an accelerated decline in lung function. This usually results from an abnormal inflammatory response of the lungs to noxious particles or gases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

what are 2 main conditions that COPD encompasses?

A

• COPD – encompasses several conditions
o Chronic bronchitis - affects bronchi
o Emphysema – affects alveloli

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

who is most effected by COPD?

A

the elderly

but can also happen in young people

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

what is the burden of COPD in terms of the economy and its prevalence in the world and the deaths its caused

A

• Major global healthcare issue
• Predicted to become 3rd leading cause of death worldwide and ranked 5th for disease burden.
• 1.2m in the UK living with COPD, 2% population (4.5% people >40) = 2nd most common lung disease in the UK
• 2nd highest contributor to respiratory mortality (lung cancer 31%; COPD 26.1%; pneumonia 25.3%)
• 29,776 COPD deaths in 2012 (15,245 males, 14,531 females)
o 2,719 aged 15-64 years
o 27,056 aged >65 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

what is the difference in epdemiology of COPD in developing vs developed countries?

A
  • In developing countries, use biomass fuel which contributes to COPD deaths more so in these countries than smoking which is more prevalent in developed countries
  • Risk factors vary according to geographical area
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

what is the link of COPD and affluence?

A

• Links to affluence
o Highest = north east and north west
o Lowest = east and south east

202
Q

is there a link between smoking and COPD?

A
  • Over 80% of COPD patients are smokers

* Only 20% of smokers become diagnosed with COPD

203
Q

what damage does smoking cause t the airspaces in the lung? what can it cause?

A

• Emphysema – caused by destruction of air spaces in the lungs
Initially smoking causes inflammation in walls between the airspaces then becomes one large space
o Start to lose its elasticity so inflates but can’t deflate properly
o As disease progresses, less and less elasticity
o Affects the amount of area for gas exchange to happen
-Bronchitis - chronic inflammation of the bronchi and fibrosis

204
Q

describe the genetic link to COPD

A

♣ Variable development of chronic airflow obstruction among smokers suggests other factors influence the onset and progression of COPD
♣ Heritability accounts for at least 30% of the variation in COPD risk
- Smoking 1st degree relatives = 3-fold risk
- Non-smoking 1st degree relatives = no risk
♣ Many candidate genes tested for association with COPD – but not replicated
♣ Genome-wide association studies (GWAS) have identified multiple genomic regions that are associated with COPD
- >20 genomic loci associated with lung function (FEV1 levels)
- Several of these also associated with COPD susceptibility e.g. CHRNA3 and CHRNA5 (nicotinic acetylcholine receptor) gene cluster on chromosome 15
• Genetics require some sort of environmental influence to progress
• GWAS- check fev1 levels and addiction (from nicotine receptor)

205
Q

how can a-1 antitrypsin deficiency affect the lungs and the liver? what gene is it encoded by? how can smoking exacerbate these effects?

A

♣ α1-antitrypsin is encoded by the SERPINA1 gene
♣ α1-antitrypsin deficiency is an autosomal recessive inherited disorder affecting 1 in 2000–5000 persons in Europe
- Protease inhibitor
- Produced in liver
- Released into blood stream and goes to lungs where it protects it from neutrophil elastase
o This can impact the lung if it’s not neutralised
- In a-1 deficiency – abnormal form of the protein is made but can’t leave the liver
- Lungs = exposed to the N. elastase
- In the liver, it builds up and so makes scar tissue in the liver so increasing the risk of liver damage
- Cigarettes increase N elastase levels and changes the protein: anti-protease balance

206
Q

what is the pathology of COPD?

A
  • In COPD – alveoli attachments degrade (become detached)
  • Wall thickens
  • Mucus builds up in lumen
  • Diameter of lumen is educed so airflow is impacted
  • Irreversible due to structural changes
207
Q

what area of the lngs does chronic infammation affect? how is this affected in smokers?

A

♣ Chronic inflammation affecting peripheral airways and lung parenchyma
♣ Inflammation increases with disease progression

  • Degree of inflammation – increased in smokers
  • Increased in mild COPD
  • Inflammation progresses even when smoking is stopped
  • (once a certain stage has been reached, stopping smoking will not affect how the disease still progresses)
208
Q

what is the effect of oxidative stress in patients with COPD?

A
  • Increased in patients with COPD from exogenous sources and endogenous mechanisms
  • Have many downstream effects
    o Inflammation
    o Making auto antibodies
    o Decreasing anti proteases
    ♣ Increases emphysema
    o Increase in tgfb
    Patients can be more susceptible to lung cancer as they are not repairing their DNA
209
Q

what is the link between ageing and COPD?

A
  • FEV1: shows our lung capacity increases as we age till 25-30 then progressively declines till 85 where you get 50% FEV1 – senile emphysema
  • In COPD, oxidative stress drives these processes: telomere shortening etc
    o Drives a more rapid decline in the ling ageing
210
Q

describe the idea of airway colonisation in COPD

A
  • In healthy people, macrophages in airways will patrol the airways to look for pathogens and phagocytose these to maintain the health
  • Efferocytosis
  • In COPD lung, there’s defect in alveolar macrophages and so have a decreased ability to phagocytose bacteria and so cannot
  • Becomes necrotic and breaks open
211
Q

what are frequent exacerbations and how do they affect COPD patients?

A

Frequent exacerbations:
♣ worse prognosis
♣ more hospital admissions
♣ faster disease progression
♣ worse health status
- Pathogens leaving airway susceptible to bacterial infection
- In COPD If you can’t clear this, leads to more issues in the airways
- Have high level of air pollution which can contribute to exacerbation – not as common
o Increase in inflammatory cells with its cytokines and also an increase in the sympotomoology (cough etc.)
o Hospitalised and so decreases quality of life
- In severe cases, causes death
- Risk = stronger If you are ‘frequent exacerbation’
o Periods of exacerbations drive a faster disease progression

212
Q

describe airway remodelling in COPD patients

A
  • Goblet cell causes increase in mucus
  • Neutrophils in sputem (what they cough up)
  • Contrast with asthma which mainly had eosinophils
  • Rather than th cells, have cytoxic X
  • In normal epithelia, severe asthma, ep cells slough from airway
    o In COPD, get squamous epithelium which is a pre-change that could become lung cancer
    o Could reverse back (not always fated to become cancerous)
  • Small increase in airway SM
  • No real BM thickening as in asthma
213
Q

what is the condition called that shows that there is a link between asthma and COPD?

A

Asthma-COPD overlap syndrome (ACOS)

214
Q

describe what is known about ACOS?

A

• Increased eosinophil count in COPD patients as well as the usual neutrophil count for COPD
o high-dose ICS, IL-5, IL-13, IL-33 blocking antibodies
o high does corticosteroid would work on this ohenotype as its asthma like
• neutrophilic infammationin asthma patients (would usually be seen in COPD patients)
o CXCR2 antagonists, phosphodiesterase 4 inhibitors, p38 MAPK inhibitors, IL-1 and IL-17 blocking antibodies, macrolides
• Asthmatic pateints with irreversible airflow obstruction
o inhaled combination therapy of corticosteroid, long-acting b2-agonist, long-acting muscarinic antagonist

215
Q

what are the available treatments for COPD?

A

• Management of stable disease:
o Reducing exposure to irritants & pulmonary rehabilitation
o Relief of symptoms – bronchodilators (long-acting β2-agonists (LABA) and/or long-acting muscarinic receptor antagonists (LAMA))
o Reducing risk (mainly of exacerbations)
• Management of exacerbations:
o Oral antibiotics and possibly oral corticosteroids (use of inhaled corticosteroids restricted)
o bronchodilators (long-acting β2-agonists (LABA) and/or long-acting muscarinic receptor antagonists (LAMA))
• Virus is a predominant trigger but there isn’t a way of curing this

216
Q

what areas are infected in respiratory tract infections?

A

the nasal cavities, pharynx and the the airways

217
Q

what are the 2 divisions of RTIs?

A

upper and lower RTIs

218
Q

what areas are affected in upper RTIs? what conditions arise from this?

A
upper RTI: affects the nasal cavities (nose and sinus) and the pahrynx (throat)
nasalpharyngitis
sinusitis
larangitis
pharyngitis
influenza
219
Q

what areas are affected in lower RTIs? what conditions arise from this?

A
lower RTIs: affect the airways (trachea, bronchioles, bronchi and alveoli)
bronchitis
bronchioitis
influenza
pneumonia
tuberculosis
220
Q

what is the burden of RTIs globally?

A

it is the 3rd leading cause of death in the world

important cause of death in both children and adults

221
Q

how common are viral RTIs? what is the occurrence in adults vs children?

A

children - get it 4-5 times anually
adults 1-2 times anually
by 75years old, will have had ~200 colds in your lie tme

222
Q

what are the 6 major respiratory viruses and briefly describe their occurrence throughout the year

A

adenovirus - low levels throughout the year
influenza A - variable incidence amount and time. very high peaks in the winter, short peaks throughout the year but affects a lot of people
influenza B - very low levels generally, peaks occasionally
parainfluenza - very low levels generally, peaks occasionally
respiratory syncytial virus - peaks every year generally at winter and affects a large amount of people
rhinovirus - spikes all throughout the ear and is detected much easier now

223
Q

what have been the stages of equipment used for various diagnoses of respiratory viruses? what has been come up with now?

A

PCR - in the 1990s, greatly improved the detection of respiratory viruses (highly sensitive). but, it cant detect what you don’t already know as a known primer is needed for it to work
High throughput screening - allows us to screen for a high number of viruses but the sensitivity isn’t good enough for clinical samples and also it is very costly and less sensitive than PCR
they have now come up with VirCopSeq-VERT which is a combination of HRT and PCR. it allows high throughput screening of viruses but with a higher sensitivity. this allows for much more effective screening

224
Q

what is the major cause of RTIs and bronchitis in infancy and childhood?

A

respiratory syncytial virus (RSV)

225
Q

when are people likely to get RSV in their life?

A

60% infants have been infected with it in their first ‘RSV season’ (i.e. winter)
by 2-3 years old, almost all children will have had it
it induces protective immunity which tends to wane overtime
RSV sis also a major cause of respiratory infection in older patients with cardiovascular comorbidity, COPD or immunocompromised patients

226
Q

what is a major determining factor as to what kind of disease you will get if you are infected with RSV?

A

most kids who get RSV in their first year of life get bronchitis due to the immaturity of their immune system
as they get to 2-3 years old, their immune system matures by getting more th1 which is protective and losing their th2 bias
if they do get bronchiolitis due to RSV, they are more likely to get post bronchiolotic wheeze which is pre asthma

227
Q

what is the virology of RSV?

A

it is an enveloped negative ssRNA of the paramyoxoviridae family

228
Q

how is RSV recognised by the host cell?

A

by many proteins as it is a ssRNA which contains a high number of nuclei
the G proteins are highly glycosylated and are know to attach the virus to the host cell
fusion proteins work to allow the virus to fuse with the host to allow its entrance in the cell which then allows the virus to self replicate

229
Q

how does G proteins allow RSV to be attached to the host?

A
  • uses annexin 11 o airway epithelial cells

- uses L.selectin and CX3CR1 on immune cells

230
Q

what are the ways that RSV can be detected by the host?

A

RSV can be detected by multiple PRRs

  • TLR2/6 – not known how
  • TLR4 - fusion f protein
  • TLR3 - dsRNA replicates
  • TLR7 - ssRNA
  • RIG-1 - ssRNA viral genomes bearing 5’ triphosphates; dsRNA replicates
  • Nod2
  • NLR3P/ASC - small hydrophobic (SH) RSV viroporin
231
Q

what is the innate response in the cell in response to RSV?

A

RSV infects the epithelial cells lining the repiratory tract ad will activate PRRs
this will cause inflammation and therefore the relsease of interferons, anti-viral cytokies etc which will lead back to the epithelia that was infected as well as neighbouring clels and so will initiate cell apoptosis
other mediators eg TNFa (proinflammatory) and chemokines eg CVCL8 and CXC17 will recruit neutrophils and NK cells causing death to cells that cause this. can also phagocytose and remove the debris

232
Q

what is the adaptive response in the cell in response to RSV?

A

RSV infecting dendritic cells will migrate to regional lymph nodes and present the antigen to cd4 helper t cells causing activation of b cells, activation of ABs and also cytotoxic t cells. this leads to a cycle of inflammatory cell recruitment and inflammation. this also could lead to bronchiolitis etc or otherwise just a cold which passes in ~7 days

233
Q

what are the 4 ways inside the cell that RSV can continue to infect you throughout your life?

A
  1. NS@ can bind to RIG1 to prevent it from interacting with MAVS which prevents/severely reduces this signalling pathway and so limits possible inflammation
  2. NS1 can bind to IRF3/7 (tf which generates type 1 interferons and so reduces the ability of this cell to respond
  3. fusion f protein and nuclear protein can induce type 1 interferon production - not sure yet how - and then NS1 and NS2 bind to STAT2 which degrades STAT2 and reduces its ability to release interferon stimulated genes
  4. F protein can bin to TLR4 and down modulate (prevent) signalling – and so reducing the host response
234
Q

what are the 4 ways outside the cell that RSV can continue to infect you throughout your life?

A
  1. secreted G protein (attachment protein) can be secreted from the cell and reduce TNF and interferon production by binding CXC3RI both on dendritic cells and CD4 cells
  2. can act as AB decay (mop up any ABs you have that are generated against RSV - by inhibiting IFN-Y production)
  3. can inhibit synapse formation between dendritic cells and t cells (by N protein)
  4. inhibit dendritic cell maturation by NS1/NS2
235
Q

what are the features of the RSV disease?

A

cytopathic effect of virus and local inflammatory response
- airway epithelial cells highly permissive to RSV
- RSV replication –> epithelial damage –> necrosis
- submucosal oedema
- mucus secretion
- bronchoconstriction
all this causes severe obstruction of airway lumen

236
Q

what is a typical feature of cells that have been infected by RSV?

A

they will have cells stuck together (fused) which form a syncytium which is when the neighbouring cells stretch and fuse together. this is a very destructive virus

237
Q

what was the problem with the treatments made for RSV?

A

formalin was a drug trialed but caused o protection, created a higher viral load, made a16-fold increase in hospitalisations and caused 2 infact fatalities

238
Q

what happens in the normal cell with RSV vs when formalin is added?

A

in the normal cell, RSV bids to TLR4 on the cell surface wand is then internalised which then triggers the activation of RIG1, MDA5 and TLRs which are all in the endosome. this causes a large tf activation and antibody affinty maturation.
when formalin is added, the RSV is inactivated due to the formalin and so although it bind to the TLR4 on the surface, it cant be internalised because as it cant replicate and so there is limited activation of the RIG1 etc and no real affinity maturation. however, this means that the system is primed so when an active RSV does come along, the response that it will create will be extremely massive

239
Q

what treatment has been proven to work for RSV in high risk infants?

A

monthly injections of anti-RSV f protein monoclonal Ab (palivizumab)

240
Q

what is the supposed link between RSV and asthma and what can it be caused by?

A

severe RSV infection in infancy has firmly been established to be a risk factor for subsequent asthma in later childhood and leads to a 4 fold higher incidence of asthma
the underlying biological mechanisms are not yet fully understood. thought to be due to:
- chronic epithelial and airway reactivity changes to developing infant lung
- lung injury altering lung function
- immunomodulatory changes (th1-th2 changes)
- genetic factors that impact on pattern of immune response to infectious agents

241
Q

how is it supposed that RSV bronchiolitis alters the Th1/th2 immune response?

A

th2 polarisation to the lung immune response happens which enhances the th2 sensitisation to aeroallergens and induces development f a chronic asthma phenotype

242
Q

what does the geneic background determine about the risk of RSV hospitalisation?

A

it determines ~20% variation

243
Q

what is an example of a susceptibility gene studies for its involvement in RSV?

A

TLR4 mutations Asp299Gly and Thr399lle over-represented in infants <12months of age hospitalised for RSV bronchiolitis compared to infants who have mild RSV
fail to translocate TLR4 to the cell surface and so there is a reduced NFkB signalling because they don’t, don’t detect the virus until its already inside the cell and replicating

244
Q

what is the difference between RSV and human rhinovirus in terms of them being triggers or inducers of RSV and HRV?

A

RSV is more of an inducer than a trigger of getting the condition - mostly children/infants affected in short epidemic seasons
HRV is more of a trigger than an inducer - anyone of any age can be infected in any season

245
Q

what can HRVs cause?

A

most common cause of upper RTIs
causes relatively benign upper respiratory tract illnesses but also exacerbations of chronic pulmonary diseases, asthma development, severe bronchiolitis in infants and children and finally it can cause a fatal pneumonia in the elderly and immunocompromised patients

246
Q

what proportion of the common colds we suffer is due to HRVs?

A

2/3rds of the colds we suffer

247
Q

what are the upper and lower respiratory infections that HRVs can cause?

A

URI: the common cold, acute otitis media (fluid build up in the ears) and rhinosinusits
LRI: bronchiolitis, community-acquired pneumonia (very common) and croup(less common)

248
Q

what do HRVs cause in healthy adults?

A

cause a self-limiting syndrome with a predominantly upper respiratory tract manifestation in healthy adults

249
Q

describe the basic virology of HRVs

A

it is a ssRNA virus - in the largest subgroup of picornaviradae family with over 150 serotypes having been identified
they are small non enveloped positive ssRNAs

250
Q

how are the serotypes of HRV categorised?

A

it was originally based on the receptor:

  1. intracellular adhesion molecule 1 - ~96 major viruses (major)
  2. low density lipoprotein receptors - ~12 minor viruses(minor)
  3. cadherin related family member 3 - ~50 ‘c’ viruses (relatively new)

another way to classify = genetically:

  1. HRV-A - 77 serotypes(ICAM-1+LDLr)
  2. HRV-B = 30 serotypes (ICAM-1)
  3. HRV-C - ~50 serotypes
251
Q

how do HRVs differ from other viruses in terms of how it infects airway epithelial cells (AEC)?

A

respiratory ciruses enter and replicate within the airway epithelial cells. HRV infect nasal epithelial cells and sometimes has been detected in lower airways
HRV is rarely associated with cytopathology of AECs
-AECs - sloughed by airway lining remain structurally intact
- disruption to barrier function by dissociation of zena occludens 1 from tight junction complex
in contrast to other viruses, the airway remains structurally intact - disruption in barrier function can occur (e.g. tight junctions are effected) which an lead to bacterial infection

252
Q

describe the signal transduction pathways and activation of the innate immune response in HRV

A

HRV binds to receptor (LDLr) gaining entry to the cell and activating PRRs both within the endosome and in the cytoplasm. this leads to release of cytokines, chemokines and b cells (eosinophils) and t cells (PMNs). HRV can also activate TLRs (not sure how this is)

253
Q

describe the acute exacerbations that arise due to HRV/RSV. what are therapeutic options for this?

A

acute exacerbations:

  • further increase in airway inflammation
  • major cause of morbidituy and mortality and accelerates disease progeression
  • significant healthcare costs - asthma:50%, copd - 70%)
  • frequency = major determiant of health status and quality of life

HRV = frequent cause of exacerbations in patients with underlying airway disease - 50-65%
same number of infections but associated with higher inflammatory response and therefore with respiratory tract infections
RSV causes exacerbations in the older populations wth cardiovascular morbidity, COPD or immuno compromised patients
therapeutic options - targeting exacerbations; currently inadequate

254
Q

why is it especially dangerous for HRV in asthma/COPD patients? describe what happens inside the cell

A

HRV infection binding to epithelial cells on the surface (receptors) and also interacting with alveolar macrophages and causing the release of chemokines and cytokines and thus leading to chemotoxins and immune cells. also leads to release of other cells impacting on airway smooth muscle and fibroblasts which is detrimental in asthma and COPD patients due to these changes to the airway

255
Q

is there an increased access to epithelial cells of HRV

A

there is some evidence suggesting that structural changes to the lungs (epithelial damage by HRV) couldredisposethe to be more susceptible to viral infections

256
Q

what studies have been done to show that there is a greater response to HRV in COPD and asthma patients. are there any supporting contrasting studies?

A

an experiment using 3 groups of people (asthma patients, COPD patients and healthy controls) were all exposed to HRV
found that the viral loads were higher i asthma and COPD patients than in healthy controls
one of the factors could be interferons - these could block viral entry and so control viral transcription ad translation
cleavage of RNA and induction of apoptosis could occur
- could lead to failure to induce a robust IFN response= uncontrolled viral replication and increased inflammatory responses
in a 2nd study, they found that asthma patients had a defective type 1 response to the rhinovirus. another group studying the same thing found the same results
however another group studying this found no difference

the first group studying this found no difference in well controlled asthma (with no interferon deficiency) and so it was in severe therapy resistant atopic asthma

257
Q

is there a susceptibility phenotype to HRV?

A
- frequent exacerbations = more susceptible to viral infection and/or more severe clinical disease after infection
in asthma: could be partly due to deficient IFN production as they aren't producing enough interferons and so get much more replication and acute exacerbations
other factors-- less antiviral th1 cytokines(higher viral load)
higher SOCS1 (suppressor of cytokine signalling 1) May link th2 inflammation with IFM deficiency
more ICAM1 expression which is the receptor that binds majority of the HRV serotypes
in COPD: deficient IFN production could be with more ICAM-1 expression
258
Q

what are the possible mechanisms in which HRV increases susceptibility to bacterial infection?

A

1 - dissociation happens because of increased ROS generation which means that bacteria can gain access to epithelial cell wall to colonise it
2 - increased expression of ICAM1: the virus can increase expression of this molecule (an its internalisation). this can also aid the internalisation of s.aureus
3 - increased expression of platelet activating factor receptor (PAFR) which is a way pneumonia gets access to cells - can cause susceptibility to bacterial infection
4 - macrophages: HRCV can infect macrophages (dont replicate in them but can cause changes i.e. less inflammatory cytokine release in response in response to bacteria and so therefore to response and kill bacteria

259
Q

what are some novel approached to treating virus induced exacerbations of asthma and COPD?

A
  • immunomodulatory strategies: inhaled interferons can enhance the innate anti-viral immune response. this is most effective when the interferons are given at the same time as the virus but the incubation period for HRV is 1-2 days before you know you are infected so it is too late
  • targeting inflammatory modulators and binding to them and their receptors
    antiviral techniques:
    blocking viral replication and function
    blocking viral binding and entry
    vaccination is difficult to develop one that will target all the different serotypes ofHRV
260
Q

why do the lungs need to sample a lot of air and what problems can this cause?

A

lungs: need to get very small molecules into the body
15-18 times a minute, theres gas in the alveoli and so the lungs and contents sample alot of air. needs to have a thin interface to be able to do this properly. have a 75m2 area which is vulnerable to infection

261
Q

what sort of pathogens can infect the pharynx and what can it cause?

A

phayrngitis - viral usuallly(70-80% of the time) and so doesnt require treatment
sometimes,can be bacterial which are treated
- infected by:
–group a steptococcus (b haemolytic) which is identified by how it grows on culture with blood on it
– mycoplasma pneumonis - not easily spotted/doesn’t grow well in culture
– fusobacterium necropphorum - invades into the veins and neck and so could affect the jugular vein, drains int the heart into the p.artery then drains into the lungs

262
Q

how is group a streptococcus (b hemolytic) identified in culture?

A

it is grown in culture with blood and reacts with the haemoglobin then turns transparent

263
Q

what colour membrane is formed in the pharynx during pharyngitis and why is this? how can this be treated?

A

• grey membrane formed due to bacteria releasing toxins to kill the epithelial cells. A gram-positive infection – Corynebacterium diptheriae
o use antitoxins (AB produced to soak up all the toxins used with antibiotics)

264
Q

what is sinusitis and how do people get it? are they bacterial or viral infections? what are the treatments available for both?

A

• sinuses have narrow openings and Is lined with mucosa, this could block the entrance which fills sinuses with puss. So, causes pain around face.
o Sinus should have air but in this condition, its filled with fluid
o Usually a virus but could be bacterial
o Viruses get better themselves

265
Q

what is a disease that is normally seen in children and caused by haemophilus influenza? why is it rare now? what is its usual structure?

A

• Acute epiglottitis – normally seen in children and is caused by haemophilus influenza
o Rare now due to new vaccine (HiB)
o Mostly capsulated but some = not encapsulated which can’t be treated

266
Q

what causes the whooping cough? can it be treated?

A

• Bordatella pertussis – whooping cough. Has many virulence factors so get irritation of respiratory tract which causes the whooping cough
o Have vaccine against it now

267
Q

what is bronchitis? is it caused by a virus or bacteria?

A

• Bronchitis – inflammation in large airway. Triggered by virus
o Most pathogens = viral
o Get some bacterial – rarely a cause in healthy bacteria

268
Q

what is pneumonia generally caused by? how can it be diagnosed and what is the hospitalisation/mortality rate?

A

o Build-up of white material in basis of lungs.
o The air sacs are filled up with secretions, dead cells and dead bacteria
o Listen as lung expands, hear crackles etc.
o 1 In 100 mortality
o stay in hospital ~ 1 week – costs a lot of money

269
Q

what is the pathogenesis of pneumonia? where does it hit the most and why does it cuse breathlessness? what is the cause of inflammation here?

A
  • alveoli = weak spot – hard to get pre-existing defences as then you wouldn’t be able to do gas exchange properly
  • alveolar macrophages do tissue repair and phagocytosis
  • when pathogen arrives, senses it using TLRs, phagocytose it
  • can do it more effectively for some bacteria
  • if you have a pathogen that’s able to subvert phagocytosis or too many pathogens, causes suddenly lost of activity here
  • pro-inflammatory response calls in neutrophils. But if they keep coming in, the normal activity of the alveoli won’t be able to work properly as they would kill the bacteria but then also self-destruct.
  • Will become breathless as you can’t do gas exchange with all this stuff in the way
    o Oxygen levels decrease
  • Inflammation causes pain
  • Inflammatory response causes fever
270
Q

describe the reslution phase of pneumonia and the

A
  • There’s a resolution phase which clears the airways and allows you to recover
    o Wait 6 weeks for repeat x ray which allows all that stuff disappears
271
Q

what are the symptoms of pneumonia?

A
  • Symptom of pneumonia - If lining of lung is inflamed, as lung expands, has to move up to chest wall, makes it sticky as the lung lining which feels rough – very painful
    o Plurosy
272
Q

what is the agent causing 40% of pneumonias? how can they be treated? what is the problem with the available treatments?

A

• Streptococcus pneumoniae – 40% of pneumonia
o Draftsman colony (gram positive cocci)
o See lots of dark purple cocci
o Split into alpha and beta
o Sugar coating the bacteria – very variable
♣ Abs we produce recognise serotypes and so protects us next time but there are so many variations that this won’t be effective for all the other 99 types
♣ Need multivalent vaccines with conjugates
♣ Have 13 valent conjugate vaccines
• If treated with this, these types reduce but the other (85 types) would increase

273
Q

what type of pneumonia arises due to warm air?

A

• Legionella pneumoniae – due to warm air (in hotter climates and bad air con etc.): warm air then bacteria = attracted to it

274
Q

what is the best way to diagnose atypical pathogens? why cant they be diagnosed normally?

A

Atypical pathogens – don’t behave the same as when you try to diagnose them in the lab
The best way to find what pathogen is in the respiratory system = to grow phlegm in culture
Sometimes able to see antigen in the urine –legionaella and streptoccochus

275
Q

what can a ball of pus with air spaces in the lungs cause ? why does this happen?

A
Ball of pus with air spaces in the lungs which communicates with lining of lungs and = outside the lungs. Get infection in lung which causes …
	Lung abscess (unusual)
	Due to lung compromise or bacteria
276
Q

how are the possible infections different when the immune system isn’t working very well

A

If immune system isn’t working very well, get a broader range of bacterial viruses and fungi causing infections

277
Q

who are affected by chronic lung infections?

A
  • those who have a compromise that makes them more prone to this i.e. getting pulmonary tuberculosis
278
Q

what is the microbiology of Tb?

A
  • Tb has a very thick waxy cell wall
279
Q

why is thought that Tb is able to survive in very harsh conditions?

A
  • Used to be an environmental organism then evolved to be a human disease
    o The fact that it used to be in the environment, means that its used to surviving in very harsh environments/conditions
  • Key components: mycolic acids and waxes
  • It’s very good at surviving inside the cells that try to kill it
280
Q

why is Tb considered to be slow and called consumption?

A
  • Tfb =alcohol fast bacilli (AFC)
  • Takes long time to make the thick waxy coat and so takes long time to develop/replicate
  • Bacteria grows at a 25 times faster rate than Tb
  • Won’t be able to see samples of it early on as it takes 3-4weeks to grow to an amount to be able to see
  • Grows slowly inside humans
281
Q

who is most likely to get Tb and how is it spread?

A
  • Resource poor countries have a higher burden of Tb
  • Tb = spread in environments where people live closely together where there is not much circulation
  • Incidence is high in non-UK born compared to UK born
    o Within UK born, if you have family from countries with high Tb prevalence, there is a higher incidence of Tb in your family
    o More common in bigger cities
  • Tb used to be called consumption
    o Because it’s a slow insidious process (losing weight etc.)
282
Q

How do you catch Tb?

A
  • Only can catch it if someone is coughing up tb germs not the environment
  • Not common as its heavy droplets that would drop to the ground and the light gets rid of it
  • Must be very close to catch it
  • If you are smear positive you’re would be diagnosed as having Tb
    o Can detect how likely you are to spread Tb into the community
283
Q

How does a granuloma form in Tb?

A
  • Tb into lungs and macrophages try to kill it
  • Not possible to kill all
  • Get a response that because macrophage and to kill the tb, t calls more cells to wall of the tb to stop it from spreading
    o Called a granuloma
  • Can withstand phagolysosome
284
Q

what is the process of antigen presenting of tb cells?

A
  • macrophage starts to take part of the protein from the tb and present it from t cells (it’s a APC), must take it to lymph nodes to find t cells to kill it
  • Cd4 t cells make pro-inflammatory cytokines which activates more macrophages to make them better at killing the tb
  • Cells surrounding the tb = are fibroblasts which seal in the tb cells with the tissue and etc. becomes puss. Forms tubercles
  • If this works kills microbacteria/stop it from working
  • Sometimes can kill this so the microbacterium can exist in a low activity state for many years
285
Q

what happens to Tb cells at lymph nodes

A
  • Tb may land and there is a response which could involve clinical disease in a small number of people
  • At the local lymph nodes, activate t cells to activate more macrophages
  • Granuloma expansion becomes an abscess
286
Q

what does a granuloma expansion become?

A
  • 5-10% people get an abscess growing in their lungs
  • soon this abscess will grow and move into the bronchioles
  • this has all the microbacterium in it which when coughing would come out into the environment
287
Q

how can you diagnose Tb?

A
  • to diagnose Tb you need to do special stains that don’t wash out with alcohol
    o its AFB (alcohol acid farse)
  • increased diagnostic sensity for AP
  • increase chance of finding Tb with the more samples of sputem you get to analyse
288
Q

what was the culture that used to be used to grow Tb? and how long does it take? what is used now?

A
  • Lowenstein jenson = used to be used as a culture to grow Tb
    o Takes 2-8 weeks in total
  • Found that liquid cultures are much better and are now used all over the UK
    o Takes 4-8 weeks in total
289
Q

how ca PCR be sed to grow Tb in culture

A
  • Can do PCR
    o Alternative to smear
    o Will tell you more before your culture is ready
    o Better sensitivity
    o Could spot any resistance strains using the rapid PCR
290
Q

what can happen if Tb gors beyond the lungs? does everyone with Tb cells get Tb? what happens if not?

A
  • Tb has the ability to go beyond the lungs
  • If it goes into the blood stream with macrophages with the Tb, could cause a granuloma and cause a disease or stay latent till it ends up anywhere in the body 30 years later i.e. in the brain, kidney etc.
  • 90% of people who have Tb, actually don’t get Tb till very late
  • 10% f these never get it
  • Tb could be sitting there and wait till it wants to reactivate
291
Q

what happens if some part of the immune system fails in Tb response? how can this be treated?

A
  • If some part of the immune system starts to fail, t cell function doesn’t work and so the Tb bacterium reactivates
    o Treated with immunosuppressant therapy
    ♣ HIV = increased risk as this is where the t cells don’t function
    Cd4 cells could be depleted or TNF alpha in response to granulomas breaking down
292
Q

describe Nocardia - where it is found, who is affected and how they become infected. also, how it is visualised

A

• Nocardia – found in the environment
o E.g. decaying planst soil water etc
• Those who are affected = those whose immune system is compromised
o I.e. diabetes, cancer HIV aids
o Chronic infections i.e. connective tissue disorders
o Alcoholism
o Transplants/corticosteroids
• (compared to Tb where anyone can be affected)
• infection = through breathing in dust that contains the bacteria
• when a hospitalised patient is infected from contaminated medical equipment
• can be seen as lacy white coverings on lungs on an x-ray

293
Q

describe meliodosis: where is found, how it affects people and who is most at risk

A
  • Melioidosis – found in tropical climates
  • Slow onset, systemic symptoms but also local systemic symptoms
  • Similar people as in nocardia which are at risk
294
Q

describe Non-Tb mycobacteria ‘NTM’

A
  • Non-Tb mycobacteria ‘NTM’
  • Microbacterium that generally lives in the environment
  • In healthy lung, can’t cause disease as its not able to live in the harsh conditions and escape the phagocytes
  • Those with their immune paresis or bronchiectasis issues would be more susceptible
  • Behaves like Tb if it has these advantages of underlying issues in the airways etc
295
Q

describe how you get chronic pulmonary disease with Histoplasma capsulatum

A
  • A mould that lives in dirt
  • In southern US and dryer pats of southern America
  • Considered in patients with pneumonia with mediastinal or hilar lymphadenopathy ..,
  • In bats – so those in caves
296
Q

how can interstitial lung disease be physiologically measured?

A

o Restriction of lung volumes (measured by FVC)
o Reduction in lung gas transfer efficiency (DLCO / TLCO)
o Hypoxia (low blood oxygen levels), particularly on exertion / exercise
o Reduction in exercise capacity

297
Q

how can chest x rays be used to test for ILD?

A

• Flattening of diaphragm
• (IPF = idiopathic pulmonary fibrosis)
o x ray shows all the fibrosis
o x ray = not great at picking up restrictive lung disease (till it’s at advanced level)

298
Q

how can spirometry be used to diagnose ILD?

A

o Look at FVC and FEV1
o FVC = total amount of air in 1 breath?
o FEV1= how much air someone can breathe out in 1 second
o ILD = restrictive lung disease: reduction in FEV1 and FVC
o In obstructive: FVC = normal/increased
o Use ratio between FEV1: FVC
o Less that 70% = obstructive

299
Q

what happens in the body to cause ILD to happen? why do patienst get breathless?

A
  • ILD = disease of gas exchange
  • Difficulty getting oxygen to diffuse across barrier of alveoli int blood stream
  • ILD- barrier is thickened (fibrosed) so oxygen has much difficulty diffusing across into the blood stream
  • Why patients get breathless
  • CO2 = usually fine to get across as it can be easy to go across the barrier
300
Q

what is the transfer factor breathing test? how can it be used to test for ILD?

A
  • Uses CO to estimate how well the lung is functioning
  • Measures amount of CO left I lungs when patient breathes out
  • Less CO left in lungs after patient breathes out, the better the lungs are functioning
  • In ILD, O2 struggles to diffuse into bloodstream get more CO left in lungs
  • Will have reduction n transfer factor in ILD patients
301
Q

how can exercise be used to test for ILD?

A
  • Incremental shuttle walk test
  • Walking bleep test around 2 cones
  • Or 6 minute walk test
  • Walking continuously around 2 cones for 6 minutes
  • Studies show that both test correlate with lung function
302
Q

describe the pathology of ILD including what causes it, the risk factors for it and its characteristics on radiology/histology

A

• Heterogenous mix of lung disease
o Inflammation
o Scarring

•	IPF (unknown cause)
•	Cause unclear, hence ‘idiopathic’
•	Fibrosis of lung parenchyma
•	Affects males more than females
•	More elderly patients
•	Smoking = risk factor
•	Characteristic radiology / histology 
o	Usual interstitial pneumonia (UIP)
303
Q

what is HRCT = high resolution CT scan

and how can it test for ILD?

A

• Fibrosis on periphery and basis of lungs
• Honeycombing (destruction of the lungs)
• Traction bronchiactosis?
o Airways being widened as scarring/fibrosis = pulling and making them wider
• In a subpleaura
• As to be in UIP pattern or it can’t be IPF

304
Q

how is IPF diagnosed -what must be investigaed? what happens in the myofibroblasts and what do they deposit?

A

IPF Diagnosis – Investigations
¥ HRCT – UIP pattern fibrosis
¥ Rule out other causes of UIP
¬ Signs and symptoms of autoimmune conditions (e.g. RA)
¬ History of environmental / occupational exposures (including asbestos)
¬ Bloods

¥ Fibroblasts repair damaged tissue
¥ Lungs get damaged
¥ Fibroblasts migrate to the lungs (to try and fix the damage), becoming myofibroblasts
¥ Myofibroblasts deposit collagen in the extracellular matrix
¥ Myofibroblasts proliferate & form fibroblastic foci
¥ The thickened tissue leads to lower gas exchange efficiency in the lungs

305
Q

what is precision medicine and what does it aim to achieve?

A
  • Sequencing people’s genome
  • Treating people with the same disease in different ways based on phenotype, differences in immunity, different co-morbidities etc.
306
Q

what are the different genes that affect peoples risk of getting IPF?

A

• Mutations in genes involved in the maintenance of telomere length
• Variations in genes that are responsible for cell adhesion and integrity
• A single-nucleotide polymorphism in the promoter region of MUC5B substantially increases the risk of IPF
o Most important= MUC5B = significantly increases risk of getting IPF
Lung microbiome in IPF
¥ Increased risk of disease progression in patients with IPF
• Increased overall bacterial burden
• Abundance of streptococcal and staphylococcal organisms
¥ Risk of IPF is increased by genetic variation in TOLLIP
• Gene encoding a protein that inhibits responses to microbes

307
Q

what is thought about the importance of bacteria in IPF? how does this relate to treatments for it?

A

Think that bacteria in lungs = important in IPF

Thinking of giving patients antibiotics to stop microbes in the lungs, may slow down progression of IPF

308
Q

what are the Key Features of IPF?

A

¥ Collections of fibroblasts (fibroblastic foci)
¥ Thickening of alveolar interstitium
¥ Destruction (honeycombing) of alveoli
¥ Affects periphery and base of lungs (UIP pattern fibrosis)
¥ Spatial heterogeneity (normal lung tissue next to abnormal tissue)

309
Q

what is hypersensitivity pneumonitis? how does its pathogenesis differ from IPF?

A

¥ Immune mediated hypersensitivity reaction in a genetically predisposed individual
¬ Recurrent exposure to environmental agents (HP inducers)
¥ Pathogenesis very different to IPF
¬ Inflammation & air trapping
¬ May progress to fibrosis and look like UIP pattern on HRCT (chronic HP)

310
Q

what is Type III Hypersensitivity caused by and what does it result in?

A
  • Pigeon droppings/feathers causes the inflammatory reaction
  • Lymphocyte produces abs to the ag
  • Abs attack themselves to ag
  • Need Prior sensitisation is required (i.e. previous exposure to antigen)
  • IgG antibodies produced by lymphocyte retain immune ‘memory’ of antigen
  • On secondary exposure, antigen-antibody complexes are formed and deposit in lungs and not adequately cleared
  • Repeated exposure leads to repeated symptoms and physiological decline
311
Q

what is hypersensitivity pneumonitis associated with? how does it cause tolerance?

A

¥ HP associated with increase of
¬ CD4+ T cells
¬ CD4+ /CD8+ ratio
¬ Th2 activity
¬ CD4+ Th17 cells involved in the development of lung fibrosis
¥ Tolerance may be mediated by regulatory T cells
¥ Many exposed individuals develop a mild lymphocytic alveolitis but remain asymptomatic

¥ It is likely that a mixture of antigens, rather than a single antigen, contributes to the sensitization and evolution of HP

312
Q

what is the Hypersensitivity Pneumonitis occupational inducer?

A

• Metalworking fluids (MWFs)
o Industrial factories that cut/grind metal
o Increased over last few years
o Coolant put into fluid after metal = cut
o Can cause HP

313
Q

how can hypersensitivity pneumonitis be classified?

A

¥ Traditional classification
¬ Acute (symptoms 4-6 hours post exposure)
¬ Subacute (symptoms weeks – 4 months)
¬ Chronic (symptoms 4 months – years)
¥ Suggested novel classification (Vasakova et al, 2017)
¬ Acute (symptoms <6 months)
¬ Chronic (symptoms >6 months)

314
Q

describe acute Hypersensitivity pneumonitis including the HRCT features

A
•	Mostly reversible with complete resolution possible
o	Different from IPF – which is progressive over time but in HP can recover
•	HRCT features:
o	Upper / middle lobe 
o	ground-glass opacities 
o	Poorly defined 
o	centrilobular nodules 
o	Mosaic attenuation 
o	Air trapping
o	Consolidation (rare)
315
Q

describe chronic HP including its HRCT features. describe the sparing of teh bases idea

A

¥ Potentially reversible
¥ Risk of progression

¥	HRCT features:
o	Upper / middle lobe fibrosis
o	Honeycombing
o	Mosaic attenuation
o	Air trapping
o	Centrilobular nodules
o	Sparing of the bases
¥	Can get similar abnormalities in IPF and HP
¥	But in IPF, affects the basis
¥	In HP, spares the basis
316
Q

what is the histology behind the diagnoses of HP

A

¥ Classic triad:
¬ Chronic cellular bronchiolitis
¬ Cellular (i.e., lymphocytes and plasma cells) chronic interstitial pneumonia
¬ Non-necrotizing granulomas and/or giant cells

317
Q

what are the most common CTDs causing ILD?

A

o Systemic sclerosis (SSc)
o Rheumatoid arthritis (RA)
o Idiopathic inflammatory myositis (IIM)

318
Q

what are some CTDs that cause ILD (not the most common)

A

o Sjogren’s syndrome (SS)
o Systemic lupus erythematous (SLE)
o Mixed connective tissue disease

319
Q

is there a link between ILD and CTD? what can ILD occur in the presence of?

A

• ILD leading cause of mortality in CTD
• Up to 15% of the patients initially diagnosed with idiopathic NSIP are later found to have CTD
• ILD can also occur in the presence of autoimmune features that do not meet classification criteria for a specific CTD
o Interstitial pneumonia with autoimmune features (IPAF)

320
Q

describe sarcoidosis including what its caused by

A

¥ Granulomatous disease characterized by the presence of non-caseating granulomas involving multiple organ systems
¬ Lungs affected in over 90% of cases
¬ Significant variations according to age, race, gender and geography

321
Q

why is a mutlidisciplinary approach required to treat ILD?

A
o	nutrition (some patients lose weight)
o	prevention vaccinations
o	adherence to medication
o	psychological assistance (patients and their families)
o	palliation of symptoms
o	long term oxygen therapy
o	management of infections
o	lung transplantation
o	non-invasive ventilation (masks etc.)
o	physiotherapy (5-6 times weekly, training)
322
Q

what is the median survival of IPF? what was the most common treatment for it?

A

o Has a median survival of 2-5 years from diagnosis
o Previously hypothesised that IPF was a disease of inflammation, leading to scarring
o Thus, immunosuppression was the mainstay of treatment
Up until 5 years ago, the main treatment was adrenous treatment till 2012 when there was a landmark journal which changed the way we treat it

323
Q

wha was the technique to treat IPF previously and why was it stopped?

A

PANTHER study (previously used)
• Includes randomised trial
• ½ patients with combination therapy which included steroids + immunosuppressant’s (azathioprine) + NAC (antioxidant)
• up until 2012m this was the standard treatment
• it was stopped as they found that patients who had this treatment (8) died within this study. Compared to 1 who died from the placebo group
• deaths from any cause or hospitalisation was significantly lower in the placebo group compared to the treatment

324
Q

what are the newer treatment options for IPF called?

A
treatment working on the fibrotic pathways, rather than immune suppression were underway
o	Pirfenidone
♣	approved by NICE in the UK in 2014
o	Nintedanib
♣	Approved by NICE in the UK in 2016
325
Q

how does pirfenidone work? including its side effects and how often the tablets should be taken

A

• It has an effect on proliferation of fibroblasts
• Has an effect on tgfb and ECM protein synthesis (also migration, CTGF, proliferation, myofibroblast trans-differentiation and apoptosis)
• Three tablets 3 tomes a day
• Main side effects: (not everyone)
o Photosensitivity (skin rash) – need to wear spf50 whenever leaving the house
o Gastrointestinal upset (nausea, weight loss, reduced appetite)
o Liver function test abnormalities (hepatitis)

326
Q

what are the names of the 2 studies that showed evidence for a positive effect of pirfenidone?

A

the capacity study and ascend study

327
Q

describe the 2 studies showing evidence for the positive effect of pirfenidone

A

1 – Pirfenidone – CAPACTITY study
• Reduced the decline in FVC over 72 weeks
• Fewer patients had FVC decline of >10% compared to placebo
• Study had 3 groups: 2 with differing doses of Pirfenidone and a placebo group
• The group with the higher dose had a less of a decline in FVC, both were better than the placebo
2 – Pirfenidone – ASCEND study
• Reduced the decline in FVC over 52 weeks
• Fewer patients had FVC decline of >10% or death
• Decline in FVC is a surrogate marker for mortality
o If you want the study to show improvement of mortality, the study would need to be followed up for more than a year
o Use decline in FVC as a marker for mortality

328
Q

what is nintendanib? including its main side effects and how many tablets are taken a day

A

• Different mechanism of action to pirfenidone
• Had already been licenced for the treatment of cancer
• One tablet twice a day
• Main side effect: diarrhoea (can be quite severe)
o Could reduce the dose and stop the diarrhoea and prescribe something to stop this
• Can cause LFT abnormities (hepatitis)
• Small increased risk of bleeding

329
Q

is a combo f nintendanib and pirfenidone especially damaging to the cell?

A

no - a recent study showed ocombo of Pirfenidone and Nintendanib = safe

330
Q

how does nintendanib work? (what does it affect)

A

• It is an inhibitor of multiple tyrosine kinase receptors
o Platelet derived growth factor
o Fibroblast growth factor
o Vascular endothelial growth factor

331
Q

describe the study that shows evidence for the positive effect of nintendanib

A

1 – Nintedanib - IMPULSES study
• Reduced decline in FVC over 52 weeks
• Fewer patients had FVC decline of >10%

332
Q

what is better, nintendanib or pirfenidone in treating ILDs?

A

Pirfenidone vs Nintedanib

  • No head:to:head comparison studies
  • A network meta-analysis concluded that pirfenidone and nintedanib provide similar benefits
  • Pooled analysis demonstrated that both treatments significantly reduced (by ~50%) all-cause mortality vs placebo
333
Q

what are recent drug trials especially targeting?

A

• Lots of studies going on in the field regarding drugs to treat IPF including ones that target fibroblasts and type2 cells etc.

334
Q

what is the available treatment for hypersensitivity pneumonitis?

A

• Main treatment is identification and exposure avoidance
o find out what patient is sensitive to and then getting rid of this?
o i.e. if it is due to their pillow, get rid of it, if its due to a pet, get rid of it, if its due to their job, may need to leave etc.
• cause unidentified in up to 60% of cases
• prognosis better in those who avoid further exposure
• progression to fibrosis may still occur in those who have removed antigen exposure
• treatment strategies focussed on inhibition of adaptive immune system

335
Q

describe the corticosteroids as a treatment for HP - a study that has been done to prove its effect

A

• corticosteroids commonly used as first line treatment
• only 1 randomised, placebo controlled study in 1992
o 20 patients received prednisolone for 8 weeks, 16 received placebo
o after 1 month, significant difference in DLCO between the treatment groups. O statistical significant difference in FVC
♣ DLCO – diffusing capacity of the lungs for carbon monoxide
♣ Determines how much oxygen travels form the alveoli to the lungs in the blood stream
o after a follow-up of 5 year no statistical significant differences were found between the treatment groups in FVC, FEV1 or DLCO

336
Q

what are the benefits and side effects of corticosteroids?

A
  • Derived from naturally occurring steroids produced by the adrenal glands
  • Decrease function of lymphocytes and cytokine activity
  • However, several undesirable consequences from long-term use…
Side effects:
•	Obesity
•	Indigestion
•	Increase risk of infectionspsychosis
•	Osteoporosis (increased risk)
•	Diabetes (increased risk)
337
Q

why is a personalised medicine approach beneficial for treating HP? what has been devised so far?

A
A personalised approach is required to ensure efficacy, compliance and tolerability
•	Use of lowest efficacious dose
•	Use of ‘steroid sparing agents’
•	Access to specialist nurse liaison
•	Monitoring for complications
o	Bone scanning (DEXA)
o	Glucose monitoring
338
Q

what are some names of immunosuppressants that have been used to treat ILDs

A
•	Methotrexate (MTX)
•	Azathioprine (AZA)
•	Mycophenylate mofetil (MMF)
•	Cyclophosphamide (CYC)
•	Rituximab (RTX)
Other agents may allow a reduction in steroid dose to mitigate long-term effects in some forms of ILD
339
Q

how does methotrexate work to treat ILDs?

A

• Inhibits folate metabolism
• Reduces T-cell ability to make proteins
• Paradoxically can cause pneumonitis
o pneumonitis – inflammation of the lungs
o hard to know if deteriorating state of person due to ILD or methotrexate (the disease of medicine)

340
Q

how does Azathioprine work to treat ILDs?

A
¥	Prodrug – converted to 6-mercaptopurine
¥	Inhibits purine synthesis
¥	Reduces T-cell turnover
¥	Can cause hepatitis
¥	Bone marrow suppression may occur
341
Q

how does Mycophenylate work to treat ILDs?

A
  • Inhibits purine synthesis
  • Reduces T-cell turnover
  • Immunosuppressive
  • Susceptible to viral infection
342
Q

how does Rituximab work to treat ILDs?

A
  • Depletion of CD20-positive B lymphocytes, thereby inhibiting their differentiation into antibody-producing cells
  • Inhibition of T cell co-stimulation
343
Q
what was found from a retrospective study of •	Mycophenylate mofetil (MMF)
and  Azathioprine (AZA) in terms of what was better tolrated?
A

¥ Retrospective study
¥ 51 were treated with MMF and 19 with AZA
¥ DLCO increased by 3.9% in MMF group and by 5.1% in AZA group
¥ no statistically significant difference
¥ MMF = better tolerated (less drop-out)

344
Q

what was found in a study testing the effectiveness of Pirfenidone in HP?

A
  • Retrospective study in Japan
  • 23 patients with chronic HP
  • Statistically significant improvement in decline in vital capacity (VC) over 6 months
345
Q

what was found in a study of MMF in CTD-ILD?

A

• A study in 2013 showed that there was an:
o improvement in FVC which was statistically significant
o improvement of TF – statistically significant
o MMF was associated with significant improvements in estimated percentage of predicted FVC% and in estimated DLCO%

346
Q

what was found in a study of rituximab in CTD-ILD?

A

A study in 2013 showed that there was a statistically significant improvement in FVC and DLCO in 33 CTD-ILD patients

347
Q

how does SSc-ILD work on DNA?

A

majority of drugs act on lymphocytes – affecting DNA synthesis

348
Q

describe studies that talk about CYC in SSc-ILD (1 and 2)

A

CYC in SSc-ILD (SLS-I)
• Mean absolute difference in adjusted 12-month FVC % predicted between the cyclophosphamide (n=79) and placebo (n=79) groups was 2.53%, favouring cyclophosphamide. This was statistically significant
• So, provides evidence that CYC has improvement compared to placebo but only 5%

CYC/MMF in SSc-ILD (SLS-II)
¥ Showed no significant difference in FVC % change between treatments (p=0·24)
¥ More patients on CYC than on MMF prematurely withdrew from study drug (32 vs 20)
¥ both equally effective but more side effects with CYC
¥ Sixteen deaths occurred (11 CYC; 5 MMF) with most due to progressive ILD

349
Q

what is a treatment for sarcoidosis?

A

¥ Not all patients with pulmonary sarcoidosis require treatment
¥ Treatment is favoured in following patients
¬ Symptomatic (breathlessness, cough fatigue)
¬ Active inflammation & progressive organ damage
¬ Worsening pulmonary function
- An important part of management involves identifying the patients that require treatment and those who can be observed without treatment

350
Q

how are Corticosteroids used in sarcoidosis

A

¥ Corticosteroids are the first-line therapy
¬ Initial doses of prednisone 20-40 mg daily
¬ No studies to define the optimal dose or duration
¬ Dose is typically tapered every 3–4 weeks to ≤10 mg
¬ Typically, patients are treated for at least 6 months
¬ Chances of relapse after therapy stopped 14-74%
¥ high chance of patients relapsing after they stop steroids
¥ Those patients who can be controlled with <10 mg/day of prednisone are usually not considered candidates for treatment with steroid-sparing cytotoxic medications.
¥ In those patients who relapsed while off steroids, reintroduction of steroids usually stabilized their disease

351
Q

how is methotrexate used in sarcoidosis?

A

¥ MTX used as second-line treatment in following patients
¬ Steroid-refractory
¬ Develop steroid-associated adverse effects
¬ As a steroid-sparing agent
• Typically begun orally at a dose of 5 mg weekly and gradually increased by 2.5 mg every 2 weeks up to 10–15 mg per week
- if steroids don’t work or if they’re having bad side effects, MTX could be given at the same time as steroids
- Benefits of MTX therapy require at least 6 months to manifests themselves. Hence tapering of steroids should be delayed for at least several months after starting MTX

352
Q

describe MTX vs AZA as treatments in sarcoidosis

A

Study testing MTX vs AZA a second line therapy for sarcoidosis:
• Both have significant steroid-sparing potency
• Similar positive effect on lung function
• Higher infection rate in the azathioprine group

353
Q

describe how Leflunomide is used in sarcoidosis?

A
  • Mean change in FVC in the 6 months prior to leflunomide was 0.1L and +0.09L in the following six months (p=0.01)
  • The median corticosteroid dose at initiation was 10 mg at baseline and 0 mg at 6-month follow-up (p<0.001)
354
Q

how is Infliximab used in sarcoidosis?

A

¥ Infliximab therapy resulted in a statistically significant improvement in % predicted FVC at Week 24
¥ To be used as a third-line agent in severe refractory sarcoidosis cases not responding to conventional immunosuppressive treatment
¥ Adalimumab is an effective alternative for patients intolerant to infliximab

355
Q

how is pulmonary rehabilitation used treat IPF?

A

¥ Exercise training significantly increased 6MWD and health related quality of life
¥ Larger improvements in IPF compared to CTD-ILD
¥ Benefits declined at 6 months in IPF but not CTD-ILD
¥ patients go 1-2 times a week to an exercise class

356
Q

what is the survival post-transplant IPF vs HP like?

A

HP may recur in the allograft and negatively impact clinical outcomes. Thus, vigilance for ongoing antigen exposure and disease recurrence after LT for HP is important. The overall survival following LT in this group is good and appears to be improved compared with IPF, the most common ILD indication for LT.

357
Q

what are Palliative Treatments and what do they involve when treating IPF?

A

Treatment targeted at mitigating symptoms
May involve:
¬ Opioids for breathlessness and pain
¬ Anxiolytics for anxiety (e.g. benzodiazepines)
¬ Supplementary oxygen
¬ Commonly, patients will experience symptoms from ILD, which do not resolve with treatment of disease

358
Q

what do long term oxygen therapies involve?

A

• Palliative or ‘long term’ oxygen therapy (LTOT)
o >15hrs per day
• Hypoxia at rest
o pO2 <7.3kPa or <8kPa if evidence of PH
• Often delivered by static air concentrator

359
Q

what are the benefits of Ambulatory oxygen

A
  • Enables greater exertion to be undertaken in patients with hypoxia on exercise
  • Historically compressed oxygen in tanks, but portable concentrators increasingly common
  • Ambulatory oxygen studies show improvements in different exercise parameters
360
Q

what are the differences in innate and adaptive immune defences?

A
Innate Immune defenses
Mucosal defenses
Mucociliary, anti-bacterial proteins
Innate cellular defenses
Macrophages and neutrophils
Adaptive immune defenses
B-cell mediated
Antibodies
T-cell mediated
Cytotoxic and helper cells
361
Q

are acquired or inherited defects more common and more dangerous?

A

acquired = more common but less dangerous

362
Q

what are the questions to be asked when figuring out if a patient with immunocompromised lungs needs treatment?

A

What is the cause and nature of the defect?
Can the underlying cause be corrected or improved by a specific treatment?
If not, what are the likely consequences of the defect (what type of infections will the patient get? How severe?)
Can we prevent infections by prophylaxis (low dose preventative anti-infective agents)
If an infection is present, identify the causative organism and treat or make a ‘best-guess’ or treat multiple potential pathogens

363
Q

what is an example of specific treatments that can be used to teat HIV, CF etc?

A

smoking -get rd of it and give nictonie replacement
Cancers and SCID, GCD - do stem cell transplantation or gene therapy for severe immune deficiency
CF - correction of CFTR
HIV - HAART
AB deficiency - immunoglobulin replacement

364
Q

what are the treatment differences for the 2 different mutations of CFTR?

A

G551D mutations affects 4% of the CF population and causes lack of functioning CFTR (doesnt open) so treat with potentiators (eg ivacaftor) to open it
F508del mutation causes misfolding of CFTR so doesnt go to cell membrane and s not stable. need to correc folding so use Correcters e.g. tezacaftor and lumacaftor. need to use with potentiator ivacaftor to see any effect

365
Q

what was found in a study showing the effect of using bot potentiators and correctors to treat F508del mutation?

A

found that there was a sustained well functioning of the channel so was successful (much higher than in placebo)

366
Q

what receptors do HIV especially effect?

A

CD$ receptors and co-receptors (CXCL4 or CCR5)- t helper cells, macrophages and dendritic cells

367
Q

what cell type do HIV affect in its progression?

A

use cd4 receptors to infect cells and use T helper cells machinery to make viral proteins and proteases to cleave the,

368
Q

describe the mechanism of viral replication

A

Glycoproteins on the HIV molecule (gP160 made of gP120 and gP 41) allow it to dock and fuse onto the CD4 and CCR5 receptors
The viral capsid the enters the cell and enzymes and nucleic acid are released
Using reverse transcriptase single stranded RNA is converted into double stranded DNA
Viral DNA then is integrated into the cells own DNA by integrase enzyme
When the infected cell divides the viral DNA is read and long chains of viral proteins are made
Assembly the viral protein chains are cleaved and reassembled
Budding here immature virus pushes out of the cell taking with it some cell membrane
Immature virus breaks free to undergo more maturation
Maturation protein chains in the new viral particle are cut by the protease enzyme into individual proteins that combine to form a working virus

369
Q

how do ARVs (HIV treatment) work?

A

ARVs work by interfering with the way that HIV virus replicates.
They do this by interfering with the docking of HIV and interfering with the action of major HIV enzymes
Reverse transcriptase
Integrase
Protease

370
Q

what are the inhibitors that HAART use for treatment of HIV?

A
-NNRTI
Non Nucleoside reverse transcriptase inhibitor
EFAVIRENZ
NEVIRAPINE
RILPIVIRINE
-NRTI
Nucleos(t)ide reverse transcriptase inhibitor
3TC LAMIVUDINE
FTC EMTRICITABINE
AZT ZIDOVUDINE
TDF TENOFOVIR
ABC ABACAVIR
-Integrase Inhibitor
DOLUTEGRAVIR
RALTEGRAVIR
ELVITEGRAVIR
-Protease Inhibitor
DARUNAVIR
ATAZANAVIR
LOPINAVIR
-- use combo of all 3
371
Q

how can you tell when you should start treatment of HIV using HAART?

A

Indications to commence therapy in HIV-positive patients include a CD4 count<350, any AIDS defining illness (eg pneumocystis pneumonia PCP, Kaposis sarcoma), neurological involvement, or to reduce the risk of transmission

372
Q

what are possible porblems with using HAART? what is being done to combat this?

A

many tablets for life
Very complex for patients (and doctors)
Missed doses encourage treatment failure and resistance
Many side effects and interactions
Combination tablets have been made by (unprecedented) co-operation between pharma companies

3 diff drugs several times a day = complicated for a chronic illness
missed doses causes resistance
some pharmaceutical companies – mixed the drugs together which can be taken once a day

373
Q

what is good compliance associated wth for HIV treatments?

A

Good compliance is associated with better suppression of viral replication/load, improved clinical response and reduced risk of viral resistance and transmission

374
Q

what is b cell defects and antibody deficiency associated with? what is there an increased risk of?

A

Anti-CD20 monoclonal antibody therapy (for haematalogical cancer, autoimmune disease)
CVID

Increased risk of:
Encapsulated bacteria
(H. influenzae and S. pneumoniae)
Oto-sino-pulmonary infection, bronchiectasis

375
Q

describe the effectiveness of using immunoglobulins as replacements in antibody deficiency and how it is prepared

A

In primary antibody deficiency (CVID or common variable immune deficiency) immunoglobulin replacement has dramatically increased survival (from 12 to over 50 years). Reduces infections including pneumonia
Prepared from pooled plasma from healthy donors (50), extensive screening for blood-bourne disease. A small risk remains
Can be administered by intravenous (usually every 3 weeks) or subcutaneous (usually weekly) lifelong

376
Q

when do you give infection prophyalxis?

A

The nature and severity of underlying condition and risk of infection
Previous infection history
The evidence that prophylaxis has benefit in this situation
Available agents, route and frequency, side effects
Treatment burden
Antimicrobial stewardship

377
Q

give 3 examples of respiratory prophylaxis in immunocompromised patients

A
Antibiotic prophylaxis in myelotoxic chemotherapy when neutropenia can be predicted both in time and severity
Antibiotic and antifungal prophylaxis in chronically neutropenic patients eg bone marrow transplant, or those with inherited defects 
Pneumocystis pneumonia (PCP) prophylaxis in HIV and immunosuppression (Thelper cells compromised)
378
Q

What is Pneumocystis Pneumonia (PCP)? and its cause

A

Life-threatening opportunistic lung infection of immunocompromised individuals
Caused by Pneumocystis Jirovecii, a fungus with some protozoal features.

379
Q

what is the conditions for considering a patient for prophylaxis in PCP?

A

Occurs in patients with reduced Th cell number or function. Consider for prophylaxis if:
HIV, CD4 <200 or previous PCP
Chronic high dose steroids +/- immunosuppressives (that affect the t cell)
Solid organ or allogeneic bone marrow transplantation

380
Q

describe the 2 different prophylaxis’s used for PCP patients

A

septrin - 960mg oral three times a week or 480mg daily, Both components inhibit folate biosynthesis
Rash (can be life-threatening), bone marrow suppression, gastointestinal problems, caution is had Previous severe reaction, neutropenia, 1st trimester pregnancy, methotrexate, renal and liver failure.
Extremely effective

Nebulised pentamidine
300mg nebulised monthly
Uncertain
Anti-parasitic
Cough
Bronchospasm
Need to protect staff
Previous reaction, severe asthma
Effective but some failures
381
Q

how would a patient with pulmonary infection (immunocompromise) present and how is it diagnosed?

A

May be non-specific – fever, lethargy or with respiratory symptoms (cough, breathlessness)
Chest Xray or CT (computed tomography) changes may give clues
Investigate with sputum samples if possible
If no diagnosis or no response to initial treatments you may need to take samples from the lung (e.g. bronchoscopy)

382
Q

what are the 2 tests that can be done to get a lung sample?

A

Sputum induction - -patient inhales something thats an irritant to help them
and
fibreoptic bronchoscopy - fibreoptic telescope – inside the lung

  • so can obtain samples from the lungs to try and grow the pathogen or you could detect the footprint using PCR
383
Q

how can you choose a good treatment of PCP?

A

High dose co-trimoxazole (120 mg/kg daily, in 2–4 divided doses for 14–21 days) oral or intravenous
Adjunctive corticosteroid therapy for moderate-to-severe PCP enhances survival in HIV. ?reduces inflammation to dying organisms
Toxicity– fluid load, rash, renal/liver toxicity, low blood potassium, neutropenia, low blood glucose
Alternatives include intravenous pentamidine (not nebulised), and intravenous clindamycin-primaquine. Very toxic as well.

384
Q

what is invasive aspergillosis?

A

Aspergillus fumigatus – environmental fungus
Affects neutropenic or immunosuppressed patients
Can be progressive and fatal
Pulmonary nodules or infiltrates on CT – a ground glass ‘halo’ is classic
Treat if classic, or undertake bronchoscopy +/- biopsy – culture, fungal cell wall components (B-glucan, galactomannan), histology

385
Q

what are treatments of invasive aspergillosis?

A

Reduce immunosuppression?
Voriconazole (a triazole) is first-line treatment
Oral or intravenous,
Loading dose required (large volume of distribution), metabolised in liver
Side effects – visual disturbance, cardiac rhythm disturbance (QT prolongation), hepatotoxic, rash, convulsions
Response rates of 50-90%
Fungal resistance emerging
Newer agents eg posaconazole and isavuconazole

386
Q

what are alternative treatments for invasive aspergillosis other than voriconazole?

A

Echinocandins (caspofungin, micafungin,anidulofungin intravenous only
Well tolerated, GI disturbance, heptotoxicity, allergic reactions
Amphotericin B – iv only but liposomal formulation to reduce toxicity; intravenous (can be nebulised for prophylaxis)
Nephrotoxic, hypokalemia, chills and allergic reactions
Both less effective monotherapy than voriconazole, echinocandins have been used with voriconazole

387
Q

what are some respiratory conditions that are due to a change in genetics?

A

• asthma, COPD, lung cancer, alpha-1 antitrypsin deficiency, cystic fibrosis, many more

388
Q

how many people in the UK are affected by CF and what is the median age of death?

A
  • UK 10,461 people affected

* Median age death 31 years old

389
Q

what is the defect in chromosomes that causes CF?

A
  • Defect in long arm of chromosome 7 coding for the cystic fibrosis transmembrane regulator (CFTR) protein
  • > 1600 mutations of CFTR gene identified
  • F508del most common mutation causing CF (49% homozygous)
  • 90% within panel of 70 mutations
390
Q

what is the prevalence of CF incidence? why is it like this?

A

• Static incidence with an increasing prevalence
o Could be because people are living longer now with the personalised medicine
o Predicted that children in 50s will live up till they’re in their 50s

391
Q

describe the pathophysiology of CF

A

Failure of mucocilary clearance and impaired immune function contribute to continues insult to bronchial wall, through the recruitment inflammatory cells and uncontrolled neutrophilic inflammation
More infections, more damage to lungs which just becomes a vicious cycle

392
Q

what kind of lng cndition is CF?

A

– it’s an obstructive lung condition

due to it having lots of inflammation, smooth muscle becomes thicker and respiratory lumen gets narrower

393
Q

how can CF be diagnosed in children? what are some clinical symptoms of it? when are people usually diagnosed by?

A

Young children – heel prick foot test – used to pick up a range of conditions including cf
• genetic profile – look for number of genes you can identify but some mutations you can’t
• clinical symptoms (most identified like this) – frequent infections, malabsorption, failure to thrive
• stool block (small bowel obstruction), thick secretion in bowels
• people with recurrent pancreatitis also could be thought to have this
• late diagnoses via (chesty child and have asthma, not get better then is screened for CF)
• mild CF picked up later in life
• males with CF, majority of them are infertile as they have an absence of their vas deferens
• abnormal salt/chloride exchange – raised skin salt or impaired nasal potential difference (sweat test).
o Measure content of sweat
o People with CF have abnormal sweat content
• 50% diagnosed by 6 months, 90% diagnosed by 8 years of age

394
Q

describe all the complications that arise rom having CF and how many people are usually affected?

A
  • bronchiectasis (a whole host of conditions) – many things can cause this including CF, airways become dilated and distorted for some reason
  • 90% of people who survive the neonatal period of CF get bronchiectasis
  • infertility: 98% male and 20% in females (obstruction to sperm getting through which makes them seem infertile)
  • pancreatic insufficiency malabsorption 85% (airways and tracts in body have thick secretion; in pancreas, ducts and tubes get blocked over time and pancreas is meant to make pancreatic enzymes and also insulin so this doesn’t work well)
  • CF related diabetes 50% by age 30 (due to the lack of insulin production caused by the pancreas)
  • CF liver Disease 25% chronic liver disease, 10% cirrhosis (liver filled with ducts and tracts that get blocked – many get liver transplant before lung transplant)
  • Sinonasal polyps 10-25%
  • Distal intestinal obstruction syndrome (DIOS) 20% (people with pancreatic insufficiency are more prone to this) – secretions in bowel = thick due to stool in bowel (affects small bowels so need lots of laxatives etc.)
  • Arthropathy 10% adults (problem with their joints)
  • Meconium ileus 10-20% infants
  • Low bone mineral density (osteopenia/osteoporosis) 10-25%
  • Peneumothorax 4-20% (lack of lung markings so can more easily get collapse of lug/puncture)
  • Allergic lung disease (ABPA) 7%
  • Skin – salty sweat/salt loss
395
Q

what stage of CF management is spent te most time on? what does this step entail?

A
•	Rescue antibiotics
o	2-week course
o	home vs hospital
o	access – peripheral/central
o	issues frequent antibiotics
-	allergies
-	renal impairment
-	resistance
-	access problems
as patients live longer, worries that they will see more of these problems
396
Q

what stage of CF management is being worked on by scientists the most? what does it entail?

A

Use maintenance preventions (trying to work at this takes most time)
• CF prevention management
o Segregation
o Surveillance –frequent review minimum every 3 months
o Eradicate infections early
o Suppression of chronic infections – antibiotics
o Anti-inflammatory – azithromyocin, steroids
o Airway clearance physiotherapy, bronchodilation and exercise
o Nutrition – pancreatic enzymes, diet high calorie and fat, supplements including vitamins, percutaneous feeding
o Diabetes – insulin
o Psychosocial support
o Vaccinations – influenza, pneumococcal
o (lung transplant)

397
Q

how can the holes of CF be fixed by personalised medicines? what can happen with and without these personalised medicines?

A

• Personalised medicine
o individual tailored or targeted medicine
o move away from a ‘one size fits all’ approach
o stratified based on predicted response or risk of disease
o genetic information is a major factor

Without personalised medicine, some people benefit from the drugs and some don’t (some people can even get adverse effects)
With personalised medicine: each patient received the right medicine and so they each benefit from this individualised treatment (using biomarkers and diagnostics to discover the right medicines)

398
Q

what are the advantages of personalised medicines?

A
  • increase adherence to treatment
  • improve quality of life
  • increase life expectancy/prolong life
  • help avoid adverse drug reactions/intolerances
  • shift the emphasis in medicine from recue to prevention
  • help control the overall cost of health care
  • direct the selection of optimal therapy and reduce trial-and-error prescribing
  • reveal additional or alternative uses for medicines and drug candidates
399
Q

why are personalised medicines used in and are beneficial in CF?

A
  • monogenic disorder (i.e. is the result of mutations(s) in a specific gene) [know that it is chromosome 7 where it happens]
  • less known influence on environmental factors on disease expression
  • well-characterised pathophysiology with clear therapeutic targets
400
Q

what are examples of monogenic and complex disorders? give examples of both

A

monogenetic vs complex disorders

  • complex disorders e.g. asthma have many environmental factors that can influence their disease
  • monogenetic disorders is mostly a genetic condition with the mutation occurring in 1 gene and has an either dominant or recessive inheritance pattern
401
Q

what can be done by using perosnlaised medicine in CF?

A
  • genotype directed therapies
  • treatment adherence support – tailored behaviour change interventions
  • targeted treatments based on infectious organisms and resistance patterns
402
Q

explain some genotype-derived therapies used to treat CF?

A

• small-molecule agents facilitate defective CFTR processing or function
o Ivacaftor G551D (6%) and other specific mutations
- improved lung function (FEV1), BMI, QoL
o Orkambi homozygous F508del – only licensed compassionate use UK (beefits don’t outweigh costs so can’t give this to everyone)
- Mixed outcomes

• Gene therapy – further research as significant problems with delivery

403
Q

what are the 5 functional classes that can be what causes CF to occur?

A
  • Don’t make any CFTR protein
  • Produce it, but can’t move up to membrane to work
  • Produce it, its trafficked but doesn’t open
  • It is produced and moves up but it is abnormal so disruption ion flow
  • Produce CFTR but in smaller quantities
404
Q

which of the 5 functional classes that causes CF do the G551D and F508del mutations come under?

A

Delta F508 – Class 2

Gff1D – Class 3 (ivacaftor is useful here) – 5%

405
Q

what is the effect of using ivacaftor and lumacaftor together to treat CF? who can receive it? (what age?)

A

Ivacaftor and lumacaftor (Orkambi) (have 2 diff dysfunctions) - combo
• Ivacaftor and lumacaftor combined
• Lumacaftor is a CFTR corrected – corrects cellular misprocessing of CFTR (e.g. folding) to facilitate transport from the endoplasmic reticulum
• Improves stabilisation and processing (folding-trafficking) of CFTR to increase its presence on the cell surface
• Class 2 mutation – F508 del/F508edel
• Age 6 and above

406
Q

list the possible challenges that could come with treating CF

A
  • Adherence to treatment
  • High treatment burden
  • High cost of certain treatments
  • Genotype determining pharmacogenomics treatments – not available to all
  • Allergies/intolerances to treatment
  • Different infectious organisms and their resistance to drugs
  • Equity/restrictions to prescribe treatments i.e. NHS England restrictions on Orkambi meaning currently only available via compassionate use program
  • Prevention vs recue treatment – disadvantages of recue – cost, time, decline in quality of life and life expectancy, side effects of recurrent intravenous antibiotic therapy
407
Q

describe the problem with adherence to treatment in CF

A
  • Adherence to medication among people with long term conditions is estimated at 30-50%
  • CF adherence to nebulisers 36%
  • Adherence often invisible –need objective data
  • Adherence complex
408
Q

what are the principle airway diseases?

A

asthma and COPD

409
Q

what are the diferent types of asthma?

A

eosinophilic or non-eosinpphilic asthma
eosinophilic has 2 subtypes
atopic = a tendency (not diagnosis) – to form iGe antibodies to common allergens (cats/pollens etc). not all with atopic get the illeess associated eg astma hayfever etc. its very common and typical for childhood asthma. lab workers could also become allergic – become allergic
non-atopic – very common. have allergy bt dont know whta theyre allergic to

410
Q

describe what happens in eosinophilic asthma

A

eosinophols are rectuied to the airway in this type of asthma
they cause airways to be destroyed>
very allergic tye of asthma – to dust, pollen etc
have igE cells on surface of mast cells which releases cytokines and allergic t cells release other things

411
Q

describe non-eosinophilic asthma

A

non inflammatory control
dont know how many sub types of thsu tehre are
some people with is have smoking related disease, people that are pverweight can have their fat cells causing them to be astmatic

412
Q

what are the 2 man things that asthma drugs need to do?

A
Reduce airways inflammation, mucus  production and bronchial hyperreactivity
Ameliorate symptoms (bronchodilatation)
413
Q

describe the COPD phenotypes

A

Persistent symptoms, hyperinflation, emphysema
Frequent exacerbators (≥2 exacerbations per year) – go to hospital for intravenous antibodies etc.
Division based on eosinophilic or non- eosinophilic inflammation?
domt know much about sub types of COPD
think COPD probably has something to do with levels of eosinophils but dont know

414
Q

what is the name of the condition of asthma and COPD together? what kind of person would have it?

A

ACOS (Asthma and COPD Overlap Syndrome)

if you have asthma at young age then start smoking, at 50 years old could have both.

415
Q

why are inhaled therapies useful?

A

Targeting - get drug to where the disease it – if you dont want to go to the wole body, just to an organ – inhalation =better
Minimising adverse effects - adverse effects minimisation – giving frugs just to surface of lungs so no adverse effects of the drug everywhere else

416
Q

what is an important factor in regulating where drugs go?

A

Particle size is one factor that governs deposition.
1-10 µm size generally held to be in the respirable range. – bigger than this, deposits in oralpharynx. smaller, will never interact with the airway wall
Depends on device delivering, nature of drug (e.g. hydrophobicity) and flow rates, any underlying disease and regional differences in lung ventilation (if not well ventiated art of lung if you have a disease, dont have an effect

417
Q

how are smooth muscles affected in asthma/COPD

A

Inappropriate and excessive contraction of smooth muscle

Hypertrophy and proliferation of smooth muscle cells, further narrowing the airway lumen

418
Q

what do short acting bronchodilators have in them?

what are B2A used for

A

Short acting bronchodilators open up constricted smooth muscle in both asthma and COPD
‘blue’ inhalers: salbutamol and terbutaline
Nebulisers for high dose therapy – inhaled through mask
Beta 2 agonists to reduce cardiac side effects (persuade smooth muscle to relax)

419
Q

give examples of different b-adrenoreceptos and the different ways that they work

A

Adenaline - rescue from anyphylaxis
Isoprenaline - does bad things to the heart - acute rescue use
Salbutamol - works very quickly longer lasting
Salmeterol - have some versions that are longer acting – hang around in membrane

420
Q

how does noradraline activate b-adrenoreceptors?

A

bind to gcpr, activate adenylate cyclase, to cAMP, pka topka*

421
Q

what are some adverse effects of cAMP activation

A

Raising cAMP may activate Na/K exchange pump driving cellular influx of potassium
Tachycardia (cardiac side effects)
Hyperglycaemia: loss of insulin sensitivity, increased liver glucose release

422
Q

what are LABAs and when are they used?

A

Long-acting beta agonists.
Valuable bronchodilators with possible anti- inflammatory actions
Never used without steroids in asthma
Often used without steroids in COPD

423
Q

what are SABAs? can they be used alone?

A

saba = short acing beta agonist – effects wear off

saba without steroid treats sypmtoms but not underlying inflammation
very powerful to reduce bronchocostriction and improve quality of life

424
Q

how do long acting anti-muscirinics work?

A

The parasympathetic nervous system regulates airways tone
Blocking the action of acetylcholine on the muscarinic receptors, leads to bronchodilatation and reduce mucus secretion
Long acting anti-muscarinics (LAMAs) such as tiotropium, glycopyrronium, aclidinium are effective bronchodilators

425
Q

how does stopping the parasympathetic nervous system affect airways?

A

if you block action of parasymp nervous system
less Ach so no bronchoconstriction
-airways the relax

426
Q

what is a therapeutic option for treating COPD without exacerbations?

A

LABA/LAMA with SABA as needed

SABAs as a top-up

427
Q

what is the use of steroids in treating asthma and COPD?

A

Steroids treat inflammation.They are therefore the mainstay of asthma therapy and prevent asthma deaths
Treat the processes that drive remodelling, airway smooth muscle proliferation, epithelial injury
Inhibit inflammation driven by NFkB and related pathways, promote epithelial integrity

428
Q

why arent oral steroids given to everyone?

A

because there are side effects
Systemic: diabetes, cataracts, osteoporosis, hypertension, skin thinning, easy bruising, growth retardation, osteonecrosis of the femoral head
Topical: hoarse voice, oral candida, skin thinning, easy bruising, cataracts (in high dose)
Adrenal suppression

429
Q

do systemic or topical steroids have more side effects?

A

topical have less side effects because its localised to the place you wan tit to go

430
Q

what are the benefits of inhaled corticosteroids?

A

Minimise systemic absorption
Reduce side effects to mostly local adverse events (but still include oral thrush, dysphonia, easy bruising, cataract)
Range of inhaled steroids of varying potency: beclometasone, budesonide, fluticasone diproprionate, fluticasone furoate

431
Q

what are some devices to allow inhalation of corticosteroids?

A

MDI – CFC free. drug suspended in propellant, breath at same time. activates drug, puff and shoots jet and breath into lungs. drug goes to where it needs to go in the lungs
not easy to take – difficult
flow rate dependent
must be timed directly to inhaling and puffing

DPI – dry powder devices – drug suspended in carrier devices e.g. lactose. suck in powder which is aerosolised

432
Q

what device is used to get over the problem of depositing corticosteroids in mouth? what can happen if not used?

A

used to get round problem of getting puffer into spacer, instead of bit that would have deposited into mouth, gets deposited in the spacer so decrease risk of oralpharyngeal X

433
Q

how is ICS used in asthma?

A

Often a combination therapy

ICS/LABA combinations are common and effective: seretide, fostair, symbicort, relvar

434
Q

how is ICS used in COPD?

A

More controversial
Reduces frequency of exacerbations (‘triple’ therapy of ICS/LABA/LAMA)
May increase risk of pneumonia
May be of particular benefit in people with asthma and COPD, or with eosinophilic COPD

435
Q

what are some immediate management options for acute asthma?

A

Oxygen 40-60% (CO2 retention not usually a problem)
Salbutamol neb 5 mg (+ipratropium neb 0.5mg if life threatening) - repeated/ i.v. infusion (much higher doses than what is in inhalers)
Prednisolone 30-60 mg (±hydrocortisone 200mg iv)
Magnesium or aminophylline i.v. (bolus/load)
ABGs
CXR if suspect pneumothorax, consolidation, or fails to respond to treatment (or is very severe)

436
Q

what are some less common/popular inhaled treatments?

A

Chromoglycate – usually in children
Nebulised antibiotics – in people with infections?
Nebulised steroids

437
Q

what are some less common/popular oral treatments?

A

Leukotriene receptor antagonists

Theophyllines – tell muscle to relax

438
Q

what is atopy?

A

atopy = tendency to form IgE to allergens?
atopic = tendency rather than an illness
can be atopic but not have illness
1 in 5 = atopic. half of these have an illness

439
Q

what are the tests for allergy/asthma?

A

wheel and flare response:
wheel = swelling around siite
slare = redness- bloody

skin prick test:
prick is so light that it doesn’t draw blood

440
Q

what is a drug that was given for the treatment of kidney transplant that is also effective in treating asthma?

A

cyclosporin to stop rejection (suppresses t cell) in kidney transplants
cyclosporin = used early on in clinical trials
found that it can cause fewer exacerbations, less need for steroids and inhibits T cell proliferation via cyclophilin and calcineurin

441
Q

what happens in response to IgE cells and mast cells in an allergic reaction?

A

Fcer1 binds IgE which coats mast cells and basophils that are in circulation
allergen comes along and cross links 2 receptors of Fcer1 and puts them together which causes a signal
mast cells then recruit mediators e.g. histamine which is what causes the redness in flare
this is very quickly followed by a range of cytokines, proteases and other mast cell mediators

442
Q

what happens in response to IgE cells and mast cells in an allergic reaction when Omalizumab is used?

A
removes ige from circulation
binds to fc region that normally sits in receptor, so ige cant occupy receptors anymore 
stops ige initiating inflammation
exceptionally effective
given as subcutaneous injection
given depending on some factors
443
Q

what are the benefits of using omalizumab?

A

Specific targeting of allergy without side effects of steroids very rare to have side effects
Reduces exacerbations by about 3/4, though has less effect on background lung function
Doesn’t replace inhaled therapies - starts as an add on therapy
Can replace oral steroids – long term can replace this

444
Q

what are the disadvantages of using omalizumab to treat eosinophilic asthma?

A

Hospital (mostly) administration given by injections (1-4 injections very 2-4 weeks)
Subcutaneous injection(s)
Side effects: ? increased risk of vascular episodes, low risk of anaphylaxis – very low risk
Uncertain use in pregnancy
Cost - £10,000-30,000 per year for each patient

445
Q

apart from omalizumab, what are 2 other drugs that can be used to treat eosinophilc asthma? describe them

A

If you have eosinophilic asthma, mepolizumab…
reduces the frequency of attacks
decreases the amount of steroids you need orally
4 weekly dosing s/c, relatively few side effects (headache, ? anaphylaxis)
No significant immunosuppression side effects
Issues over cost; NICE appraisals and implementation
convenient
nurse goes once a month and do injections in people homes
works very well

Reslizumab: intravenous anti-IL-5.
does same thing but its intravenous
single injection without having to find a vein but this reslizumab- more painful and needs someone with high level of skill to do it - -this is less convenient

446
Q

what is a drug that could possibly be used to treat COPD

A

mepolizumab - not sure yet if it can be used as not 100% sure that eosinophils are the cause of COPD although it has been seen in COPD exacerbations
early trials have found that they have a limited benefit when used in COPD patients

447
Q

describe another way of treating asthma that is not to do with targeting eosinophils directly - describe the drugs that exist to treat them and ate the way that they work

A

reslizumab, mepolizumab and benralizumab are all used and work as anti-il5 agents
Neutralising IL-5: reslizumab, mepolizumab
Blocking IL-5 receptor and causing immune system- mediated eosinophil death: benralizumab
works in2 ways:
- doesnt allow il5 to be recognised by e
- coats e in itself so immune system kills it *(as it recognises it as foreign)

448
Q

how does benralizumab work to treat atshma?

A

works in 2 ways

  1. doesn’t allow il5 to be recognised by eosinophils by binding to its receptor so cant react. immune system sees the cells covered in the drug which it knows to be foreign so it gets rid of it
  2. coats the eosinophil in benralizumab so the immune system kills it because it is recognised as being foreign
449
Q

what is the name of the drug that targets il4 receptors and what has its effectiveness been in astha treatment

A

used to treat only eczema
works by targeting the il4 receptor
clinical rials have shown that it can reduce frequent exacerbations by 60%, improve lung function and also decrease the need for oral steroids

another study found that compared to a control group, those treated with Dup had a 3.5% hospitalisation (compared to 6.5%)
of the 210 participants who had been using oral steroids, 50% had been completely taken off them and 80% significantly reduced their steroid use by 1/2

450
Q

how do Tezepelumab work?

A

works by binding to TSLP which is a member of the cytokine family and is a central regulator of the type 2 immunity. once bound, stops them from interacting with the TSP receptor. study showed that those who received this drug had lower rates of clinically significant asthma exacerbations compared to the placebo. regardless of the asthma group, it worked i.e. on eosinophilic and neutrophilic related asthmas

451
Q

how do Azithromyocins work to treat asthma? what are some problems with its use?

A

targets nfkb etc to reduce infammation
fewer acute attacks and better control of asthma
not sure how it works completely
infection and inflammation both improved
used more and more – so more people in public = exposed to this drug
az = used a lot in other infections and so the use of this too much risks it being not effective later in these infections (resistance)

452
Q

describe the AMAZE study and what it found

A

400 participants, placebo-controlled
AZM 500 mg 3x wk
Minimal S/Es, mainly diarrhoea, hearing impairment withdrawals placebo group only
Inc ex smokers, ICS/LABA but few LAMA
60% reduction in exacerbations regardless of asthma phenotype
Some increase macrolide resistant organisms such as PsA

453
Q

what are side effects of using AZM drug to treat asthma? what was used before it?

A

causes diarrhoea
other then that, very few side effects
used to only be used to neutrophilic asthma etc

Macrolides used to be used but had very little evidence

454
Q

what types of asthma can AZM be used to treat?

A

both eosinophilic and non-eosinophilic asthmas

455
Q

why are AZM seen as better than biologics? but what is a disadvantage of AZM?

A

Biologic: £5k - £30k/ year
AZM: £100-200/ year
(-) is a lack of license

456
Q

what are the 5 considerations that doctors ned to make before prescribing antimicrobials?

A

Are antibiotics necessary?
What is the site of infection?
What is the organism sensitivity?
What is the appropriate or available route of administration?
What antibiotics are safe for the patient?

457
Q

what is the difference between anti-microbials and antibiotics?

A

most of the agents used are semi-synthetic derivatives of antibiotics and so are called anti-microbials

458
Q

what are the main causes of sore throats (viral, bacteria etc)?

A

Laryngitis – viral
Tonsillitis – viral&raquo_space; bacterial
Strep throat(streptococcal infections)
Glandular fever – viral (Epstein Barr)

459
Q

what do doctors use to test whether the sore throat is a bacterial infection or not? is this reliable?

A

hey use the centor criteria
Tonsillar exudate
Tender anterior cervical adenopathy (neck glands)
Fever over 38°C (100.5°F) by history
Absence of cough.
If 3 or 4 of Centor criteria are met, the positive predictive value is 40% to 60%. The absence of 3 or 4 of the Centor criteria has a fairly high negative predictive value of 80%.

460
Q

what are the names of bacteria, viruses and common pollutants that cause exacerbations in COPD? what is the most likely cause?

A

Bacteria
Streptococcus pneumoniaeandHaemophilus influenzae»Moraxella catarrhalis.
Staphylococcus aureus(influenza season).
Pseudomonas aeruginosa
Viral
rhinoviruses, influenza, parainfluenza, coronavirus, adenovirus and respiratory syncytial virus.
Common pollutants
nitrogen dioxide, particulates, sulfur dioxide and ozone
more often caused by viruses/pollutants than bacteria

461
Q

what is the problem with COPD being caused by a bacteria?

A

it is hard to tell if the cause is bacterial and so if it isn’t, giving antibiotics would be useless but if it is, would have missed this
also, takes several days to be able to find out if COPD has been caused by the bacteria after several days of them being diagnosed

462
Q

what is an anti-viral?

A

prevents transcription of genetic material

a small molecule that works by blocking nucleic acud synthesis r binding to a target site

463
Q

What is an antibiotic?

A

Antibiotics as defined by Semar Waksman are;
“ agents produced by micro-organisms that kill or inhibit the growth of other micro-organisms in high -dilution”
Produced by micro-organism so gastric juice (acid), is not antibiotics.
Penicillin discovered by Alexander Fleming in 1929 was the the first antibiotic.

464
Q

what do antimicrobials include?

A

Antibiotics
Antibacterial agents
Antivirals

465
Q

what are some common viral infections that affect the respiratory tract?

A

influenza A and B and RSV

466
Q

what is a part f the cll that can be targeted by a competitive inhibitor to stop viral replication? describe it. what are examples of a drug for this?

A

neurominodase – as virus comes out of cell, must disconnect from moitee using neuromindoase enzyme. virus cant bud if there is a neuromindase inhibitor and so can’t infect other cells
Oseltamivir ‘tamiflu’ or Zanamivir ‘Relenza’

467
Q

how effective are neuroaminodase inhibitors in treating colds?

A

shorten the symptoms of influenza-like illness by about half a day in adults (but not in asthmatic children), compared to a placebo.
no reliable evidence for reducing the risk being admitted to hospital or developing pneumonia, bronchitis, sinusitis or otitis media

468
Q

what is the problem if you start treatment of cold with neuroaminodase inhibitors too late?

A

by then, the virus would have already spread as this inhibitor is meant to stop the virus leaving and transfecting other cells. so, it would be ineffective

469
Q

what is a good antiviral that targets RSV? explain how it works

A
Ribavirin
is a nucleoside inhibitor.
an analogue of guanosine 
stops viral RNA synthesis
good in vitro activity against RSV
--antiviral ribavirin
prevents nucleic acid synthesis
G analogues like RSV, once the enzyme (causing rep), when it puts it in the chain, stops next nucleotide from jining onto it (its an analogue that doesnt have the right side chains so doesnt allow next acid to bind) – so tops rep of RSV
470
Q

what are some problems with using ribovirin treat RSV. but what is a conflict to these disadvantages

A
  • shown to have devastating effects in young children
    Possible shorter duration, less need for ventilator or oxygen
    Early use of inhaled ribavirin has been shown to reduce morbidity and mortality in adult bone marrow cell transplant recipients
471
Q

how do antibacterials generally work? what parts of the bacteria do they target?

A

defined as points of biochemical reaction crucial to the survival of the bacterium
penicillin-binding proteins in cell wall
cell membrane
DNA
Ribosomes
Topoisomerase IV or DNA gyrase
the crucial binding site will vary with the antibiotic class

472
Q

how does penicillin binding proteins work? what are some examples?

A

BINDING TO CELL WALL AND INHIBITION OF CELL WALL SYNTHESIS – Penicillin binding proteins PBP
disrupts the ability to maintain and carry on building cell wall so the cell dies
BETA LACTAMS (PENICILLINS AND CEPHALOSPORINS)
GLYOPEPTIDES

473
Q

how do metronidazole prevent bacterial survival?

A

INTERFERENCE WITH NUCLEIC ACID SYNTHESIS OR FUNCTION
e.g. METRONIDAZOLE,
RIFAMPICIN

474
Q

how can you stop bacterial DNA from being read in the cell?

A

INHIBITION OF DNA GYRASE

e.g. FLUOROQUINOLONES

475
Q

how do aminoglycosides and macrolides inhibit the survival of bacteria?

A

INHIBITION OF RIBOSOMAL ACTIVITY AND PROTEIN SYNTHESIS
AMINOGLYCOSIDES, TETRACYCLINES,
MACROLIDES, CHLORAMPHENICOL

476
Q

what is used to terminate bases of bacteria (stop it from being made) and how can this be used as an anti-bacterial drug?

A

INHIBITION OF FOLATE SYNTHESIS AND CARBON UNIT METABOLISM

SULPHONAMIDES AND TRIMETHOPRIM

477
Q

list the pathogens that exist that can cause community acquired pneumonia

A
Streptococcus pneumoniae (40%)
Mycoplasma pneumoniae (~10% peaks in epidemic seasons)
Chlamydophila pneumoniae (~10%)
Legionella pneumophila and other spp.(<5%)
Haemophilus influenzae (<5%)
Klebsiella pneumoniae (rare; homeless and in hospital)
Staphylococcus aureus (low % in community but increased after influenza and in hospital especially)
Viruses (≥10%)
478
Q

why is it harder to treat atypical pathogens compared to other ones?

A

atypical pathogens – have certain characteristics that make tem harder to detect and so need diff treatment

479
Q

describe streptoccchus pneumoniae

A
Gram positive cocci
Alpha haemolytic, optochin sensitive
>>90 serotypes
any age but increases at extremes of age
can be severely ill with respiratory failure or sepsis
‘invasive pneumococcal disease’
480
Q

what can be used to treat S. Pneumonia?

A

best drugs in clinical traisl – beta lactams – amoxixillin
most time amoxicillin works

macrolides – inhibit ribosome
fluroquinolones – inhbit DNA gyrase

481
Q

is beta lactams have been the most successful treatment of s.pneumonia in trials, why are macrolides and fluoroquinolones used?

A

macrolides and fluoroquinolones are not used in UK as penicillin and b lactams work
in other countries, b-lactam has too much resistance and so these ones are more commonly used
macrolides more commonly used in those allergic to blactams

482
Q

describe H. influenzae, Steph. aureus and Klebsiella Pneumoniae

A
H. influenzae
Gram negative cocobacilli
Ealry 20th C thought to be cause of Influenza
Encapsulated – e.g. HiB 
Unencapsulated ‘non typable’ NTHi
Staph. aureus
May complicate recent influenza, 
ventilator-associated pneumonia too
Klebsiella pneumoniae
Gram negative bacilli, Enterobacteriaceae
Normal flora of mouth and intestines
homeless, alcoholic, Hospital –associated
483
Q

state drugs to treat H. influenzae, Steph. aureus and Klebsiella Pneumoniae

A
H. influenzae
-β-lactam antibiotics:
Amoxicillin +clavulanic acid ‘co-amoxiclav’
Tetracyclines: Doxycycline
But NOT Macrolides
S. aureus
- β-lactam antibiotics:
Flucloxacillin
Cefuroxime
MRSA?
Vancomycin
Linezolid

Klebsiella pneumoniae
β-lactam antibiotics:
Co-amoxiclav
Cephalosporins

484
Q

list the atypical pathogens, what existing drugs they respond to and why they are difficult to detect

A
Legionella pneumophila
Mycoplasma pneumoniae 
Chlamydophila pneumoniae
Chlamydophila psittaci
Coxiella burnetti

dont respond to B lactams but do respond better to macrolides and fluoroquinolones

485
Q

what is teh analogy ESKAPE used for?

A

The most common and serious MDR pathogens have been encompassed within the acronym “ESKAPE,” standing for Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.

486
Q

describe both early and late onset Hospital acquired Pneumonia - which are more highly resistant?

A
Micro-organisms
Early onset ≤5d of hospital 
organisms similar to CAP and also anaerobes
Late onset >5d;
Staph. aureus including methicillin resistant (MRSA)
Pseudomonas aeruginosa
Acinetobacter baumanii
Klebsiella pneumoniae
487
Q

what is used to treat TB and when?

A

TB is curable
Treat with at least 4 drugs for at least 6 months
Standard treatment in UK:

rifampicin
isoniazid
both for 6 months (or 12 months if in CNS)
\+
pyrazinamide
ethambutol
or the first 2 months
488
Q

why are rifampicin and isoniazid given for 12 months if TB is in CNS?

A

extra considerations if tb - – if in CNS, drugs given for longer to make sure you’ve gotten rid of all of it

489
Q

how do the different special anti-microbials for TB work?

A

rifampicin – tagets rpob
pyrazinamide- has to be converted to where the bacterias are. inhibits fatty acids syntahse (cell wall fromation), disrupts chnges in intracellular ph makign it less lilely to survive
isoniazid – KatG – place develops in microbacteria. if it doesnt fit twith the compound, bcteria will survive. if changed int pe
stop unfolding of DNA heix and prevemtign transcription adnd translation

490
Q

what are the 4 ways that bacteria resist antibiotics?

A
  • Change antibiotic target
  • Destroy antibiotic
  • Prevent antibiotic access
  • Remove antibiotic from bacteria
491
Q

how can an antibiotic be destructed/destroyed when a bacteria becomes resistant?

A

The antibiotic is destroyed or inactivated
Beta lactam ring of Penicillins and cephalosproins hydrolysed by bacterial enzyme ‘ Beta lactamase’ now unable to bind PBP
Staphylococci produce penicillinase so penicillin but not flucloxacillin inactivated
Gram negative bacteria phosphorylate and acetylate aminoglycosides (gentamicin)

492
Q

how can there be prevention of antibiotic entry?

A

modify the bacterial membrane porin channel size, numbers and selectivity
Pseudomonas aeruginosa against imipenem,
Gram negative bacteria against aminoglycosides

493
Q

how can you remove

antibiotic from bacterium? give specific examples

A

Proteins in bacterial membranes can act as an export or efflux pumps
Level of antibiotic is reduced
S. aureus or S. pneumoniae resistance to fluoroquinolones
Enterobacteriacae resistance to tetracylines

494
Q

what are the 2 wasy that resistance can develop?

A
  • intrinsic - Naturally resistant
    Example of intrinsic resistance:
    Aerobic bacteria are unable to reduce metronidazole to its active form
    Anaerobic bacteria lack oxidative metabolism required to uptake aminoglycosides
    or some bacteria- dont have target sites to begin with – these are intrinisc classes of resistance
  • acquired: spontaneous gene mutation or Horizontal gene transfer
    (conjunction, transduction and transformation)
495
Q

describe how horizontal gene transfer happens n acquired resistance and how spontaneous gene mutation occurs

A

conjugation - Sharing of extra-chromosomal DNA plasmids “Bacteria sex’
Transduction - Insertion of DNA by bacteriophages
Tranformation - picking up naked DNA

spontaneous gene mutation - 
New nucleotide base pair
 change in amino acid sequence 
change to enzyme or cell structure 
reduced affinity or activity of antibiotic
496
Q

what is the difference between primary and acquired resistance? and what is used to treat them

A

Primary Resistance
Innate property eg Pseudomonas and penicillin

Acquired Resistance
Due to mutation or gene transfer
Chromosomal eg M.tuberculosis
Plasmid mediated eg MRSA

497
Q

what should be considered when determining the potency of an antibiotic against a bacterium?

A

protein binding,
pharmacokinetics,
distribution into the site of infection,
the adequacy of the patient’s host defenses
exposure of an organism to an antibiotic needed for its eradication
musn’t be toxic so ned to find tis minimum inhibitory concentration

498
Q

what are the steps required in a good pharmacokinetic drug?

A

(1) release from the dosage form;
(2) absorption from the site of administration into the bloodstream;
(3) distribution to various parts of the body, including the site of action and
(4) rate of elimination from the body via metabolism (LIVER) or excretion (KIDNEY) of unchanged drug.

499
Q

what needs to be considered when thinking about if an anti-biotic is safe for patients to use?

A
intolerance, allergy and anaphylaxis
side effects
age 
renal function
liver function
pregnancy and breast feeding
drug interactions
risk of Clostridium difficile (5 ‘C’s)

if kidneys arent working, get toxic levels of drug
liver not workng, may metabolise the drug too fast/slow etc
very rarely give 1 drug to soemone
interactions betwenen drug and others given – key
antibiotic associated diarrhoes – utgrowth of bacterial inefction (Clostridium difficile) – can allow this which is usually resistant to cause toxic affects in the GI tract

500
Q

what are the 5 C’s in risk of Clostridium difficile?

A

Ciprofloxacin

Clindamycin

Cephalosporins

Co-amoxiclav (augmentin)

Carbapenems, e.g. meropenem (for most difficult to treat patients)

501
Q

what are some advantages and disadvantages of using nuclear missile or snipers to be antimicrobials?

A
Nuclear missile
Will work (for now)
Expensive
Adverse effects
Harder to give
Promotes resistance
Sniper
Might miss
Narrower AE profile
More tolerable
Saves other choices
cheaper
502
Q

describe the different antibiotic therapy treatment options for those with low-severity – high severity CAP

A

Offer a 5‑day course of a single antibiotic amoxicillin, to patients with low‑severity community‑acquired pneumonia.

Considerdual antibiotic therapywith amoxicillin and a macrolide for patients with moderate‑severity community‑acquired pneumonia.

Consider dual antibiotic therapy with a beta‑lactamase stable beta‑lactam[1]and a macrolide for patients with high‑severity community‑acquired pneumonia.