TI Flashcards

1
Q

What is Primary Cell Culture?

A

A technique where cells are isolated from the body tissues and grown in culture, trying to create in vivo conditions.

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

What are examples of non-haemopoeitic primary cultures?

A
  • Liver
  • Muscle
  • Fibroblasts
  • Skin
  • Nerves
  • Endothelial Cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are examples of haemopoietic primary cultures?

A
  • Stem, progenitor cells
  • Monocyte, macrophages
  • T and B cells
  • Dendritic cells
  • Osteoblasts
  • Erythrocytes
  • Megakaryocytes, platelets
  • Neutrophils, Eosinophils, Basophils, Mast Cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the 6 features of primary cell cultures?

A
  • cells derived directly from tissues
  • interpatient variability
  • finite lifespan
  • cells divide and/or differentiate
  • cells carry out normal function
  • cell lines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What happens in the disaggregation of cells?

A
  • cells are allowed to migrate out of an explant
  • mechanical dissociation
  • enzymatic dissociation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What type of cell doesn’t need to be disaggregated in primary cell culture and why?

A

Haemopoietic cells because they are already disaggregated

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

What are the sources of stem cells?

A
  • Bone marrow
  • Umbilical cord blood
  • Mobilised peripheral blood
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the path from stem cells to blood cells?

A
Stem cells...
Early progenitors...
Late progenitors...
Immature precursors...
Red cells/neutrophils/platelets!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What do the stem cells that sit out of the cycle do?

A

either divide to produce identical clones of themselves or differentiate

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

At what point do stem cells become visually distinguishable?

A

immature precursors

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

What does CFU stand for?

A

Colony forming unit. (the letters at the end show which lineages they are limited to)

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

What are the constituents of bone marrow?

A
  • Areas of tightly packed cells - progenitors and early stem cells
  • Blood vessels
  • Fat
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are the defining features of stem cells?

A
  • pluripotent
  • self-renew
  • rare
  • responsible for engraftment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the defining features of progenitor cells?

A
  • undifferentiated
  • undistinguishable by morphology
  • committed to one or more lineages
  • detected in colony forming assays
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the defining features of precursor cells?

A
  • immature but recognisable
  • starting to differentiate
  • few final divisions before they become mature cells and move into the peripheral blood
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What do haematopoeitic growth factors do?

A

They are polypeptide growth factors, also known as cytokines.
They bind to the cell surface transmembrane receptor and stimulate growth and survival of progenitors.

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

About the structure of stromal cells…

A

they have an extracellular matrix and adhesion molecules on their surface. They provide cytokines.

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

Why do stem cells need to be processed?

A

stem cells are few in number and so we want to enrich and purify them

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

What are processing methods of stem cells?

A
  • Erythrocyte lysis - ENRICHMENT
  • Density gradient centrifugaton - ENRICHMENT
  • Adherence depletion - ENRICHMENT
  • Antibody depletion - PURIFIED STEM CELLS
  • Antibody selection - PURIFIED STEM CELLS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Why are progenitors called CFUs?

A

Because they grow to form colonies of mature cells, containing up to 1000s of cells.

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

What do you need for a colony assay?

A
  • semi-solid medium (eg agar)

- growth factors

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

What happens in colony assays?

A

Larger progenitors are forced to mature and divide into colonies using growth factors when put into culture on a semi-solid medium. We can quantitate the number of CFUs we had originally and can use a microscope to identify the different CFUs.

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

What is LTBMC?

A

long-term bone marrow culture

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

What are the steps of LTBMC?

A
  • bone marrow
  • ficoll gradient
  • liquid culture
  • CFU
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What happens in LTBMC?

A

We use flasks in which the stromal cells are at the bottom. The stem cells bury into the stromal cells, which stimulates them to divide. The stem cells emerge from the stromal cells as committed cells (stem cells remain in the stromal cells) and move into the supernatant. There is a direct relationship between early stem cells and later progenitors.

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

What are the applications of primary cell culture techniques?

A
  1. Research - basic haematopoeisis and carcinogenesis
  2. Testing toxicity of chemotherapeutic agents and carcinogens
  3. Generate cells for stem cell transplantation/manipulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

How are cells isolated from the blood?

A
  1. Density centrifugation
  2. Fluorescence activated cell sorting (FACS)
  3. Immune-precipitation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

How are cells isolated from solid tissue?

A

Mechanical and enzymatic disruption using collagenase, dispose and trypsin

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

What are the pros of primary culture?

A

Unmodified

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

What are the cons of primary culture?

A
  • aberrant expression of some genes
  • variable contamination
  • poor growth characteristics (50-100 divisions)
  • inter-patient variability
  • phenotype instability
  • molecular manipulation is difficult
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the idea model for cell culture?

A
  • Good growth characteristics
  • Phenotype stability
  • Defined population
  • Molecular manipulation readily achieved
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the two ways in which cell lines can be derived from primary culture?

A
  1. Spontaneous - from tumours or prolonged culture, multiple ill-defined mutations, transformed phenotype
  2. Genetic manipulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What do we target to generate cell lines?

A

Processes that regulate growth and ageing…

  • p53
  • retinoblastoma gene (Rb)
  • telomerase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is the role of p53 in a normal ageing cell?

A
  • telomere binding protein protects the telomere ends (TTAGGG)
  • when the telomere gets too short, the binding protein is lost
  • p53 binds to the unprotected chromosomes
  • activated p53 triggers growth arrest or cell death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is the t-antigen?

A

A viral protein which interacts with normal p53 and Rb. This can cause increased growth without loss of function.

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

What is HPV/what does it do?

A

A viral protein - E6 targets p53 for degradation, E7 binds to Rb

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

What viral genes are commonly used to generate cell lines?

A
  • SV40; T and t antigen
  • HPV; E6/E7

…p53 and Rb bind to different viral proteins

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

What is the role of telomerase?

A

Telomerase prevents the erosion of telomeres. Some cells need telomerase and silencing of Rb for ‘immortalisation’. E6/E7 and telomerase are believed to maintain a differentiated phenotype.

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

What are the steps in the derivation of cell lines?

A

Plasmid…Gene for selection…Growth promoting gene
Transfection
Selection pressure added
Colonies selected

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

When generating cell lines, what hurdles have to be overcome?

A
  • getting the DNA into the cells

- getting the cells to stably incorporate the DNA once inside

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

What are the 5 methods of transfection?

A
CaPO4 co-precipitation
Lipofection
Electroporation
Viral transfection
Nucleofection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is lipofection?

A
  1. positively charged lipoplexes interact with the negatively charged cell membrane
  2. taken up by endocytosis
  3. release from endosome
  4. transport to the nucleus
  5. entry to the nucleus is inefficient and may need mitosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is electroporation?

A

An electrical field is applied to the cells in order to increase permeability of the cell membrane, allowing chemicals, drugs or DNA to be introduced to the cell.

The pores reseal when they have been created by the electrical current, and the rate of this resealing is dependent on temperature.

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

What is viral transfection?

A
  • Exploits the normal mechanism of viral infection
  • Usually has high transfection efficiency
  • There are three gene-targeting vectors commonly used based on three types of viruses; retrovirus, adenovirus, and adeno-associated virus.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is nucleofection?

A
  • combination of electroporation and lipofection
  • increased efficiency particularly of non-dividing cells
  • technology is protected under patent
  • different solution and protocols are used for each cell type
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the disadvantages of cell lines?

A
  • RAPIDLY DIVIDING CELLS OFTEN LOSE DIFFERENTIATED FUNCTION
  • therefore in making cell lines, growth and function are at odds
  • the ideal solution would be a cell line that divides when you want more and stops when a study of function is requited
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the means to overcome loss of function of cells in cell lines?

A
  • conditional mutant eg T-antigen of SV40

- changing culture conditions such as the ECM, co-culture or 3D culture

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

What is 3D culture?

A

An artificially created environment in which biological cells are permitted to grow and interact with their surroundings in all three dimensions

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

What are the advantages of 3D cultures?

A
  • genes expressed as in vivo
  • cell-cell communication is re-established
  • cells are orientated in the same was as in tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the three ways of Light Microscopy?

A
  • histology
  • phase contrast
  • time lapse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is Stoke’s Shift?

A

Due to energy loss, the emitted light is shifted to longer wavelength relative to the excitation light. It is the difference between the two peaks on an excitation-emission graph.

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

What is photobleaching?

A

Bleaching of fluorochromes - due to high intensity illumination the flurophores might permanently lose their ability to emit light

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

How can you avoid photobleaching?

A
  • Work with reduced excitation light intensities or use grey filters
  • Use shorter exposure times/higher gain settings and longer intervals during time lapse studies
  • Use anti-bleach in your mounting media
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are fluorescent proteins?

A

These proteins are naturally found in light-producing cells of cnidarians. Fluorescent proteins can be fused with other proteins and introduced in cells via transfection. This allows live study of fluorescent tags in living cells/organisms.

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

What is the benefit of confocal microscopy over wide field microscopy?

A

Higher z-resolution and reduced out-of-focus-blue make confocal images crisper and clearer

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

What is the negative of confocal microscopy compared to wide field microscopy?

A

Only a small volume can be visualised by confocal microscopes at once. Bigger volume need time consuming sampling and image reassembling.

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

What are the characteristics of stem cells?

A
  • can self-renew

- can differentiate into a range go more specialised cells

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

What is meant by potency of stem cells?

A

Their ability to differentiate

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

What is multi potency?

A

can only form one type of cell - i.e. HSCs can only form blood cells, but various types of blood cell

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

What is totipotency?

A

Can produce all of the differentiated cells in an organism. Can give rise to the embryo and the tissues that support its development, such as the placenta. These supporting tissues are known as the trophoblast.

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

What is pluripotency?

A

Can give rise to all the cell types that make up the body. This is similar to totipotency but don’t make the supporting tissue, only the embryo.

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

What do multipotent stem cells do for the body?

A

(aka Adult stem cells) function as a repair system for the body, replenishing old or damaged tissues

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

What is asymmetric stem cell division?

A

produces one stem cell and one differentiated cell. QUIESCENCE

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

What is symmetric cell division?

A

Produces 2 stem cells. ACTIVATION - leads to expansion of the stem cell pool and occurs due to injury/disease

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

What is the stem cell niche?

A

This is the area in which stem cells reside. It is the microenvironment where you find the stem cells, and it interacts with the stem cells to determine their fate. It also regulates stem cell activity.

The niche helps to maintain the balance between symmetric and asymmetric stem cell division.

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

What is the Extracellular Matrix in terms of adult stem cells?

A
  • key component of the stem cell niche
  • two way interaction; stem cells also remodel the ECM
  • critical regulator of stem cell function
  • mechanical features can determine cell fate
    …brain stiffness induces neuron formation…
    …muscle stiffness induces muscle formation…
    …bone stiffness induces osteoblast formation…
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

How is the lifespan of a stem cell extended?

A
  • can undergo more cell divisions than somatic cells
  • length of telomeres determines how many divisions can occur
  • have high expression of telomerase which rebuilds the telomere
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is the therapeutic potential of adult stem cells?

A
  • have clinical applications
  • can be autologous - won’t be rejected
  • fewer safety concerns - no need to immunosuppressants, not linked to tumour genesis
  • easier to get sufficient numbers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Where are Mesenchymal Stem Cells (MSCs) found?

A
Primarily found in bone marrow but also in...
placenta
umbilical cord blood
adipose tissue
adult muscle
corneal stroma
dental pulp
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Why do MSCs have therapeutic potential?

A
  • homing capacity - tend to home to damaged tissue sites
  • high differentiation potential
  • production of trophic factors
  • immunomodulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is osteogenesis imperfecta?

A

Also known as ‘brittle bone disease’, this is a genetic disorder in which osteoblasts produce defective type I collagenase. Symptoms include osteopenia, multiple fractures, severe bony deformities and shortened stature.

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

What are limbal stem cells?

A

Corneal renew and repair are mediated by stem cells of the limbus.

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

What is transdifferentiation?

A

The process by which an adult cell can change from its initial differentiated state to another. This is largely an in vitro phenomenon.

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

What are cancer stem cells responsible for?

A

The growth of tumours is fuelled by limited numbers of dedicated stem cells that are capable of self-renewal. Many studies have suggested that CSCs are resistant to cancer therapy which could explain relapse as the CSCs can relocate and form a tumour in the new location. Therefore, if we target therapeutics specifically to CSCs we may be able to cure the cancer permanently.

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

What can we use to target cancer stem cells?

A
  • surface markers
  • signal cascades
  • ABC cassette
  • the microenvironment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What does MRD stand for?

A

Meaningless Random Differences

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

What does DWC stand for?

A

difference is worth considering

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

What does SUD stand for?

A

statistically unreliable differences

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

What does FCD stand for?

A

fairly convincing differences

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

What is the advantage to the visual approach to data analysis?

A

Immediate visual summary of effect size and confidence

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

What are the disadvantages of the visual approach to data analysis?

A
  • does not give a precise measure of confidence

- comparing one confidence interval to another cannot be translated into a numerical unit

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

What are the advantages to using p-values?

A

gives a single number that summarises whether your data looks random

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

What are the disadvantages of using p-values?

A
  • very little information
  • doesn’t tell you effect size
  • doesn’t tell you confidence levels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What is a type 1 data error?

A

When we get p

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

What is a type 2 data error?

A

When we get p > 0.05 even though there is a real effect. This often occurs when you haven’t repeated the experiment enough, or your sample size is too small.

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

What should you do if the data differs substantially from the normal distribution?

A
  • mathematical transformation e.g. take a log of the data
  • non-parametric test
  • Mann Whitney U test instead of T-test
  • Wilcoxon signed ranks test instead of paired T-test
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What should you do when comparing more than two groups?

A
  • not valid to do a series of t-tests for each pair
  • multiple testing increases the frequency of the type 1 errors
  • an alternative is to use 1-way analysis of variance (ANOVA)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What does an ANOVA test do?

A

Looks at data from all groups and asks how likely or unlikely it is that the data as a whole is random. ANOVA compares the variance within each group to the variance between groups.

The variance within each group gives you a measure of random variation.

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

In ANOVA testing, if the variance between groups is small compared to within groups what does this suggest?

A

This suggests that the different between groups could be explained by random variation.

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

In ANOVA testing, if the variance between groups is large compared to within groups what does this suggest?

A

This suggests that it is unlikely this can be explained by random variation.

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

What does it mean if the p-value for ANOVA is less than 0.05

A
  • unlikely the differences between groups is random
  • valid to ask which differences are responsible for this
  • can do individual tests between each pair of the groups
  • t-tests or other ‘post hoc’ tests
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What is pluripotency?

A

the ability of a cell to give rise to every tissue in the body

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

What are the two types of pluripotent stem cell?

A

embryonic stem cells (ES) and induced pluripotent stem cells (iPS)

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

What are the stages of derivation of an ES cell?

A
  • cleavage stage embryo
  • cultured blastocyst
  • isolated inner cell mass (grown on layer of irradiated mouse fibroblast feeder cells)
  • cells dissociated and replated to avoid overgrowth (and new feeder cells)
  • established ES cell cultures
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

what is the hinderance of ES cell lines?

A

only 10% of embryos produce these stem cells

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

The adult body is formed from a single cell (the oocyte) by…

A
  • migration
  • cell division
  • differentiation into different types (tissues)
  • apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

what drives differentiation of ES cells?

A

proteins secreted by cells called growth factors

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

What are the stages of pancreatic development?

A
  • formation of the GUT TUBE
  • formation of the dorsal and ventral BUDS
  • BRANCHING
  • endocrine progenitors MIGRATE in mesenchyme, where they differentiate and form ISLETS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What do the pancreatic islets contain?

A

the insulin-secreting beta cells

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

What are current projects of tissue engineering?

A
  • biodegradable polymers for nerve regeneration
  • hydrogels for musculoskeletal repair
  • electrospun fibres to allow generation of artificial tendons
  • windpipe made of glass
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What are therapeutic targets of regenerative medicine?

A
  • heart disease
  • type I diabetes
  • CNS trauma
  • neurodegenerative disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What are the disadvantages of embryonic stem cells?

A
  • ethically contentious
  • transplantation would involve immunosuppression
  • immune matching involves generating >1000 variable ES cell lines in the UK
  • far more difficult to derive than first anticipated
  • currently 300+ human ES cell lines world wide and very few clinical trials
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What are induced pluripotent stem cells (iPS)?

A

a type of pluripotent stem cell that can be generated directly from adult cells - formed when you force the expression of 4 genes in fibroblasts (skin cells)

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

What are the yamanaka factors?

A

When first investigating iPS cells, scientists found there were 3 genes that caused complete loss of colonies when they weren’t included:

  • factor 14 (oct 4)
  • factor 15 (sox 2)
  • factor 20 (klf 4)

They also noticed that removal of cymc (factor 22) allowed colonies to form but they didn’t look right.

They went on to show that these 4 factors alone could cause the formation of ~160 colonies, whereas the most a 3 factor combination could form was ~50

These 4 factors are known as the YAMANAKA FACTORS and are still used today.

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

What are methods of pluripotency assessment of iPS cells?

A
  • compare cell morphology
  • look for expression of pluripotency associated genes using PCR
  • check for expression of markers by fluorescent staining
  • look for proteins that should be expressed in the nucleus (oct 4, Nanog and sox 2) and on the cell surface (Tra160 and SSEA4)
  • functionally assess their pluripotency
  • functionally to form chimeric mice using green fluorescent protein, the GFP gene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

How has the original iPS technique been adapted to improve efficiency and safety?

A
  • using range of viruses and plasmids to deliver the genes

- safety is a consideration because many of the genes put in to make iPS genes are oncogenes

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

What are the main stages of iPS reprogramming?

A
  • initiation stage
  • maturation stage
  • stabilisation stage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What happens in the initiation stage?

A

A key feature of initiation stage is MET. The starting cell population has a mesenchymal phenotype and when reprogramming is initiated they start to become more rounded and closer together. This is completed by day 6.

By day 21 they have formed colonies of pluripotent stem cells which have a very high nuclear-to-cytoplasmic ratio, and are tightly packed epithelial-like cells.

As well as MET they cells increase proliferation as a result of the cymc, lin 20 and shp 53 expression.

They also resist signals from other cells causing them to apoptose or senesce.

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

What are the advantages of iPS cells?

A
  • share most of the attributes of ES cells i.e. self renewal and pluripotency
  • can be generated directly from patients own cells so they are autologous and won’t be rejected
  • no ethical issues
  • present us with the opportunity of taking patients blood or skin cells and can reprogram then to iPSCs to study disease in test tubes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What are the disadvantages of iPS cells?

A
  • current production methods involve inserting genes that could cause cancer
  • these current methods are too inefficient to guarantee production of iP cells for each patient
  • iPS cells may not differentiate as reliably to all types of cells
  • great potential but need to address these challenges
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is the structure of antibodies?

A

Antibodies produced are a heterogenous mixture of Igs to different epitopes. They have variable regions and constant regions, light chains and heavy chains.

The variable regions vary amongst different Igs and is the specific region that ‘sticks’.

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

What is the normal immune response?

A

The normal immune response is polyclonal - involving several monoclonal lymphocytes recognising different epitopes and their progeny

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

What are epitopes?

A

part of an antigen on its surface

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

What are the reasons for target antigens having multiple epitopes?

A
  • promotes a range of diverse specific antibody binding sites - not just a SINGLE antibody recognising a SINGLE antigen, will be a single antibody recognising a single epitope.
  • different antibodies recognise different parts of the same antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the advantages of using antibodies as a reagent?

A
  • high specificity
  • possible avidity (how well they tick together) for virtually any antigen including novel synthetic molecules
  • better than alternative - peptides need conjugation with large ‘carrier’ molecules to be immunogenic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What are considerations of the production of polyclonal antibodies?

A
  • cost
  • amount of antigen available
  • volume of sera required
  • phylogenic relationship between Ag origin and host
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is the process of the production of polyclonal antibodies?

A

1 - immunise antigen to activate B cells (add adjacent and then boost several times a few weeks apart)
2 - 4-8 weeks collect blood, clot, aliquot serum (test for activity)
3 - store (freeze) at -20C or 4C

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

What are the disadvantages of producing polyclonal antibodies?

A
  • ethical issues with using animals
  • every animal is different so may not be reliable for a specific test
  • need to make sure the antigen being given to the animal is the same each time which is difficult if the virus is rare e.g. HIV or ebola
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What is the alternative to making polyclonal antibodies?

A

making monoclonal antibodies

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

What are monoclonal antibodies?

A

they were first seen when looking at cancer cells. the cells are immortal so constantly produce the same antibody (as they come from the same cell). Overproduction of the same ‘monoclonal’ antibody can be dangerous, useful in diagnosis of some B lymphocyte tumours.

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

What are the advantages of making monoclonal antibodies?

A
  • high specificity
  • low possibility of cross reactivity with unrelated Ag
  • inexhaustible supply of consistent homogenous reagent
  • commercial applications
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

How are monoclonal antibodies produced?

A

1 - immunise animal with antigen of choice (test bleed reactivity for 3 weeks - antibody should be going up, then boost every day for 3 weeks)
2 - one day post booth harvest B cells
3 - take a myeloma cell (=neoplastic B cells) from a tumour culture
4 - fuse in a gel for 2/3 days then treat with a drug
[unfused B cells die - not immortal]
[unfused tumour cells die - from drug]
[fused cells are both B cells and tumour cells so they LIVE as they are immortal and resistant to the drug]
5 - plate out individual fused hybridomas and grow
6 - test each hybridoma medium supernatant for specific antibody by testing binding to antigen (ELISA)

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

What else can we do with monoclonal antibodies?

A
  • can re-engineer DNa encoding heaving and light V region which can be fused to activate molecules to target activity e.g. a toxin that kills tumour cells = therapeutic usage
  • can combine antigen binding site engineered in mouse with Ig constant regions form humans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is immunoblotting (western blotting)?

A

separates and detects specific proteins from a protein micture

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

How is immunoblotting done?

A

1 - denature cell line by treating with denaturing reagent e.g. SDS
2 - fractionate by length of polypeptides using SDS-PAGE
3 - transfer to a robust membrane eg nitrocellulose or nylon sheet
4 - hybridise to specific labelled antibodies to see what was specific (hybridise with a specific primary antibody, was, hybridise with a secondary antibody specific for primary antibody conjugated to fluorescent label, chemo-luminescent label or dye label)

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

What are the used of immunoblotting?

A
  • to determine the size (MW) of antigen
  • to see how a disease is progressing
  • to screen samples for antibody presence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is immunocapture used for?

A
  • to screen samples for antigen presence - use antibody to capture beta-HCG antigen from urine (pregnancy test)
  • rapid ELISA for fluA antigen
  • to screen samples for multiple antigen presence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What is immunopreciptation?

A

we can use this when we have a sample full of other things that we don’t want to denature (as would happen in western blotting when they were put through SDS system)

  • sample labelled
  • add and bind specific antibody
  • add Fe binding protein A and precipitate
  • wash to purify: add SDS to elute Ag from Ab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are the uses of immunoprecipitation?

A
  • to detect proteins of interest in a sample
  • to purify native proteins of interest
  • to characterise MW of proteins
  • to study structure of proteins or antibody/antigen complexes
  • to detect specific antibodies in a sample
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What is affinity chromatography?

A
  • purifies a soluble molecule from a mixture

- requires specific and reversible binding

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

What are the three stages of Affinity Chromatography?

A

STAGE 1: couple of ligand (antigen antibody) to a solid matrix in a chromatography column
STAGE 2: pass the sample through column allowing binding and discard the rest of the mixture
STAGE 3: elute off bound molecules. Low pH buffers disrupt non-covalent bonding

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

What are the advantages of Affinity Chromatography?

A
  • reusable

- can purify late quantities (manufacture)

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

What is immunomagnetic separation?

A

The same idea as affinity chromatography except instead of beads being stuck in the column, they are magnetic, and you use a magnet to pull them to one side.

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

What is the process of immunomagnetic separation?

A
1 - mix sample and beads and bind
2 - apply magnet
3 - pour or wash supernatant
4 - resuspend purified beads
5 - elute off beads if necessary
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are the uses of immunomagnetic separation?

A
  • mostly pulling pathogens out of sample that are very complex
  • pathogen specific Ab’s allow purification and concentration from complex sample e.g. toxigenic E.Coli from faeces
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What characterises epithelial cells?

A

They have tight junctions between the cells that restrict the passive diffusion of ions/substances across the epithelial cell layer.

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

What are the two oppositely facing cell surfaces?

A

luminal, apical, mucosal membrane: faces a hollow or fluid filled chamber

basolateral, serial membrane: normally close to a network of blood vessels

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

What is involved in ion transport processes in the lung epithelium?

A
  • tight junctions
  • polarised epithelium
  • vectorial transport of ions (ion channels, pumps, transporters)
  • transcellular and paracellular pathways for water movement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What is the driving force for Na+ entry?

A

the driving force is the different between the electrical and diffusional forces acting on an ion

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

What are the 3 potential sites for ion movement in epithelia?

A

1 - across the apical membrane
2 - across the basolateral membrane
3 - via the paracellular pathway

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

What is the equilibrium potential?

A

the membrane voltage (Vm) at which the driving force for net ion movement across a membrane is zero and is given by the Nernst Equation

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

What are methods of investigating transepithelial ion transport in whole organs?

A

in vivo/in vitro

water/solute transport

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

What are methods for investigating transepithelial ion transport in an isolated epithelium?

A
ussing chambers
short circuit current (ISC)
ion flux ratios
membrane permeabilisation
micro electrodes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What are methods for investigating transepitheial ion transport in membranes?

A

vesicles
patch clamps
artificial bilayers

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

What are ways of looking at water/solute?

A

1 - clearance methods i.e. absorption of glucose
2 - gravimetric measurement of net fluid absorption/secretion (Iv)
3 - marker infusion

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

What is the Ussing Technique for isolated epithelium in vitro?

A

1 - Short circuit current (ISC)

2 - Radiotracers

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

How does Short Circuit Current (ISC) work?

A
  • the resistance of epithelia to current flow by paracellular pathways is high
  • creation of a transepithelial potential (Vt)
  • values for Rt and Vt can be obtained by passing a current across the epithelium and applying Ohm’s law
  • the current flow required to maintain Vt at 0mV is the short circuit current (ISC)

Sum of ALL active electric ion fluxes across the epithelium

–> ion replacement, pharmacological blockade, permeabilisation studies

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

How do radio tracers work?

A

the measurement of unidirectional transepithelial radioisotope fluxes can be used to identify transport of specific ions irrespective of the transport mechanism

net fluxes across an epithelium are calculated from eh differences in oppositely directed unidirectional fluxes

ussing predicted that the unidirectional fluxes of an ion moving via simple diffusion should be of equal magnitude (Ussing flux equation)

fluxes that do not obey this equation are active transport processes

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

What is the simplest case where the Ussing flux ratio applies?

A
  • identical solutions either raid elf the membrane (Ca=Cb)
  • no PD i.e. short current conditions (Vt=0)

Jab/Jba = 1. exp (0)=1 — diffusion
Jab/Jba > 1 — active absorption
Jab/Jba

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

Why will fluxes of any single ion only represent a proportion of ISC?

A

Active transport correlated with ISC

If PD=0, ISC=sum of all net ion fluxes (I ionic)

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

What is flow cytometry?

A

a technique which simultaneously measures several physical characteristics belonging to a single cell in suspension. This is done by light scatter and fluorescence

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

What is the difference between flow cytometry and flow sorting?

A

flow cytometry = measuring properties of cells in flow
flow sorting = sorting (separating) cells based on properties measured in flow. Also called fluorescence-activated cell sorting (FACS)

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

What are methods of visualisation of antibodies?

A
  • fluorescence microscopy

- flow cytometry

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

What are the basics of flow cytometry?

A

Cells in suspension flow in single files through an illuminated volume where they scatter light and emit fluorescence that is collected, filtered and converted to digital values that are stored on a computer.

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

What is fluidics? (flow cytometry)

A

Need to have cells in suspension flow in single file. this is accomplished by injecting the sample into a sheath fluid as it passes through a small orifice. Same fluid flows in a central core that does not mix with the sheath fluid - laminar flow. Introduction of a large volume into a small volume - hydrodynamic focussing.

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

About lasers…

A

Lasers have a single wavelength of light (or more rarely a mixture of wavelengths).

They can provide from milliwatts to watts of light.

They can be inexpensive, air-cooled units, or expensive, water-cooled units.

Lasers provide coherent light (single frequency).

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

What is light scatter?

A

When light hits a cell it is scattered in two directions. Forwards scatter is forwards and proportional to the size of the cell. At 90 degrees is side scatter, which is proportional to the granularity of the cell.

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

Why can we detect the four colours when we add antibodies and fluorochromes?

A

Because there are filters and one detector detects only light from one fluorochrome.

Emission is overlapping - PMT1 will only measure a very narrow wavelength of light, as will 2,3,4 etc

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

What are the electronics of flow cytometry?

A
  • processing of signals from detectors - analogue-digital conversion
  • emitted light is converted into an electronic signal by the detectors. We can capture our data and analyse it on the computer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What is stokes shift?

A

the energy difference between the lowest energy peak of absorbance and the highest energy of emission

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

What does immunofluorescence use?

A

FLUOROCHROMES AND DYES

  • fluorescein isothiocyanate (FITC) - GREEN
  • phycoerythrin (PE) - ORANGE
  • peridinin chlorophyll protein (PerCP) - RED

dozens of flourochromes on the market so lots of options about what laser and what antibodies you want to use.

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

Where can the single cells in suspension in flow cytometry be obtained?

A
  • peripheral blood
  • bone marrow
  • fine needle aspirate
  • fresh tissue
  • CSF and other fluids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What are the two ways of labelling by immunofluorescence?

A

DIRECT: monoclonal antibodies (MoAbs) are pre conjugated to fluorochromes. This is a one-step-process.

INDIRECT: unconjugated monoclonal antibodies. The primary antibody doesn’t have the fluorochrome, but we put a secondary antibody on the antibody which does have the fluorochrome, and it binds to the primary antibody. This is a two step process. This may allow a stronger signal (more fluorochromes). This method may have to be used if you can’t buy or make an antibody with the fluorochrome on it.

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

What is represented by the area under the curve of a histogram?

A

the cells that have the particular fluorescent integrity of whatever parameter is being measured

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

What is gating? (flow cytometry)

A

A more sophisticated method. We can draw regions around the cells we know because of the side scatter on a dot plot and can make the machine only display cells in that gate on the basis of the two fluorochromes, so will be able to see the populations what whichever cell type we decide to look at.

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

How many populations can be identified and quantitated using 3 antibody colours?

A

8

with 2 we can only see 4… but if we used 4 we would see more, etc etc

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

What are univariate cell cycle methods? (flow cytometry)

A

in the simplest method, cellular DNA is detected using a fluorescent dye that binds preferentially to DNA.

Propidium iodide is most commonly used. It undergoes a dramatic increase in fluorescence upon binding DNA. It requires permeabilisation of the plasma membrane.

There are no antibodies or antigens - we are just adding something that binds to DNA. We have to permeabilise the cells to get the propidium iodide in - basically we have to punch holes in the membrane to allow it to enter.

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

About Propidium Iodide (PI)…

A
  • excited at 488nm

- emits in the red

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

Where are the main peaks in the cell cycle under flow cytometry?

A
  • the main peak is G0-G1
  • the following smear is S phase
  • there is another peak when they’re in G2M

there is sometimes a peak at the beginning showing apoptotic cells (or can be argued to just be fragments of DNA)

we can draw regions around these cells and see what proportion of the cells are in these phases

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

How does the assay using propidium iodide work?

A
  • PI can not normally cross the cell membrane, so living cells will look negative on flow cytometry
  • if the PI penetrates the cell membrane, it is assumed to be damaged
  • cells that are brightly fluorescent with PI are damaged or dead, they will look positive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What are detection methods of apoptosis?

A
  • by staining with PI (cells fixed)
  • phosphatidyl serine, can be detected by incubating the cells with fluorescein-labelled Annexin V, and PI - i.e. using two stains together (cells not fixed)
  • by staining with 7-aminoacinomycin D (cells not fixed)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

How does the use of Annexin V and Propidium Iodide work in flow cytometry?

A

Annexin V goes to the phosphatidyl serine. In a live cell it appears totally negative because the PI can’t get in and the annexin V can’t bind as the phosphatidyl serine is all on the inside of the cell.

In early apoptosis the PS is flipped onto the outside of the membrane so annexin V can bind. However, the membrane is still in tact so PI can’t get in, so the cells will appears green but not red.

Later on in apoptosis/necrosis we lose the integrity of the membrane. The annexin can bind (green) and the PI can also get into the cell (red) so these cells will be positive for both green and red.

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

About 7-Aminoactinomycin D (7-AAD)…

A
  • Excited at ~488nm
  • Emits at ~660nm
  • DNA specific - intercalates in G-C regions
  • long emission wavelength, with FITC and PE labelled Ab for simultaneous evaluation of DNA content and 2 colour immunofluorescence using only 488nm ex.

We can label any of these things with fluorochromes and can look at different caspases at the same time. We can measure where the cell is in the apoptotic pathway.

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

What are the applications of flow cytometry?

A
  • immunophenotyping of leukaemia and lymphomas
  • detection of MRD
  • stem cell enumeration
  • CD4/CD8 in HIV
  • measurement of intracellular cytokines
  • study of cell cycle, viability and apoptosis
  • measurement of cell proliferation
  • assessment of transfection efficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

How does cell sorting separate the droplets that we want?

A

When it recognises one of the droplets that fits into the category that we want the machine charges the stream so that the droplet containing the cell that we want breaks off the nozzle from the vibrations, and it is pulled into the plates and separated from the rest of the population.

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

What is an example of success in drug development?

A

LIPITOR: the biggest drug blockbuster in history with sales of $11 billion (before patient expiry)

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

What is an example of failure in drug development?

A

TORCETRAPIB: raised blood cholesterol, strong scientific basis and rationale, but failed clinical testing in 2006 after Pfizer spent $1 billion. Increased mortality rates as compared with placebo.

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

What are the three stages of a drug development project?

A
  1. ) DRUG DISCOVERY: candidate molecules chosen on basis of pharmacological properties
  2. ) PRECLINICAL DEVELOPMENT: non-human studies. Toxicity testing, pharmacokinetic analysis and formulation
  3. ) CLINICAL DEVELOPMENT: volunteers and patients. Efficacy testing, side-effects and potential dangers.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What are examples of drug targets?

A
  • functional proteins
  • receptors
  • enzymes
  • transport proteins

…not all are actually ‘draggable’ however…

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

What are possible non-biological limitations of drug development?

A

cost and complexity of drug discovery and development (regulation)

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

What are methods of lead finding and optimisation?

A
  • cloning of target protein
  • assay to measure functional activity
  • automated systems to allow for speed and economy
  • high-throughput screening of large compound libraries
  • natural products, fungal, plants, bacteria

Lead optimisation, complex chemistry to increase potency, selectivity and stability

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

What happens in the preclinical development of drugs?

A
  • pharmacological testing for hazardous acute effects
  • preliminary toxicology testing
  • pharmacokinetic testing for absorption, metabolism, distribution and elimination
  • chemical and pharmaceutical development to assess feasibility of large-scale synthesis and purification as well as stability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What happens in the clinical development of drugs?

A

volunteers and patients, efficacy testing, side-effects and potential dangers…

PHASE I: safety and tolerability (MTD)
PHASE II: small scale efficacy/placebo controlled
PHASE III: large scale efficacy/randomised and blinded
PHASE IV: post marketing surveillance

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

What is avastin?

A
  • first specific anti-angiogenesis drug
  • in 2013 was the second biggest selling oncology product
  • forecast to be the top selling cancer drug for 2018
  • approved for colorectal, lung, kidney and ovarian cancers and eye disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What is angiogenesis?

A

the formation of new blood vessels form pre-existing vessels

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

What are the functions of angiogenesis?

A
  • blood vessels supply oxygen, nutrients, remove waste and allow immune surveillance
  • physiological angiogenesis is essential for organ growth in the embryo and repair of wounded tissue in the adult
  • abnormal genesis leads to disease –> inefficient vessel growth: stroke, MI, ulcerative disorders and neurodegeneration; excessive vessel growth: cancer, inflammatory disorders, pulmonary hypertension, blindness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What are the three types of angiogenesis?

A
  • developmental angiogenesis/vasculogenesis
  • normal angiogenesis
  • pathological angiogenesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What does developmental angiogenesis/vasculogenesis occur?

A

organ growth

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

Where does normal angiogenesis occur?

A

wound repair, placenta during pregnancy, cycling ovary

190
Q

Where does pathological angiogenesis occur?

A

tumour angiogenesis (gives the tumour nutrients from the blood and allows metastatic spread via the blood), ocular and inflammatory disorders

191
Q

What is the difference between angiogenesis and vasculogenesis?

A

vasculogenesis is defined as the differentiation of precursor cells (angioblasts) into endothelial cells and the de novo formation of primitive vascular network, whereas angiogenesis is defined as the growth of new capillaries from pre-existing blood vessels

192
Q

What is tumour hypoxia?

A

hypoxia is a strong stimulus for tumour angiogenesis.

Hypoxia is low oxygen tension (

193
Q

What is the mechanism of action of avastin?

A
  • monoclonal antibody
  • binds to VEGF
  • prevents VEGF binding to VEGF receptors on endothelial cells
194
Q

What parameters is MRI contrast sensitive to?

A
  • T1 relaxation
  • T2 relaxation
  • Water diffusion (micro)
  • Flow (macro)
  • perfusion
195
Q

What three ways can MRI tell us things?

A
  • anatomical
  • functional
  • response to treatment
196
Q

What can ANATOMICAL MRI show?

A
  • abnormal tissue or vascular structures
  • presence of lesions
  • structural changes (learning, ageing)
197
Q

What can MRI show about FUNCTION?

A
  • blood flow: perfusion, brain activation - motive taste or experiencing pain
  • metabolic (biochemical changes)
198
Q

What can MRI show us about RESPONSE TO TREATMENT?

A
  • lesion change: disappearance, shrinkage, change in appearance
  • functional changes: normal blood flow, activation changes
199
Q

How does the basic NMR instrument work?

A

There is a very strong uniform magnetic field along the bottom of the magnet (whole thing) 30,000x stronger than Earth’s magnetic field.

MR imaging is formed with magnetic field gradients to code the image signal in space.

Linear variations of magnetic field coding the signals so you know where they’re coming from so hat you can see slices instead. You can then build up the image.

MRI scans are good because you can rotate things if the patient is not lying straight etc

200
Q

Where does the magnetisation in tissue come from in MRI?

A

it comes from the protons in water and the fat in tissue

water within hydrogen atoms gives most of the magnetisation. the nucleus is a proton that spins on its own axis - it has a single positive charge. Nuclear Magnetic Resonance can interact with this magnetisation.

201
Q

What series of signal acquisitions is an MRI image built up from?

A
  • Oblique: any slice orientation is possible
  • Multislice: acquisitions on many different slices within the TR
  • 3D: acquisitions from a full 3D volume of tissue
  • Gated: RF pulses triggered to ECG for cardiac imaging
202
Q

What is T2 dependent on and what does it increase with?

A

T2 is very dependent on how mobile the water is in the tissue and increases with:

  • oedema, an increase in the water content
  • demyelination, a loss of brain tissue structure
203
Q

What is T2 reduced by?

A

T2 is reduced by the presence of paramagnetic ions

  • Fe from blood breakdown products
  • Gd from contrast agents
204
Q

What is T1 dependent on?

A

T1 is very dependent on the presence of paramagnetic ions that reduce T2

T1 is also dependent on how mobile the water is in the tissue, and T1 increases slightly with oedema

205
Q

Is T1 lower in white matter or grey matter?

A

lower in white matter because of myelinated neutrons

206
Q

How do contrast agents work?

A
  • water in the vicinity of the contract agent experiences strong fluctuating magnetic fields, hence T1 and T2 are reduced.
  • bind strongly to the Gd - don’t get free Gd ions in the patient. Wherever the Gd is, the T1 and T2 are produced.
207
Q

How does a functional MRI work in a Blood Oxygen Level Dependent (BOLD) contrast?

A
  • gradient echo images (GRE) are very sensitive to the rpesense od non-uniformity of the magnetic field in tissue (T2* relaxation –> T2* decreases as the uniformity decreases)
  • deoxygenated blood contains deoxyhaemoglobin (dHb), which is paramagnetic, and so distorts the magnetic field
  • if blood becomes more deoxygenated, then the dHb levels increase and the T2* decreases
  • In a GRE image, this means that the image gets larger
208
Q

How does the brain respond to a decreased level of oxygen?

A

vasodilation to increase blood flow to areas of the brain require for function (moving, reading, speaking)

209
Q

What is MRS?

A

A 1H spectrum of biochemicals can be obtained from a localised region of the brain using three slice selective pulses

210
Q

What is the basis of the magnetic resonance spectrum?

A

A stronger shielded nucleus has a lower resonant frequency

211
Q

What are the safety issues of MRI/MRS?

A

No ferromagnetic objects in the examination room; scissors, stethoscopes, wheel chairs, gas cylinders - hearing aids, watches, spectacles (dentures - image quality)

212
Q

what are contraindications to the use of MRI/MRS?

A
  • pacemakers
  • first trimeter pregnancy
    aneurysm clips
  • metallic foreign bodies (orbit x-ray, shrapnel)
213
Q

About the sound waves used in ultrasonography…

A
  • mechanical radiation, propagating by vibration of elements (molecules) of a medium
  • manifests as longitudinal pressure (compression) wave and transverse (shear) wave
  • sound production requires a vibrating source

Sound is a physical wave that requires a medium to propagate through.

214
Q

What is the different between longitudinal and transverse waves?

A

Longitudinal: waves same way as direction
Transverse: waves perpendicular to direction

215
Q

What are we only interested in the megahertz range of sound in ultrasonography?

A

because we need very short wavelengths to be able to resolve structures in the body that are very close together

216
Q

What material properties determine the speed of a sound wave?

A

density and bulk modulus (stiffness)

the density of the material and how rigid it is determines the speed. If a material is squishy it slows down the speed, and conversely dense materials will increase the speed of sound.

the waves travel a bit slower in fat, very much slower in air, and much faster in bone

217
Q

What types of interactions are there of ultrasound in tissue?

A
  • ABSORPTION: ultrasound is converted into heat by friction
  • REFLECTION: some energy reflected from a boundary
  • INTERFERENCE
  • REFRACTION: some or all of the energy is diverted from its original path when the beam hits the boundary away form the normal, this can produce artefacts in the image
  • DIFFRACTION: divergence of beam from sound source when it goes through a narrow gap. this can give us less focus or no information at all
  • SCATTER: some of the energy is dispersed in all directions. This causes the grainy texture of the images produced, this is also an artefact
218
Q

What is absorption of ultrasound waves dependent on?

A

the tissue properties, frequency and the distance travelled.

The higher the frequency, the more you are vibrating the molecules so you will lose more energy. For deeper structures we have to alter features to get enough echo strength coming back to be able to see something.

We use lower frequencies to see deep structures for this reason, but can use higher frequencies when looking more superficially.

219
Q

Where do reflections occur in ultrasonography?

A

At tissue boundaries where there is a change in acoustic impedance (Z)/ a tissue interface

the amplitude (or intensity) of the reflected wave depends on the difference in the acoustic impedances (reflection coefficient) - the greater the difference, the stronger it will get reflected back

220
Q

What are specular reflectors?

A

Returns echoes to the transducer only when the sound beam is perpendicular to the interface.

If you hit the surface at 90 degrees you get a crisp white bright line in the image. When you hit it away from the normal, it will bounce back at an angle and you will lose some of this clarity

221
Q

how does interference occur in ultrasonography?

A

she of the little wavefronts coming off the scatter can summate together and line up which will increase their intensity - this is constructive interference

if they don’t line up you will get destructive interference as they cancel one another out.

These effects together give the grainy appearance - it is totally meaningless and is nothing to do with the texture.

222
Q

What is Attenuation in ultrasonography?

A

the loss of power or amplitude the ultrasound signal as it passes through tissue

ATTENUATION = ABSORPTION + SCATTER + REFLECTION

usually attenuation is nearly proportional to frequency - attenuation increases with frequency: absorption factor.

223
Q

What are the basic acoustic features of an ultrasound machine?

A
  • TRANSMITTER
  • TRANSDUCER
  • RECEIVER, ADC AND PROCESSOR
  • IMAGE DISPLAY
224
Q

What does the TRANSMITTER do in US?

A

to energise the transducer (PRF). Issues pulses all the time, like a clock, everything is related back to the transmitter for timings of echoes coming back etc

225
Q

What does the TRANSDUCER do in US?

A

piezoelectric ceramics convert the electrical energy into mechanical vibrations. The ultrasound probe, the piezoelectric material which makes the ultrasound and directs the echoes coming back.

Made of ~250 elements of varying materials - often silicon or a composite material. They are all individually wired up to individual electrodes.

226
Q

What does the RECIEVER, ADC and PROCESSOR do in US?

A

to detect and amplify backscattered energy, i.e. detect what is noise and can be discounted

227
Q

What does the IMAGE DISPLAY do in US?

A

a flat screen device which present the ultrasound image

228
Q

What is the matching layer of the transducer for in ultrasonography?

A

it is there to allow as much of the sound through into the body as possible. If it was not there and we put the transducer straight onto the skin then there would be a big difference and we would get a lot of reflection; the matching layer is a pathways between the elements and the body

229
Q

What is the backing material on the transducer for in ultrasonography?

A

it is a damping element to stop echoes coming out of the back of the probe and bouncing around which would cause confusion. Sound coming back will hit it and cause it to vibrate gently, and produce avery small electric current - a piezoelectric element.

230
Q

Do we use continuous or pulsed waves for US imaging?

A

Continuous

231
Q

What is B-mode us ultrasonography?

A

B-mode is ‘brightness mode’ - this gives us little dots on the display instead of a line. the brightness shows the strength of the echo and further down the image means it took longer to get back - i.e. placement on the image shows the time it took to get back. These dots gradually build up over time

232
Q

The basics of B-mode imaging (ultrasonography)…

A
  • large number of pulse echo lines
  • each echo is displayed along a line as a dot
  • the brightness of each dot is determined by the strength of the echo
  • the distance down the display relates to its depth below the transducer
233
Q

What does electronic focussing of ultrasonography mean?

A

This means that the operator has control.

When the elements fire, its not just one but normally a group of them that operate. If we adjust the timing, the outer ones fire a bit earlier, then the inner one.

The wave fronts interfere with each other and focus in a tissue - this allows you to focus in the area of interest.

Varying delay times enables focussing at different depths.

234
Q

What is time gain compensation (TGC)?

A

echoes coming from deeper structures are weaker and must be amplified to produce uniform tissue echo appearance.

this is manually controlled and has a profound effect on the quality of the ultrasound image presented for interpretation.

they go through standard linear amplification systems

the echoes coming back from further in the body will be much weaker and so the machine has to compensate for this or the images will get darker and darker until it is black.

the echoes that take longer to come back are amplified a bit more to compensate for that loss - “time gain amplification system”. The operator can alter that using the slides on the machine and this will change how much amplification occurs.

235
Q

How can we avoid causing artefacts in gain and time-gain compensation?

A

we can just alter the front or the back of the image, basically to make the image look more aesthetically pleasing

236
Q

What is the doppler effect in ultrasonography?

A

we can look at tissue movement by bouncing the US off RBCs and detect their forward movement - we do this by looking at the Doppler Shift Effect.

If we have a target that isn’t moving and hit it with ultrasound waves, then its reflection will have the same frequency.

If we have a target that is moving towards us, then we will get a compression of the wavefront and the frequency is higher in the reflection.

If it is moving away then the wavefront will get stretched out and the frequency will be lower in the reflection.

US machines use this to analyse blood flow as the sound bounces off RBCs. We can also get a good estimation of its speed using this technique and a complicated equation.

237
Q

What is colour doppler imaging of ultrasonography?

A

RED: anything flowing towards the probe face
BLUE: anything moving away from the probe face

there is a scale at the side with the hue of the colours giving us an idea of the speed of the sound.

238
Q

What is ‘spectral trace’ doppler imaging in ultrasonography?

A

a measurement of speed, gained by using a spectral gate, which you place over the blood vessel, which you flick into spectral doppler mode and get detailed analysis of it flowing through the gap.

this will give measurements of peak velocity, systolic velocity and average velocity.

this method is a lot less invasive as we don’t have to inject anything but we still get a lot of information

239
Q

Info on COLOUR Doppler…

A
  • information on speed and direction of blood flow
  • image noise results in random flashes of colour
  • angle dependent (no flow shown at 90 degrees)
240
Q

Info on POWER Doppler…

A
  • only detects blood flow, but more sensitive to low volume flow
  • angle independent
  • better boundary detection
  • very sensitive to soft tissue motion
241
Q

What are the two standard scanning planes?

A
TRANSVERSE SECTION (TS): right side of patient on left side of image
LONGITUDINGAL SECTION (LS): cranial aspect of the patient on the left of the image

this is the reverse for cardiologists/echocardiography

242
Q

What are the advantages of using ultrasound?

A
  • real time guidance
  • multiplayer
  • visualisation of blood flow
  • speed
  • portable
  • cost
  • non-ionising radiation
243
Q

What are the disadvantages of using ultrasound?

A
  • sound penetration (bone and gas)
  • operator dependence - only one person doing the images and they give the definitive opinion which is often a diagnosis
  • patient size and very gassy patients - worst imaging, 100% deflection from air, fat slows down waves
  • artefacts
  • have to be careful with early pregnancy as the warming effect can be measurable in a foetus
244
Q

What is acoustic enhancement in ultrasound?

A

When a region has a lower attenuation coefficient than adjacent tissues the machine overcompensates. This provides a clue to tissue composition.

Echoes coming back are stronger and look brighter, and this can be a useful artefact in the cases of liver cysts for example.

245
Q

What various areas can ultrasound be used?

A
  • ABDOMINAL IMAGING
  • PELVIS (GYNAECOLOGY AND PROSTATE)
  • SMALL PARTS
  • OBSTETRICS
  • VASCULAR
  • PAEDIATRICS
  • CHEST
  • BRAIN
  • CARDIOLOGY
  • ENDOSCOPIC/INTRAVASCULAR
  • INTRAOPERATIVE/LAPROSCOPIC
246
Q

What is interventional ultrasound?

A
  • real time and portable - offers advantage over the image guidance
  • biopsies, FNA, drainage procedures, vascular access
  • guidance for ablation procedures (e.g. RFA)
  • sterile probe covers required
247
Q

What are microbubbles in ultrasound?

A
  • Diameter 1-7 micrometers (able to cross pulmonary circulation)
  • gas filled
  • cross capillary beds
  • vascular markers
  • duration: vascular phase 5-10 minutes
  • resonant frequency 2-5 MHz
  • very safe
248
Q

What is freehand elasticity imaging (ultrasound)?

A

Ultrasound transducer is used to compress tissue.

The compression is simultaneously imaged and the images are further processed to generate stiffness images - “elastograms”

249
Q

What are clinical applications of sonoelasticity?

A
  • BREAST: improve differentiation between benign and malignant disease, assessment of appropriate therapeutic treatment
  • THYROID: provides additional features of malignancy, targeting biopsy of complex lesion
  • PROSTATE: to improve visualisation of prostate cancers, targeting biopsy
  • PANCREAS: to differentiate between benign and malignant tumours
  • LIVER: (WIP) to observe progressive degree of liver fibrosis
250
Q

What are the advantages of 3D/4D ultrasound?

A
  • volume measurement
  • viewing the orthological plane
  • assessment of surface and complex shapes
  • separation of B-mode data from colour
251
Q

What are the disadvantages of 3D/4D ultrasound?

A
  • acquisition
  • movement artefacts
  • artefacts in general
252
Q

What is multiplanar reformatting (MPR)?

A

produces images in the 3 orthogonal planes. Can be rotated or scrolled through. Power doppler information can be combined and on 4D systems this can be viewed in real time.

253
Q

Why is there an increased interest in ultrasonography?

A
  • increased demand for imaging
  • variable want for ultrasound service and out of hours access
  • affordable, high quality, portable machines
  • move towards point-of-card diagnostics
  • improved patient pathways/service improvement
  • guidelines advocating the use of ultrasound in procedures
254
Q

What is the FAST scan (ultrasound)?

A
  • performed in A+E for detection of haemoperitoneum
  • scan for free fluid
  • sensitivity 94% and specificity 98%
  • in the low risk patient a negative ultrasound has a very negative predictive value
  • recognises increased demand for ultrasound
  • training to be adequately funded
  • training to consist of a combination of practical and theoretical
  • standard of training to be equal to that of a radiographer
  • foster relationships between radiological and non-radiological practitioners
255
Q

How many levels of training in ultrasound are there?

A

3

256
Q

What are the standards and QA set out in ultrasonography?

A
  • evidence of scanning protocols
  • minimum scan members (20 per year at level 1)
  • minimum equipment standard
  • regular equipment maintenance
  • policy on infection control
  • policy on image recording
  • reporting of images BFCR (06) 1
  • incident reporting
257
Q

What is the function of neurones?

A

generations of action potentials, synaptic communication

258
Q

How does Na+/K+ ATPase generate electrical activity?

A

Na+/K+ ATPase produces an unequal distribution of Na+ and K+ ions to reduce osmotic pressure and prevent cell lysis.

This leads to creation of electrochemical gradients for Na+ and K+.

259
Q

What main principle is electrophysiology based on?

A

Ohm’s Law: Voltage (V) = Current (I) x Resistance (R)

Another way of describing resistance is as a reciprocal of conductance (Siemens,G1) R=1/G

The less resistance to the flow of current between the pd of two points, the more conductance there is…
(V=IR, rearrange to V=1/G) = Ohm’s law = I = GV

260
Q

What are the three main micro electrode techniques?

A
  • extracellular recording
  • intracellular recording
  • voltage-clamp recording

all measure activity of many/different types of ion channels - macroscopic recordings

261
Q

How is extracellular recording done?

A

two micro electrodes are places outside cells. Records cell-induced membrane potential changes between two electrodes e.g. APs. Single cells: single-unit recording. Group of cells: multi-unit recording

262
Q

What are uses of extracellular recording?

A

Record electrical activity in anaesthetised/conscious animals, e.g. Hubel and Wiesel showed specific neurones in visual cortex responded to different visual stimuli

263
Q

How is intracellular recording done?

A

One micro electrode is places inside, the other ground electrode is placed outside the cell. Records membrane potential across the membrane of single cell - allow manipulation of external solutions (e.g. control electrolytes, apply drugs). This does not alter intracellular composition of cells.

264
Q

What are uses of extracellular recording?

A
  • Record electrical responses to external stimuli e.g. neurotransmitters
  • measure changes in conductance - opening/closing of ion channels
265
Q

How does voltage-clamp recording work?

A
  • one microelectrode measures voltage - which is ‘clamped’
  • one microelectrode applies current to and from the cell
  • uses a ‘clever’ feedback amplifier - measure V and I at the same time

The power of the voltage-clamp is to measure membrane current produced by opening of ion channels at a specific membrane potential

266
Q

Why is the giga-ohm seal important in the voltage-clamp technique?

A
  • reduced background noise - can record very small currents (
267
Q

What configurations of the gig-ohm seal are there (patch configurations)?

A
  • cell-attached patch
  • inside out patch
  • whole-cell recording
  • outside-out patch
268
Q

What are the uses and problems of the cell-attached patch?

A

USES:

  • agonists applied to pipette solution
  • eg ligand gated (eg nicotinic receptors)
  • voltage steps applied through the pipette
  • eg voltage gated (Na+ channels)
  • agonists applied to extracellular solution (non-pipette solution)
  • eg GPCRs (muscarinic receptors)

PROBLEMS:

  • resting membrane potential cannot be controlled
  • can not control the composition of the intracellular medium
269
Q

What are the uses and problems of the inside-out patch?

A

USES:

  • agonists can be applied directly to the cytoplasmic surface of the cell membrane
  • eg activation mechanisms of 2nd messenger systems Ca2+, DAG, IP3, protein kinases (PKC, PKA)
  • control composition of both extracellular and intracellular media

PROBLEMS:
- very reductionist approach - will the channels behave normally?

270
Q

What are the uses and problems of whole-cell recording?

A

USES:

  • agonists applied to pipette solution
  • eg 2nd messenger gated
  • agonists applied to extracellular solution
  • eg ligand-gated/GPCR
  • voltage steps applied through the pipette
  • eg voltage-gated channels (Ca2+ channels)
  • control composition of both extracellular/intracellular media

PROBLEMS:

  • measures only macroscopic current - not individual channel openings
  • important intracellular components may dialysis out of the cell
271
Q

What are the uses and problems of the outside-out patch?

A

USES:

  • unlike whole cell recording can measure single channel openings
  • agonists can be applied to pipette solution
  • eg ligand gated/GPCR
  • voltage steps applied through the pipette
  • eg voltage-gated channels (Ca2+ channels)
  • control composition of both extracellular/intracellular media

PROBLEMS:
- important intracellular components may be dialysed out of the cell

272
Q

What is the equation for blood pressure?

A

BP = CO x TPR

273
Q

What is the equation for blood flow?

A

blood flow = pressure gradient/vascular resistance

274
Q

What is vascular resistance?

A

Vascular resistance relies on vascular tone. It regulates diameter and resistance.

275
Q

What is the structure of the vascular wall?

A
  • endothelium
  • smooth muscle - these are the muscles that contract, giving the vascular tone
  • perivascular nerves
  • elastic and fibrous tissues
276
Q

How is vessel diameter/tone controlled?

A

If the fibres contract they will narrow the vessel and vice versa. They lie perpendicular to the blood flow. This is what constitutes vasoconstriction and vasodilation.

277
Q

What is the law that relates to vascular resistance?

A

resistance is INVERSELY proportional to the 4th power of the radius (r) of a vessel, and proportional to viscosity of blood (curly n) and the length of the vessel (L)

POISEULLE’S LAW

278
Q

What is isometric tension recording?

A

measure tension generated by the vascular wall while the diameter of the vessel remains constant

can be done in large organ baths or wire myograph

widely used in vitro techniques to investigate the functional properties of isolated vessel segments

279
Q

How is isometric tension recording done in large water baths?

A
  • for large vessels (internal diameter > ~1.5mm)
  • segments are mounted as ring preparations on two fixed steel hooks
  • kept in a chamber with physiological salt solution at 37*C with oxygen
  • they are viable for >6 hours
280
Q

How is isometric tension recording done by wire myograph?

A
  • for small vessels (internal diameter ~100-400 micrometers)
  • segments are mounted as ring preparations on two fixed steel wires
  • kept in a chamber with physiological salt solution at 37*C with O2
  • viable >12 hours
  • smaller arteries play a bigger role in vascular resistance

–> DISSECTION –> MOUNTING –>

281
Q

What is EC50?

A

the concentration at 50% of the maximum response

282
Q

What are the advantages of isometric tension recording?

A
  • examine functional properties of small or large vessels
  • freshly prepared vessels are viable for many hours
  • detailed investigation into the mechanisms of action
  • compare vascular function in control vs diseased states
  • can be combine swift other experimental techniques
283
Q

What are the disadvantages of isometric tension recording?

A
  • dissection and mounting of vessels requires skills and suitable apparatus
  • isolation of vessels may remove influences from surrounding tissues
  • difficult to study longer term changes in vascular function using the same vessels
  • cannot measure myogenic contractions
  • cannot measure flow induced relaxations
284
Q

What is a pressure myograph?

A
  • for small vessels including arterioles (>50 micrometers)
  • segments mounted on 2 glass microcannulae and pressurised to an appropriate transmural pressure
  • kept in a chamber with physiological salt solution at 37*C with oxygen
285
Q

What is the myogenic response?

A
  • an intrinsic property of vessels (auto regulation)
  • stretch of smooth muscle causes vasoconstriction
  • stabilises blood flow despite increase in BP (between 40-100mmHg)
  • contributes to basal vascular tone

Shear stress = frictional force on the endothelium

286
Q

What is flow induced vasodilation?

A
  • shear stress on endothelial calls leads to vasodilation

- impaired in cardiovascular diseases including atherosclerosis and ageing

287
Q

What are the advantages of isobaric diameter recording?

A
  • examine functional properties of small vessels, including myogenic constriction of flow-induced dilation
  • freshly prepared vessels are viable for many hours
  • detailed investigation into the mechanisms of action
  • compare vascular function in control vs diseased states
  • can be combined with other experimental techniques
288
Q

What are the disadvantages of isobaric diameter recording?

A
  • dissection and mounting of vessels requires skills and suitable apparatus
  • isolation of vessels may remove influences from surrounding tissues
  • difficult to study longer-term changes in vascular function using the same vessels
289
Q

What are isolated vascular beds?

A
  • cannulate supply artery to a vascular bed
  • e.g. measure perfusion pressure under constant flow rate
  • flow = perfusion pressure/vascular resistance
290
Q

What is the Langendorff Heart Preparation?

A
  • cannulate the ascending aorta (retrograde perfusion)
  • measure coronary perfusion pressure under constant flow rate
  • viable for a few hours
  • can also examine left ventricular contractions and heart rate (if eft beating naturally)
291
Q

What are the differences between in vitro vs in vivo cardiovascular techniques?

A

IN VITRO

  • isolated small vessels
  • isolated vascular beds

IN VIVO
- ultimate preservation of surrounding tissues and regulation through hormones and innervation?

292
Q

What are the advantages of doppler flowmetry?

A
  • examine changes in blood flow in vivo (natural surroundings are largely preserved)
  • compare vascular function in control vs disease states
  • can be adopted for non-invasive measurements
  • allow long-term measurement of blood flow in the same vessels
293
Q

What are the disadvantages of doppler flowmetry?

A
  • relative measurements of blood flow
  • quality of signal (signal:noise ratio) depends on tissues and machines
  • invasive procedures and anaesthesia may be necessary
  • expertise requires for experimentation, data analysis and interpretation
294
Q

What are examples of major psychoses?

A
  • schizophrenia
  • schizoaffective disorder = schizophrenia and bipolar disorder
  • delusional disorder
  • some depressive and manic illnesses
295
Q

Why is schizophrenia an important disease to study?

A
  • early onset
  • prevalent
  • disabling and chronic
296
Q

What is the prevalence of schizophrenia?

A
  • affects up to 1% of the population
  • no significant influence of culture, ethnicity, background, socioeconomic groups
  • increased in urban areas
  • no different between men and women (men 1-25 years and poorer response to therapy, whereas women 20-30 years)
297
Q

What are early symptoms of schizophrenia?

A

Before the disease is recognised there is often a phase in late teenage years associated with social isolation, interest in fringe cults and social withdrawal

298
Q

What is the classification of schizophrenia?

A

For one to be schizophrenic, 2+ of the following must be present for a significant period of time:

  1. delusions
  2. hallucinations
  3. disorganised speech, emotional blunting and meaningless behaviour
  4. grossly disorganised or catatonic behaviour (e.g. sitting still for ages/abnormality of posture)
  5. negative symptoms (eg affective flattening)
299
Q

What are the criteria for diagnosis of schizophrenia?

A
  • must have positive and negative symptoms
  • gradual or sustained deterioration of work, interpersonal relationships, communication and self care
  • duration of symptoms has to be at least 6 months of disturbances
  • exclude substance abuse/general medical conditions
  • relationship to pervasive development disorder
300
Q

Does schizophrenia follow a classic mendelian model?

A

no but there is some familial correlation e.g. twin studies and parents etc

301
Q

What environmental factors may predispose someone to schizophrenia?

A
  • born in winter months increased risk - some form of damage to the child, e.g. damage to corticolimbic system
  • viral epidemics - development in vitro
  • obstetric complications (oxygen deprivation)
302
Q

What is schizotaxia?

A

genetic makeup deemed to be necessary for the development and manifestation of schizophrenia, along with extreme stresses within the surroundings

303
Q

What is a prodrome?

A

an early symptom indicating the onset of a disease or illness

304
Q

What are general symptoms of schizophrenia?

A
  1. abnormal ideas
  2. abnormal perception
  3. motor, volitional and behavioural disorders
  4. formal though disorders
  5. emotional disorders
  6. psychophysiological changes
305
Q

About motor, volitional and behavioural disorders…

A

peculiar forms of motility, stupor, mutism, stereotypy, mannerism, negativism, spontaneous automatism, impulsivity

  • stereotypes: purposeless, repetitive acts
  • bizarre postures; strange mannerisms
  • altered expression - grimacing

states of catatonia or bouts of extreme hyperactivity
impulsive behaviour e.g. violent acts and reasonless murder

306
Q

What is a formal thought disorder?

A
  • disturbances in thinking - intellectual speech
  • derailment of speech
  • loosening of associations; failure to follow train of thought to its conclusion
  • poverty of speech - manifests as distorted or illogical speech
307
Q

What is an emotional disorder?

A
  • loss of natural reaction to social cues (affective flattening)
  • reduced facial expression
  • apparent indifference to emotive topics
  • cold and detached
  • cannot establish a rapport in social dialogue
  • inappropriate affect - shallowness of emotions (e.g. laugh at death)
308
Q

What are the two types of psychophysiological changes in schizophrenia?

A

EVENT-RELATED POTENTIAL (ERP) ABNORMALITIES

  • deficit in processing of sensory information associated with medial temporal lobe dysfunction
  • a reproducible finding in schizophrenics

EYE TRACKING CHANGES OBSERVED IN 50% OF SCHIZOPHRENICS

  • dysfunction in connections between frontal eye fields and the temporal and parietal cortices
  • a robust phenotype marker of familial risk of schizophrenia
309
Q

What are the positive signs of schizophrenia?

A
  • delusions of perception and loss of control of own thoughts and actions
  • bodily sensations due to outside forces
  • alienated or disorientated thought
  • sudden fully formed delusion in the wake of normal perception
  • thoughts make no sense to normal people
  • hallucinations
  • voices talking about the person
  • bizarre behaviour
310
Q

What are the negative symptoms of schizophrenia?

A
  • loss of empathy with/in others
  • inappropriate or blunted mood; repetitive activity
  • incongruity of emotions (blunted emotions)
  • social withdrawal
  • inability to experience pleasure
  • poverty/paucity of speech
  • attention deficit
  • difficulty planning
  • impaired judgement

NEGATIVE SYMPTOMS ARE REFRACTORY TO DRUG TREATMENT

311
Q

What structural correlates does the neurobiology of schizophrenia cause?

A
  • deficits of neuronal matter: smaller brains and longer ventricles particularly in medial temporal lobe and left hemisphere
  • alterations in thalamus and basal ganglia - functional analysis of brain: electroencephalogram abnormality reported in 80% of schizophrenics, but the changes are non-specific
312
Q

What is hypofrontality (in schizophrenia)?

A

decreased blood flow to the prefrontal cortex

313
Q

What is the corticolimbic system responsible for?

A

memory, emotion and behaviour

314
Q

What brain areas are affected in schizophrenia?

A
  • limbic system
  • prefrontal cortex
  • prefrontal cortex

MRI scans show dramatic loss of grey matter (parietal, temporal, frontal cortices) during the evolution of schizophrenia

315
Q

What is the limbic system?

A

subcortical forebrain structures (hypothalamus, hippocampus and amygdala) - influences emotion and motivation

316
Q

What are the consequences of damaged prefrontal cortex? (e.g. phineas gage)

A
  • altered personality
  • impatient
  • impulsive
  • unpredictable
  • undecided
  • tendency to indulge in profanity (at odds with previous behaviour)
  • decreased ability to alter behaviour on basis of experience
  • attention is prone to distraction

prefrontal cortex is important in cognitive function and emotional expression in survival.

damage or infection of temporal love structures can cause psychotic symptoms.

317
Q

What is the difference between ‘drug-free’ and ‘drug-naive’ studies?

A

drug-free: stopped for a while so its not in their system any more
drug-naive: never taken the drug before

318
Q

What neurotransmitters are involved in schizophrenia?

A

DOPAMINE: a key mediator
CHLORPROMAZINE: dopamine receptor agonist, provided first significant improvement in schizophrenics - symptomatic relief
LYSERGIC ACID DIETHYLAMIDE (LSD): acts on several 5-HT receptor subtypes

Hyperactivity in the mesolimbic dopaminergic system and hyperactivity in mesocortiyal dopaminergic projections.

319
Q

What is the Dopamine theory?

A
  • infers that hyperdopinergic activity leads to psychosis
  • dopamine receptor agonists(eg amphetamines) can induce psychoses - especially in those who are already predisposed
  • there is a correlation between D2 receptor affinity (Ki) and clinical antipsychotic efficacy
  • structural deficits in the brain suggest a loss of control of dopaminergic function
  • multiple studies have shown an increased dopamine release in striatum (SCZ > controls) after amphetamine treatment consistent with increased phasic dopaminergic activity
  • consistent evidence of abnormal D2 receptor number or affinity is lacking
320
Q

What else may play a role in the disease process other than the Dopamine Theory?

A

5-HT, GABA and glutamate may also play a role in the disease process.
BUT most of the neuroleptics interfere with dopaminergic transmission.

Dopamine receptors: D1-5

321
Q

Whites the serotonergic link to psychosis?

A

There is a link from LSD to psychosis.

Phenylcyclidine (PCP/Angel Dust), antagonist at NMD (N-methyl D-aspartate) glutamate receptor type

  • use is associated with psychotic state
  • paranoia
  • agitation, hallucinations
  • social withdrawal
  • reduced emotional expression

PSP (and ketamine) can precipitate psychotic episodes in susceptible patients. PSP use suggests that deficits in glutaminergic transmission may be associated with schizophrenia.

Postmortem on brains of schizophrenics show loss of neurones in mediodorsal thalamus.

322
Q

What is dual cortical deficit?

A
  1. loss of thalamic axon terminals
  2. loss of dopaminergic innervations of prefrontal cortex
  • impairment of GABAergic cortical transmission
  • reports of significant alterations in subpopulation of cortical GABAergic cells in schizophrenics

involves many NTs:
dopamine, 5-HT, glutamate, GABA, CCK, somatostatin

323
Q

What are the aims of therapy of schizophrenia?

A
  • inhibit most florid and subjective behavioural disturbances of psychosis
  • restore patient to normal life in society
  • some agents diminish the negative symptoms of schizophrenia (amotivation, flattened effect and social withdrawal)

Amotivation:incapacity to perceive one’s behaviour and its outcome

324
Q

What are neuroleptic drugs?

A

There are two classes: typical and atypical.

Atypical neuroleptics cause less motor disturbances. There is a strong correlation between the potency of the drugs and their affinity for D2 receptors in the CNS.

The agents bring about improvement in acute positive symptoms of the disorder. Reduce disturbances of delusional thinking, hallucinations, inappropriate saviour and anxiety –> the drugs take time (days or weeks) to work

325
Q

What is the significance of neuroleptic drugs taking time to work?

A
  • you get secondary effects which may be important for therapeutic effects
  • a range of pharmacological effects are associated with the agents
326
Q

What are typical antipsychotics?

A

PHENOTHIAZINES eg chlopromazepine, thioridazine, fluphenazine
D2, alpha1, M receptor agonists, H1 blockage - causes sedation

BUTYROPHENONES eg haloperidol
D2, alpha1 receptor antagonist

THIOXANTHINES eg flupentixol
less sedating; EPS

The atypical (novel) neuroleptics differ from each other and typical neuroleptics in their pattern of receptor binding
eg respiradone, aripiprazole
327
Q

What are side effects of clozapine?

A

weight gain, aesthetic jaundice, etc

needs to be administered in a hospital setting

as it causes sedation is also interferes with H1 receptors to some extent

328
Q

What are side effects of typical antipsychotic drugs?

A

idiosyncratic reactions, anticholinergic reactions, antihistaminergic reactions, antiagrenergic, increased prolactin secretion - impotence etc, miscellaneous reactions (rashes, liver and bone marrow toxicity)

mainly due to the mode of action of the drugs, but can be unrelated –> dopaminergic pathway, blockade of muscarinic, alpha-adrenergic and histamine receptors

some drugs have individual side effects. Some work on a variety of receptors - they are ‘promiscuous’

329
Q

What are extrapyramidal side effects (EPS)?

A

parkinson’s like symptoms…

rigidity, tremor, restlessness, slowing of voluntary movements, postural instability, dystonia

330
Q

What is tardive dyskinesia?

A
  • uncontrollable involuntary movements of face and limbs

- benzodiazepines and beta-blockers may be helpful

331
Q

What are the 4 dopamine pathways?

A
  1. ) mesolimbic (SCZ - increased DA causes positive symptoms)
  2. ) mesocortiyal (SCZ - DA hypoactivity: negative and cognitive and affective symptoms
  3. ) nigrostriatal (drugs - EPS and TD drug side effects)
  4. ) tuberohypophyseal (Drugs - hyperprolactihemia side effects, D2 antagonists reduce activity in this pathway therefore prolactin levels rise)
332
Q

What is the striatum?

A

A subcortical part of the forebrain that helps coordinate body movements associated with behaviour and motivation. Loss of dopaminergic innervation to the striatum is involved in Parkinson’s disease.

333
Q

How is the tuberohypophyseal dopamine pathways related to D2 antagonists?

A

normal secretion acts to block prolactin to a certain extent. typical neuroleptics interfere with the pathway, producing elevated prolactin levels as side effects

334
Q

What order is the electromagnetic spectrum?

A

R.M.I.V.U.X.G

335
Q

What is ionising radiation?

A
  • radiation that causes ionisation when it interacts with matter
  • types used for medical imaging are gamma rays and x-rays
  • we use ionising radiation because it is PENETRATING
336
Q

What is the direct action of ionising radiation?

A

This is when the radiation is incident upon the DNA and can cause damage at that level.

Direct effects occur once a threshold level of damage has been reached.

337
Q

What is the indirect action of ionising radiation?

A

Because most of the body is water, the indirect action is most predominant. The radiation is ionising so it splits the water into ions. They can also combine to form H2O2 which is highly reactive and can go on to interact with other molecules in the body and cause damage

338
Q

What damage can ionising radiation cause on cells?

A

The cell could mutate to such a level that the cell dies, could lose control of its growth function (cancer) or could be affected at the DNA level and cause genetic changes.

339
Q

What are direct effects of ionising radiation?

A
  • only at higher radiation dose
  • threshold effect
  • e.g. Erythema and hair loss
340
Q

What are indirect effects of ionising radiation?

A
  • risk of cancer induction
  • risk of genetic change in subsequent population
  • effect proportional to radiation dose - no threshold
  • ALL RADIATION HAS RISK
341
Q

What particles are involved in radionuclide therapy?

A

alpha particles: 2 neutrons, 2 protons (positive)
beta particles: electrons (negative)

emitted following the radioactive decay of an unstable nucleus

342
Q

What particles are involved in PET scanning?

A

positrons: the electrons interact with matter to create gamma rays

emitted following the radioactive decay of an unstable nucleus

343
Q

What particles/radiation is involved in gamma-camera imaging (e.g. SPECT)?

A

gamma rays: penetrating radiation

emitted following the radioactive decay of an unstable nucleus

344
Q

What particles/radiation is involved in X-ray imaging?

A

x-rays: spectrum of electromagnetic radiation

artificially produced in an x-ray tube

345
Q

What is the attenuation of x-rays?

A

X-rays are essentially an attenuation map - the image appears darker in areas where more x-rays penetrate the film.

Attenuation increases with…

  • higher atomic number
  • higher density

bone has a high atomic number and high density so the beam will be highly attenuated, meaning it stops some of the beam transmitting through it so we get quite allow x-ray signal.

muscle is less dense with a lower atomic number, and the lung is just air with a bit of tissue so very low attenuation.

346
Q

What is TRANSMISSION imaging?

A
  • radiation directed through the patient
  • transmission map collected is essentially an attenuation map
  • good at showing structure, especially between tissues of different densities and atomic number
347
Q

What is EMISSION imaging?

A
  • the radiation is administered to a patient in the form of a tracer
  • emitted radiation is detected outside the patient

patient is injected with a radio-pharmaceutical, putting the radiation inside the patient, and then detect the radiation/gamma rays which are emitted FROM the patient and then looking at the distribution of that emission.

348
Q

How does the X-ray tube work?

A

X-ray tube is within a vacuum. We have a positively charged target. The target and the filament are separated by a potential difference. Heating the circuit controls the voltage.

We have control over the amount of x-rays produced and their energy.

the current controls the amount of electrons, and so, therefore, the x-rays produced. Control of the energy of the x-rays produced is done by varying the voltage across - it accelerates the electrons so that when they interact with the positive charged target, the interaction produces x-rays.

349
Q

What kind of detectors are there for planar x-ray?

A

FILM HARDCOPY: film processor with tanks of chemicals, high resolution

COMPUTED RADIOLOGY COMPUTER SCANNER: phosphor plate, special laser scanner of CR reader that reads and digitises the image, digital enhancement and archiving

DIGITAL RADIOLOGY: flat panel detector, fully digitised system

350
Q

Why is a compression plate used in mammography?

A

Used to reduce the breast thickness, improves resolution and lowers radiation dose.

351
Q

What is fluoroscopy (real time x-ray)?

A
  • real time imaging
  • a catheter is fed inside an artery and radio opaque dye is injected
  • show blood flow inside vessels and can be used to assist with interventions
  • interventional
  • want to see if the surgeon is guiding it through the right place
352
Q

How is x-ray intervention used in coronary angiography?

A
  • real time imaging using an image intensifier called fluoroscopy
  • a cardiac catheter is fed inside the aorta
  • radio-opaque contrast agent used to identify areas of occlusion
  • treatment may be either balloon angioplasty or insertion of a stent
353
Q

What are the limitations of planar x-ray?

A
  • cannot distinguish between overlying tissues
  • tissues other than those being observed reduce the contrast in the image
  • historically partially solved by moving the film cassette and x-ray relative to the patient to blur out overlying tissues, called ‘tomography’
  • superseded by Compound Axial Tomography, now abbreviated to CT.
354
Q

How does a CT scanner work?

A

the patient is on a bed and is moved into the scanner. The scanner has an x-ray tube and a detector which rotates round the patient the bed moves out.

355
Q

What is helical scanning?

A
  • AKA spiral scanning
  • introduced in the late 1980s
  • continuous rotation
  • continuous table feed
  • the patient moves through as the scanner rotates
356
Q

What is helical MSCT?

A
  • multislice
  • faster scan
  • more coverage each rotation
  • modern variations operate in helical MSCT - multiple slice CT
  • can cover more of a patient per rotation of the CT tube so much faster scanning technique
357
Q

How can CT scans be used in looking at disease progression?

A
  • measurement of the size of the left inguinal lymph nose shows progression of disease
  • imaging is used for monitoring response to therapy
358
Q

How can CT scans be used in treatment planning?

A
  • external beam radiotherapy irradiates normal tissue as well as tumour
  • multiple beams are used to spare normal tissue
  • CT is used to define the area to be treated and the direction of the radiotherapy beams that are used
359
Q

What is the basis of nuclear medicine?

A
  • indirect radioactive tracer, patient is emitting the gamma rays
  • time amounts of tracer (nmol) (x-ray is 10’s of g!)
  • image depends on the metabolism of the tracer: FUNCTIONAL IMAGING

We are making an image of the distribution of a radioactive tracer. Nuclear medicine only shows function. It may reflect anatomy but without metabolism, the tracer will not be taken up. Nuclear medicine is a functional modality.

360
Q

What is gamma camera imaging? (basic)

A
  • uses single photon emitting radio nucleus
  • can operate in 2D (planar) and 3D (SPECT)
  • SPECT: single programme emission computer tomogram

Gamma cameras have imaging ‘heads’. They re used for radionuclides that decay with direct emission of gamma rays - the most common radionuclide is Tc-99m (half life is 6 hours)

361
Q

What is the basis of PET scanning?

A
  • positron emission tomography
  • uses positron emitting radio nucleus
  • always 3D
  • a ring of scintillation detectors supported in a fixed gantry
  • operated in ‘coincidence mode’ - only photons emitted from an annihilation event are recorded
362
Q

What is half life?

A

half life is the time taken for the radioactivity to reduce to 50%

363
Q

How does dynamic renal transplant work (gamma camera)?

A
  • cameras positioned above the patient
  • Te99m DTPA injected IV
  • camera positioned able the patient
  • gamma camera records gamma rays and collects image over time
  • functional time-activity curves are obtained
364
Q

What is SPECT ‘datscan’?

A

DaTSCAN — I-123 FP-CIT — Ioflupane

  • Binds to the dopamine transporters (DAT), on the neutrons
  • Parkinson’s disease has reduced uptake in the Putamen differentiates from Essential Tremor
365
Q

What happens in Beta+ particle emission?

A
  • both positron and electron are annihilated

- 2 gamma rays are created at 180 degrees to one another

366
Q

What is the metabolism of EDG?

A

FDG is a glucose analogue which enters cells in the same way as glucose.

There is good reflection of the distribution of glucose uptake and phosphorylation by cells in the body

Glucose –(phosphorylation)–> glucose-6P –> glycolysis

FDG –(phosphorylation)–> FDG-6P –X–> metabolically trapped

367
Q

What is neurodegenerative dementia?

A
  • FDG reflects metabolic activity
  • alzheimer’s disease (hypo metabolism, mostly in temporal and parietal regions)
  • Pick’s disease (fronto-temporal hypometabolism)
368
Q

What are examples of hybrid imaging?

A
  • PET-CT
  • SPECT-CT (used to localise uptake)
  • PET-MR
369
Q

What does a michaelis-menton curve represent?

A
  • relates velocity to [substrate]
  • Km = substrate concentration at which 50% maximal velocity is reached

We can measure the velocity of an enzyme and plot it against the concentration of the substrate.

Vmax is when the enzyme is working at full rate, achieved when you have an infinite amount of substrate.
A very high Km means the enzyme doesn’t bind the substrate very strongly.

We can use this information to set up our assay, we want to be close to the Vmax and so we can approximate how much substrate is needed. We would possibly use around 10xKm because we can’t put in an infinite amount.

Measuring the production of the product is another way of measuring the velocity of the reaction. Take the gradient of this to get the velocity of the enzyme reaction.

If you have a very valuable substrate and can’t put in 10xKm then it might be depleted over time and you may get product inhibition.

If there are some impurities in the reaction then it takes a long time to get ready and then goes into a linear phase before the reaction tails off.

370
Q

What conditions do we need to consider for activity measurement of enzymes?

A

Temperature: Q10: relative reaction rates at 2 temperatures differing by 10*C

Optimum pH: can vary from 1.5-10

Precise substrate concentration and ionic strength of buffers.

We have to optimise the temp as if its too low the reaction will be too slow, and it it is too high then it may denature the enzymes (~40*C) and it will become labile. If you increase the temp by a factor of 10 you get approximately doubling of the rate - this is Q10.

Ionic strength of suffers is also important - some require a certain amount of salt in the buffer for them to work.

371
Q

What are the units of enzyme activity?

A

INTERNATIONAL UNIT (IU): one unit catalyses conversion of 1micromol of substrate in 1 min (temp not defined)

KATAL: one katal converts 1 mol substrate/second (1kat = 60mol/min = 6x10^7 IU. 1 IU = 1micromol/min - 16.67 nkat)

  • can standardise them in some places and constitutions
  • under defined conditions
  • katal is used in industry and is on a much larger scale
  • normally just use the international unit (IU)
372
Q

what are practical considerations of enzyme assays?

A
  • enzyme purity and nature - in a lab we want a very pure enzyme
  • enzyme stability - under assay conditions
  • quality of substrates, solvents and buffers - high quality, uncontaminated
  • assay mixture - make up everything together, only thing to do is add the enzyme to start the reaction
  • mixing to start the reaction

ASSAY: measure the activity of that enzyme under particular conditions

373
Q

What are some methods of continuously monitoring enzyme catalysed reactions?

A

ABSORBANCE: one component may absorb UV lots, and this may decrease as it changes to product
FLUORESCENCE: of the substrate or the product
OPTICAL ROTATION: when we convert the substrate to the product with a different chiral form, it will change the rotation of polarised light
CONDUCTIVITY: release of proton/consumed proton
VISCOSITY: polymer into monomer - change viscosity
RADIOCHEMICAL: when impossible to use any of the above, measure formation of product by measuring the radioactivity. Need separation to separate the substrate and the product to do this.

374
Q

How does spectrophotometry work?

A
  • if substrate or product absorbs light, reaction can be followed by the change in absorbance –> A - log10 I0/I
  • I0 is intensity of incident light and I is that of the transmitted light
  • Absorbance is related to concentration of light absorbing substance (Beer-Lambert law: A = eco)

Relationship between [substrate] and absorbance/ Proportional to path length that the light has to go through. If you double path length, absorbance will decrease by 2x. It is dependent on the intrinsic absorbance of the molecule you are following.

We can plot for an enzyme reaction the change in absorbance and can work back from this to the concentration of the substrate.

375
Q

What are general analytical problems in enzyme assays?

A
  • standardisation of assays
  • quality control (must know the expected variation of the control samples - coefficient of variation (CV))

standard deviation (SD) of control values/mean of the control values x 100 (%)

376
Q

How do we use internal controls in assays on large scale?

A

there is protocol for this. to enable yo to standardise the assay you need to put tubes in with known amount of enzyme activity.

every so often these come up and throughout the day you will measure the activity of the enzyme in these controls. you will know if the machine is working properly throughout the day because of this. Ways of knowing if it is to isn’t by looking at the standard deviation of the control values and monitoring that throughout the day.

377
Q

How does lactate dehydrogenase work?

A

Takes lactate and NADH and converts the lactate to NAD. NADH is highly absorbent and we can follow the reaction by watching the absorbance of NADH fall.

NADH is more stable than NAD - we can follow this continuously by looking at the decree in absorbance of NADH.

The forward reaction from pyruvate is favoured because:

a. equilibrium to the right
b. maximal velocity higher to right
c. NADH more stable than NAD

Lactate dehydrogenase is released by blood cells in anaemia.

378
Q

What is alanine?

A

Alanine is produced largely in the liver. In liver dysfunction the level of ALT increases massively se we get a large increase in activity. ALT switches over the amino and kept groups in L-alanine and 2-oxoglutarate. We can use the previous reaction in an assay.

There is another assay as liver enzymes using aspartate which makes oxaloacetate which is a substrate for malatedehydrogenase.

379
Q

How do we measure alkaline phosphate (ALP)?

A

4-nitrophenol phosphate (colourless) –> 4-nitrophenol (yellow) + phosphate (colourless)

We can set up a reaction to measure the formation of the product directly without enzymes first. We can remove the phosphate from the substrate, which is done at very high pH (10.3).

We can set up there reaction to use a particular phosphate which is colourless (4-nitrophenol phosphate), this is a synthetic molecule which is not found in the body. When alkaline phosphatase works on it we get 4-nitrophenol + phosphate. 4-nitrophenol is yellow and by monitoring how much of this we have, we can work out how much enzyme we have.

380
Q

Where is alkaline phosphatase found in the body?

A

Liver dysfunction, especially bile duct problems, cause increased levels of alkaline phosphate. There is also a bone form which is released in bad conditions.

Bone and liver alkaline phosphatase are slightly different in structure as they have different sugars on them and they are different isoforms. they can be detected immunologically. We can run a gel and see which is increased in its presence in the blood, which is a very useful assay.

381
Q

How can we use enzymes to measure the substrates in the blood?

A

We want to measure glucose in diabetics for example. One of the things we can do is use the reaction of hexokinase with ATP that will make G6P and ADP. We can use absorbance to measure any of these. G6P is the substrate for G6PHD, which converts NAD to NADH. This can be used to measure the amount of starting glucose and the final NADH level.

This reaction is completely specific for G6P, meaning it doesn’t measure anything else in the blood so this is an advantage. Glucoseoxidase is specific to glucose.

382
Q

what are examples of clinically important enzymes?

A

Amylase: acute pancreatitis
Alkaline phosphatase
: liver, bone
Alanine aminotransferase: liver disease
Aspartate aminotransferase: liver infarction
Gamma glutarmyltranspeptidase: hepatobiliary disease
Lactate dehydrogenase: haemolysis, lymphoma
Creatine kinase
: skeletal disorders

  • = isoforms - separating of bone and liver ALP useful
383
Q

What are tests of association?

A
  • correlation
  • regression
  • chi squared test
384
Q

What is the Pearson correlation coefficient (R)?

A

Measures whether 2 variables that you have measured vary together i.e. does X tend to go up as Y goes up?

R = 0 --> no correlation
R = 1 --> perfect correlation
R = -1 --> perfect negative correlation, one variable goes up as the other goes down

(some statisticians use R^2 instead of R)

You also need a p value to say if the correlation is statistically significant. The p value is a measure of whether yo results look like they could be purely random data.

385
Q

What does the Pearson correlate assume?

A
  • the two variables are independent of each other
  • both variables are normally distributed; but the test is robust, in practice shape of distribution is not crucial, but others can cause an artifactual positive correlation
  • the relationship between the variables is linear
386
Q

What is the difference between correlation and regression?

A

Correlation is used when both variables are measurements, which may have random errors or random variation. These usually result from observational studies.

Regression is used for experimental studies where we have set one variable in advance e.g. time or dose of drug

387
Q

What is linear regression?

A

Linear regression estimates the best straight line through the data. It returns a value for the intercept (a) and slope (b) for the line y = a + bx

If you know biologically that y must be 0 when x=0, then you can ask for the best line that that goes through the origin (intercept = 0).

While regression estimates the best straight line through the data, it is just an estimate as our data is subject to random errors.

388
Q

What is the chi squared test?

A

sometimes the data from a study are counts for the number of individual events falling into a particular category.

There may appear to be an association between two factors but this could just be random variation.

If the data can be presented as a 2xn table, then you can test for differences using the chi squared test.

389
Q

What happens in split brain patients?

A

Split brain patients have undergone surgery to cut the corpus callous, the main brain bundle of neuronal fibres connecting the two sides of the brain.

Input from the left visual field is processed by the right hemisphere and vice versa. A word is flashed briefly to the right field of view and patient asked what they saw. Left hemisphere is dominant for verbal processing so patients answer matches the word. When a word is flashed to the left field of view and the patient is asked what he saw, as the right hemisphere can’t communicate with the left, the patient is unable to say what he saw, but he can draw it.

The corpus callous can be cut to reduce the spread of epilepsy. These patients helped prove hemispheric lateralisation of function.

390
Q

What is electroencephalography (EEG)?

A

EEG is the recording of the brains electrical activity through electrodes on the scalp.

P300 wave related to cognitive function.

391
Q

What is trans-magnetic stimulation (TMS)?

A

Electromagnetic induction induces weak electric currents - can cause activity in specific or general parts of the brain - repetitive transcranial magnetic stimulation (rTMS), is being tested as a treatment for various disorders including migraines, strokes, Parkinson’s disease and strokes.

392
Q

What is construct validity?

A

the model has a sound theoretical rationale (neurobiological or psychological mechanisms, aetiology), the way humans get the disease is similar to how animals do.

393
Q

What is face validity?

A

phenomenological similarity between the model and the disorders being modelled (symptoms)

394
Q

What is predictive validity?

A

manipulations known to influence the pathological state should have similar effects in the model (drugs work the same in both)

395
Q

What kinds of stroke can be observed in animal models of the CNS?

A
  • global ischaemia: 2 or 4 vessel occlusion
  • focal ischaemia: middle cerebral artery (MCA) occlusion
  • haemorrhagic: inject collagenase into rat brain - causes bleeding
396
Q

What is a behavioural test for stroke?

A

Morris water maze - test of memory where rodent uses visual cues to try and find the underwater platform

397
Q

What is the very basis of Parkinson’s disease?

A

Loss of dopamine neutrons in the substantia nigra

398
Q

What are the neurotoxins for Parkinson’s disease?

A

6-OHDA: toxin kills dopamine (and NA) neurones but no extrastriatal loss and no Lewy bodies.
MPTP: toxin kills dopamine cells (see MPP+)
Rotenone: agrochemical but neurotoxic
MPP+ from MPTP: inhibits mitochondria
LPS: lipopolysaccharide which induces inflammation

399
Q

To be said to have depression, what symptoms need to be present?

A

5+ of these symptoms need to be present in the same 2 week period (nearly every day/all day) and represent a change from previous functioning…

  1. depressed mood
  2. markedly diminished interest or pleasure in (almost) all activities
  3. significant weight loss when not dieting or weight gain or decrease in appetite
  4. insomnia or hypersomnia nearly every day
  5. psychomotor agitation or retardation
  6. fatigue or loss of energy
  7. feelings of worthlessness or excessive guilt (inappropriate)
  8. diminished ability to think or concentrate, or indecisiveness
  9. recurrent thoughts of death, suicidal ideation without a specific plan, or a suicide attempt or a specific plan for committing suicide
400
Q

What various induced ‘feelings’ could we sue to make a rat depressed?

A
  • chronic mild stress (different small stressors most days for ~6 weeks)
  • learned helplessness
  • social defeat
  • maternal separation
  • olfactory bulbectomy
  • disruption of pair bonds
401
Q

What is ‘learned helplessness’?

A

Rat learns that it cannot escape form an electric shock and gives up trying. Also demonstrated in dogs - in the end they just lay down and whine.

402
Q

What is social defeat?

A

smaller rat bullied by a bigger rat

chronic stress that occurs in the presence of persistent loss in aggressive social encounters especially in the absence of social support

403
Q

What is maternal separation?

A

Take young pups away from mother very early.

We get an increase in ultrasonic vocalisation (USV) of some frequencies and on return of pups to mothers we get an increase in USV at 50Hz (happy sound). There is some debate about what each USV frequency might mean.

404
Q

What is an olfactory bulbectomy?

A

destroy the olfactory bulb.

Whiskers and smell are very important to rats - a lot of brain is used in processing the information from its snout and whiskers. A rat that has has its whiskers or olfactory bulb removed might be ‘depressed’

405
Q

What is disruption of pair bonds (depression)?

A

separate male and female gerbil partners

oxytocin? mongolian gerbils mate for life so if you separate them, they become ‘depressed’

406
Q

What are the ways of trying to measure depression in a rat or mouse?

A
  • sucrose choice test (good model)
  • forced swim test
  • tail suspension test
407
Q

What is the sucrose choice test?

A

‘Depressed’ animals may seem to choose water over sucrose when offered the choice.
Chronic mild stress reduces sucrose intake, wuline (chinese herbal medicine) appears to be antidepressant as it normalises sucrose intake. Most clinically used antidepressants would normalise sucrose intake.

408
Q

What is the forced swim test?

A

Measure swimming and escape behaviour (climbing). Depressed rats are less active in both respects.

antidepressants (fluoxetine and desipramine) caused increased swimming and escape behaviour

409
Q

What is the tail suspension test?

A

Mouse/rat struggles to escape and eventually gives up - measure latency to giving up. Antidepressants increase this latency.

410
Q

What tests are designed to detect anxiolytic actions of drugs but not selective effects in animal models?

A
  • elevated plus maze
  • social interaction test
  • black/white box
  • object burying
411
Q

What is the elevated plus maze?

A

rodents prefer close arms to open arms. placed in centre and number of entries and amount of time spend in the open and closed arms is recorded.

Anxiolytic drugs increase time in open arms.

412
Q

What is the social interaction test?

A

used to examine social interactions between 2 rats or mice in a novel environment. 2 rodents that don’t know each other will sped some time interacting, anxiolytic drugs increase this time.

413
Q

What is the light-dark box test?

A

rodents prefer dark to light spaces - a rat or mouse is places in the dark part of the box.

Measure:

  1. latency to leave the dark box
  2. time spent in light box
  3. number of crossings between the boxes

anxiolytic drugs may increase time spent in the light box

414
Q

What is the object burying test?

A

introducing a new object of aversion to a familiar cage causes anxiety in rats and mice, stimulating them to engage in burying behaviour. Anxiolytic drugs reduce this behaviour.

415
Q

What is latent inhibition - pre-pulse inhibition?

A
  • use a conditioning paradigm
  • associated bell with food
  • if we use the bell many times beforehand, it takes longer to condition the animal/person
  • not the case with schizophrenics - they learn the conditioning (e.g. the bell) very quickly
416
Q

What symptoms have to be present to be diagnosed as having a drug addiction?

A
  1. recurrent use resulting in a failure to fulfil major obligations at work, school or home
  2. legal problems resulting form recurrent use
  3. continued use despite significant social or interpersonal problems caused by the substance use
  4. recurrent use in situations which are physically hazardous (e.g. driving while intoxicated)
417
Q

What are symptoms of substance dependence:

A
  1. substance abuse, PLUS…
  2. increase in tolerance
  3. withdrawal symptoms
  4. compulsion to use the drug
418
Q

What is conditioned place preference?

A

if you give a rat cocaine always in the white compartment and placebo (saline) in the black compartment, the rat will always choose to spend relatively more time in the white compartment as it associates this with feeling good

419
Q

What effect does cocaine withdrawal have on social interaction?

A

decreased social interaction

2 rats initially spend ~70s of 5 mins interacting. if one rat is given cocaine (1mg/kg) then it spends more time interacting (~120s on day 7 and 140s on day 14). if the rats cocaine is stopped at day 14, on the following day it will only spend ~10s interacting - this is a withdrawal symptom. Some drugs (e.g. ondansetron) can reduce these withdrawal effects

420
Q

What is locomotor sensitisation?

A

on 1st occasion a rat is injected with cocaine is moves around a little. if it is given cocaine every day for a week then given the same dose it initially got, it moves around more. after about 2 weeks of cocaine it moves around even more - this is locomotor sensitisation

421
Q

What can be observed with intravenous (IV) self administration of rats with cocaine?

A

the rat has an IV line in its jugular vein. it can press a bar for cocaine through the IV line. at its preferred dose it will self administer cocaine all day and eventually overdose if allowed - rats like cocaine and will work hard for it

422
Q

What is the wisconsin card sorting test?

A

schizophrenics have problems with this test.

the subject has to determine a rule (e.g. must attend to colour/shape/number of symbols on the card). a related test has been developed for rats, not involving cards. this is a test for cognitive function in humans.

423
Q

What is the adaption of the Wisconsin card sorting test for rats?

A

digging task for cognitive function: attentional set-shifting

the rat must learn rules e.g. food is in the cumin disk - only allowed to dig in one pot for food

in some cases PCP (phenylcycxlindine) leads to long term cognitive dysfunction and PSP treated rats can take longer to learn the rule changes i.e. they have cognitive deficits

424
Q

What is the technique for measuring NTs in living animals?

A
  1. anaesthetise animal
  2. place animals head in stereotaxic frame
  3. drill hole into skull
  4. insert guide cannula (sometimes)
  5. insert probe
  6. do experiment then kill (overdose) or allow animal to recover
425
Q

What are the advantages and disadvantages of using anaesthetised animals in measuring NTs?

A

ADVANTAGES:

  • animal feels no pain, less stress?
  • able to better control the experiment

DISADVANTAGES:

  • some anaesthetics influence transmitter release
  • cannot correlate transmitters with behaviour
426
Q

What is the correlation between dopamine and sexual behaviour?

A

increased dopamine levels found during sexual encounters

427
Q

What effect does 5-HT have on exercise?

A

with 5-HT rather than glucose there is an increase in exercise from 90 minutes to 270 minutes (in male rats)

428
Q

What are the different ways of measuring NTs in vitro?

A
  • brain slices
  • organotypic brain slices
  • prisms
  • cell cultures
  • synaptosomes
429
Q

How are brain slices performed?

A
  • kills rodent humanely (anaesthetised or cervical dislocation)
  • open skull
  • take out brain
  • cut out region of interest
  • slice section of brain of interest with ‘vibrate’ - typically 300-400 micrometers thick
  • vibrate moves blade forward slowing with sawing action
430
Q

What is a brain slice chamber?

A

A typical brain slice chamber can be kept alive for hours with heated (e.g. 32*C) oxygenated artificial cerebrospinal fluid (ACSF, contains salts and glucose). In this case a water bath heats the ACSF

431
Q

What is the method of organotypic brain slices?

A

Same as for brain slices except…

  • take thinner slices (e.g. 200 micrometers)
  • take tissue from very young rodents (e.g. PND7)
  • use antiseptic technique (sterilise)
  • culture slice in ‘incubator’ (long-term experiment)
  • use an air-liquid interface
  • use -culture medium’ (e.g. horse serum and antibiotic) not ACSF (just salts and glucose)
  • can use ‘co-cultures’ e.g. a piece of cortex cultured with a piece of accumbent - will they make appropriate connections?
  • NC3Rs - reduce animal use in research –> reduction, refinement, replacement of vertebrates

Recent data suggests that one can use older rodents (e.g. 40 days old). Normally we see a loss of cells and structure after culture. Here, and antisense, which blocks gap junction formation, promotes slice health after 14 days in vitro.

432
Q

What are prisms?

A

A rush block of brain, not used much anymore - record from thin site where tissue is viable - it gets more oxygen and salts than deeper tissue and so it is healthier.

433
Q

What can we use for neuronal cell cultures?

A
  • neuronal cell lines (from frozen)
  • dissociated tissue from neonate (e.g. PNd7)
  • other cell lines with neuronal characteristics and use nerve growth factors to induce a neuronal phenotype (e.g. PC12 cells)

We can record ‘quantal’ dopamine release from adrenal chromatin cells (quantal release: release from a single vesicle)

434
Q

What can BDNF induce?

A

neuronal phenotype in PC12 cells - increased dopamine transporters and ‘dendrite growth’

435
Q

What can epidermal growth factor (EGF) reportedly induce?

A

the functional expression of dopamine receptors in cells (Missale et al)

436
Q

What are synaptosomes?

A

the brain sample is homogenised then spun in a centrifuge at different forces (g) until the sample has a band of ‘‘synaptosomes’’ which are basically just synapses but contain many receptors, uptake sites and enzymes which are involved in transmitter synthesis, release and metabolism.

437
Q

What can whole animal in vitro preparations tell us about NTs?

A
  • the different afferent and efferent connections are intact
  • can drugs cross the BBB?
  • subjected to metabolism in the gut and liver
438
Q

What can brain slice in vitro preparations tell us about NTs?

A
  • no information on systemic metabolism but this cn be useful if one only wants to determine the exact pharmacological mechanisms of a drug
  • can measure NTs with better spatial accuracy - one can see the brain nuclei down the microscope, in vivo one must use a stereotaxic frame
  • only have some afferent and efferent connections
439
Q

What can single cell in vitro preparations tell us about NTs?

A
  • no afferent or efferent connections
  • limited information on drug metabolism
  • but a much simpler system to get simple mechanistic answers e.g. on QUANTAL release from single or few transmitter vesicles
440
Q

What are the different techniques to measure NTs?

A
  1. radio-labeling
  2. HPLC
  3. push-pull cannula
  4. microdialysis
  5. voltammetry
  6. aperometry
441
Q

How is radio labelling done?

A
  • incubate brain slice in tritiated dopamine [3H]DA
  • dopamine neurones take up radioactive dopamine through DAT
  • when dopamine is released one can measure the amount of dopamine which is proportional to the radioactivity
  • not used so much anymore
442
Q

How is HPLC done?

A

sample is injected into HPLC column, which separates the transmitters and metabolites based on their size and charge. in this example, 2 dopamine metabolites DOPAC and HVA come out of the column first, then dopamine and finally a metabolite of 5-HT - 5HIAA

Typically, areas under the curve are measured and compared with standards i.e. known quantities of transmitter

443
Q

How is push-pull cannular done (in vitro transmitter release)?

A

not used much any more. buffer is pushed down the cannula, which is in the brain, then drawn back up - test to see what transmitters are picked up. micro dialysis has superseded this technique.

444
Q

How do we measure in vivo NT release using a micro dialysis probe?

A

The micro dialysis probe is implanted in the brain under general anaesthetic some days prior to experimentation (can be used in humans).

The tip is the only part exposed to the brain and transmitters cross the semi-permeable membrane by diffusion down their concentration gradient. Note you can also add drugs to the brain by having them in your buffer.

445
Q

What is voltammetry?

A
  • uses small carbon fibre electrodes inserted directly into the brain
  • electrode oxidises the transmitter and the transfer of electrons is measured as a small current
  • oxidation varies with the potential of the electrode
446
Q

What are the advantages and disadvantages of voltammetry?

A

ADVANTAGES:

  • near to real time (e.g. 10 samples/second)
  • great spacial resolution (8x30 micrometer tips)

DISADVANTAGES:
- currently limited to only a few compounds: dopamine, noradrenaline and serotonin and some of their metabolites, NO etc - they must oxidise at low voltages

447
Q

What happens in voltammetry at the carbon fibre electrodes?

A

a glass electrode filled with a single CF is pulled (heated and pulled). The carbon tip is trimmed to about 20 micrometers.

Apply a triangular voltage waveform (-1 to +1.4 V) to the CFE and record the current flowing through it. At about 600mV dopamine oxidises (more current) and at about -200mM dopamine reduces (less current).

V = IR, current doesnt follow voltage because of capacitive properties of carbon. A sample takes 20ms

448
Q

What are examples of where we can use voltammetry?

A
  • in brain slice
  • in brain slices can measure anoxia dopamine release in caudate
  • in anaesthetised rats
449
Q

What is in vivo voltammeter (freely moving)?

A

Voltammetry is fast enough to correlate transmitter release with behaviour on a sub-second time scale - can look at many different behaviours, and behaviours that only last a second, not just sleep, locomotive etc

450
Q

What is aperometry?

A

This is similar to voltammeter except that the voltage is not changed, i.e. a constant voltage is applied to the carbon electrode (for dopamine = 600mV).

Thus, we do not get any information about oxidation or reduction peaks. The main advantage is that sampling is essentially continuous, i.e. ‘real-time’.

451
Q

What is the pharmaceutical drug discovery process?

A
  • test in simple system (e.g. synaptosomes, cell culture)
  • go on to more complex system (e.g. brain slices)
  • test in whole animals: toxicity data, effect in animal models
  • test in humans
452
Q

What are characteristic pathological features of alzheimers disease?

A
  • shrunken brain
  • ventricles are swollen
  • thickened and sunken sulci
  • thinner grey matter ribbon
453
Q

About familial AD…

A

We now know that the most common gene mutation is on chromosome 21 and it is APP. This protein can have a little bit cut off of 40 aa. This fragment can form Abeta which forms the plaques.

454
Q

What are presenilins?

A

Presenilin 1 on chr 14 and presenilin 2 on chr 2

Presenilins are mutations associated with familial, early onset AD. It is quite rare and occurs before the age of 30.

455
Q

What is meant by ‘Swedish families’?

A

There is a swedish/london/netherands family and all have familial AD. They each have a slightly different mutation and as it is rare they are referred to as for example the Swedish family.

Swedish family mutation: double mutation at aa670-671 in APP.

These families are sometimes mentioned within the same breath as Down’s syndrome. The Down’s trisomy is an extra version of chromosome 21 and they have an extra copy of APP. It is possible that Down’s Syndrome is a kind of mirror of AD.

456
Q

Why can AD be referred to as the 21st century plague?

A
  • in recent years deaths from many other disease have decreased
  • however deaths from Ad have increased
  • this may be because people are living longer so more people are ending up getting it
  • causing a massive drain on healthcare budgets and time and is a sad way to live
457
Q

How does AD develop with age?

A

as we get older pathology accumulates in our brains and can take us to a recognised state of dementia. We can’t be diagnosed as having dementia until a symptomatic threshold is reached.

This may have been preceded by a preclinical phase we don’t know about… the brain may have absorbed some of this pathology - loses some neurones and can still cope.

458
Q

How might AD therapy work?

A

If we could delay AD onset by even two years this would be very good.

Therapy and a ‘cure’ may not be about stopping it, but slowing it and reducing risk. Once you have full dementia there is nothing that can be done - there is no reversal and lost neurones can’t be replaced.

459
Q

How are cognitive scales used in dementia?

A

Cognitive scales (MMSE, CAMCOG, MOCA) allow you to assess the patient. This is useful as even if a patient is very good to begin with and is then average after time, this still shows a decline.

460
Q

How does ADAS-Cog work?

A

The patient is given a list of words to remember and after two minutes they are tested to see how many they remember. This is a good method but is hard to replicate reliably over different countries because the words have to be different in different languages so this makes it less fair.

461
Q

What are senile plaques in AD?

A

extracellular deposits of Abeta fibrils also in downs syndrome teenagers

462
Q

How is Abeta cleaved?

A

Beta is cleaved from APPby secretase enzymes: alpha, beta and gamma.

  • beta secretase = BACE
  • gamma secretase - presenilins are essential cofactors and produce Abeta42 and Abeta40

The three secretase enzymes have an intracellular C terminus and an N terminus that sticks out.

463
Q

What is the difference between healthy and unhealthy Abeta cleavage?

A

In healthy patients alpha-secretase cleaves APP forming fragments and no harm is done.

If it gets excessively chopped by beta secretase and then gamma secretase, we end up with tiny fragments which form plaques from the Abeta - they can be 40 or 42 aa in length.

464
Q

What are PIB scans?

A
  • used wth Abeta in diagnosis
  • small molecules have been developed and are given to the patient
  • they pass through the BBB and bind selectively to amyloid plaques
  • Abeta is everywhere in AD patients and there is loads of binding
  • this is a way of determining amyloid load in live patients
  • would be great if we had a way of getting rid of the amyloid but we don’t know how at the minute
465
Q

About tauopathy and tangles…

A
  • Tau is part of the neuronal cytoskeleton
  • essential and is a type of scaffolding for neurones
  • it is bad if it is HYPERPHOSPHORYLATED (some phosphorylation is normal)
  • this makes the filamentous tau protein crinkle into a tangle
  • fragments inside neurones and cerebral cortex

Hyperphosphorylated tau looks like little balls/clumped, when it should look like neat parallel railway tracks.

466
Q

About hyperphosphorylated tau…

A
  • filaments are in nerve cell process
  • wants to keep it intact and going where it is meant to via connections
  • if filaments become defected and tangle, the axon becomes pathological and may die
  • neutron may live but the circuit won’t work

SOME PHOSPHORYLATION IS NORMAL - BUT TOO MUCH IS NOT

467
Q

Are plaques or tangles more important in the molecular pathogenesis of AD?

A

We don’t really know how plaques and tangles come and how they contribute. There is debate as to whether tau or amyloid is more important…

  • nerve cells grown in petri dish from mice
  • grown in culture and treated with Abeta
  • 4 days later many of the axons are gone and those remaining are regressing and fragmented
  • this was done again wth mice genetically altered to have the tau protein gene removed
  • the same process is done with its nerve cells
  • they are resistant to Abeta and are unaffected

The logical conclusion of this is that tau is needed for Abeta to have an effect. Abeta comes first, causing the defective tau and tagging and therefore the neutron dies

468
Q

About cerebral amyloid angiopathy…

A

Amyloid not only found in plaques - can be found around blood vessels also.

Abeta40 is found clustered around the vessel and Abeta42 forms a messy corona around that - they are very distinct.

In someone who has this, they tend to have both 40 and 42. 40 is a feature of the vascular type and 42 is labelled a a plaque. Can have vascular amyloid as well as plaque based.

Vascular is also damaging but less so than plaques.

469
Q

What are the current AD therapies?

A

CHOLINESTERASE INHIBITORS:

  • Donezepil
  • Rivastigmine
  • Galantamine

Memantine - NMDA receptor antagonist
…and Immunotherapy

470
Q

What is the rationale for the use of cholinesterase inhibitors in AD?

A

The cholinergic system is involved in memory and Ach is a major contributor in these circuits. Many people with mild cognitive impairment (MCI) see a benefit from the cholinesterase inhibitors but not all.

471
Q

How is it thought that immunotherapy could be used to treat AD?

A

We know that Abeta is a protein and that it is what causes the problem. Therefore, if we can make an antibody then it will find the Abeta in the plaques and stop them from forming and could even get rid of them.

472
Q

What is the Elan vaccine trial?

A
  • vaccine AN-1792
  • active immunotherapy (n=372)
  • full length Abeta 1-42 peptide and adjuvant
  • 15 AD patients: aseptic meningoencephalitis
  • terminated in jan 2002 after negative results and a death

As the remaining 14 patients died over time their brains were examined. It was seen that in 3 of the patients all the plaques had completely gone. However, these patients still had a score of 0 on MMSE (highest 31 and is healthy) and so this showed that Abeta does its damage very early on and so we think that it is too late in severe dementia to do any help. If we looked for tau in these brains it would be abundant and neuronal cell death would be prevalent.

Basically, the vaccine worked - but it didn’t. It is too late when the AD has become symptomatic.