Microbiology Flashcards

(451 cards)

1
Q

What has happened 4.5 Gy ago?

A

Gy = a billion years

4.5 Gy - Earth was formed: a supernova (explosion) gave rise to the sun and planets (material clumped and fused under the influence of gravity, it was then cooled)

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

What happened after Earth’s formation?

A

4 Gy - formation of water and atmosphere (collision with icy comets and volcanic outgasing (vapour))

3.9 Gy - oldest sedimentary rocks - Greenland (Earth cooled down and water condensed to form oceans)

3.5 Gy - stomatolites (sediments of alternating layers of limestone and bacterial communities ‘microbial mats’)

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

What are some microfossils?

A

Found in stromatolites

Filamentous prokaryotes which look like Leptothrix
Colonial cyanobacteria which look like Entophysalis
Algae which look like Red algae

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

What are the four theories which explain the origin of life?

A

1) Chemical origin: how were simple molecules (aa’s and bases) created abiotically ‘prebiotic soup’

2) RNA world: why is RNA likely to be the first macromolecule to underpin abiotic reactions

3) Apparition of cellular life: how could prebiotic chemistry lead to a cellular life

4) Panspermia: life came from space

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

How did we get from biological building blocks to cells?

A

4.3-3.8 billion years ago:
Biological building blocks (amino acids, nucleosides, sugars) –> RNA world (catalytic RNA and self-replicating) –> Protein synthesis (translation) –> DNA (replication, transcription) –> lipid bilayers (cellular compartments) - early cells had high rates of HGT

3.8-3.7 billion years ago:
Divergence of bacteria and archaea (components of DNA replication, transcription and translation all in place)

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

What is the chemical origin of life theory?

A

Different theories:
1861 - synthesis of sugars from atmospherically produced formaldehyde raining onto basic minerals
1953 - synthesis of amino acids from lightning in a reducing environment
1960 - synthesis of nucleobases from polymerisation of ammonium cyanide in solution

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

What is the Miller experiment?

A

Based on the hypothesis of a reducing atmosphere - replicated the early Earth’s atmosphere and saw most amino acids can be formed from simple chemicals

Added H2, methane and ammonia with spark - formed amino acids - requires high H2 concentration

We are assuming that the conditions were like that.

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

What is the primordial soup model?

A

Precursors of ribonucleotides, amino acids and lipids have arisen through chemicals under distinct geological conditions.

All life building blocks can be produced from the same pool of compounds under distinct conditions:
Hydrogen cyanide - precursor
Hydrogen sulfide
UV

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

What is the RNA world hypothesis?

A

Could have been the first macromolecule encoding complex information.

  • 4 building blocks (compared to 20 amino acids)
  • requires less energy than DNA to form and degrade
  • uracil is formed early in biochemical pathways
  • some viruses use ssRNA as genetic material
  • some have catalytic activities RIBOZYMES
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Who discovered ribozymes?

A

Thomas Cech and Sydney Altman

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

What are the activities of ribozymes?

A
  • cleave and ligate RNA molecules (splicing)
  • replication
  • form peptide bonds (ribosomes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Why is compartmentalisation beneficial?

A

Protect from the environment
Selective barrier
Concentration of molecules (increases rate)

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

How to create compartmentalisation?

A

Phospholipids - spontaneously assemble to form protocells

This trapped amino acids and nucleic acids - first primitive cellular form of life

pH dependant

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

How did compartmentalisation occur to give rise to the last universal common ancestor (LUCA)?

A

Hypothesis 1: surface origin
Primitive cells were formed spontaneously on Earth’s surface from a prebiotic soup - natural selection led to optimise metabolic processes

Hypothesis 2: subsurface origin
Life appeared in more stable conditions on ocean floor in hydrothermal mounds. Geothermal heated water from fissure of sea floor + cool ocean water (iron, silicates, carbonates, Mg) = porous montmorillonite clays
H2 and H2S used as source of electrons to form organic molecules (ATP/Fatty acids)

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

What is panspermia?

A

Life originated from space: Chandra Wickramasinghe and Fred Hoyle

Evolution is driven by constant influx of viruses from space causing pandemics:
- Spanish flu
- Covid-19
- Candida auris

However - very little evidence

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

How big are microbes?

A

From 10 nm to 1mm

Poliovirus - 20 nm
Influenza virus - 100 nm
Enterococcus faecalis - 1-2 micrometres
Dimeregramma (golden algae) - 50 micrometres
Amoeba (protist) - 100-750 micrometres

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

When did we see the first cell using a microscope?

A

Leeuwenhoek in 1674

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

What is binomial nomenclature?

A

Carl Linnaeus - defines kingdom, class ect.

In 1735

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

Who made the kingdoms?

A

1866 - Haeckel made 3 kingdoms - animalia, plantae and protostista

1938 - Copeland made 4 - added monera (prokaryotes)

1959 - Whittaker made 5 kingdoms - added fungi

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

Who made the 3 domains?

A

Carl Woese - bacteria, archaea, eukarya

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

What are the three analytical methods of microbial diversity?

A
  • genetic plasticity drives diversity
  • taxonomy
  • phylogeny
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is genetic plasticity?

A

Drives microbial diversity - ability of an organism to change its phenotype in response to different environments

  • haploid genome
  • rapid multiplication (binary fission)
  • horizontal gene transfer (free DNA = transformation, bacteriophage = transduction, plasmid = conjugation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is taxonomy?

A

Classification of organisms - binomial nomenclature
DK (animal, fungi, plants) PCOFGS

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

What is arthropoda?

A

Phylum - segmented animals with hard skeletons

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
How do we do phenotypic analyses?
For taxonomy - morphology/differential staining - can do in hospital - metabolic properties - biochemical tests and unknown bacteria are mixed - results show what organism it is - phage typing - fatty acid profiles - use extracts to form methyl esters then do gas chromatography - mass spectrometry - last 4 are with pure isolates
26
What is genotypic analyses?
For taxonomy - DNA/DNA hybridisation: 75% - same species, 25-50 - same genus - Fluorescent In Situ Hybridisation (FISH) - rDNA (16s) sequencing - Multi locus sequence typing/fingerprinting - Whole genome sequencing
27
What is phylogeny?
Study of evolutionary history of organisms - evolution Measured by comparing genetic information - uses sequences called molecular clocks - line them up and see similarities. These encode for conserved proteins with similar functions and are undergoing random and neutral mutations.
28
How do we show phylogeny?
Through cladograms (phylogenetic trees) - rooted - unrooted
29
What are the three domains?
Eukarya Archaea Bacteria
30
What are the differences between the three domains?
Archaea: prokaryotic, no peptidoglycan, mem lipid are branched carbon chains attached to glycerol by ether linkage, first aa is methionine, no antibiotic sensitivity, no rRNA loop, no tRNA arm Eukarya: membrane lipids are straight carbon chains attached to glycerol by ester linkage, first aa is methionine, no antibiotic sensitivity, no rRNA loop, there is a common arm of tRNA Bacteria: same membrane lipids as eukarya, first aa is formylmethionine, there is antibiotic sensitivity, there is a rRNA loop and there is a common arm in rRNA. Sometimes eukarya and bacteria are similar, sometimes eukarya and archaea is - shows how analysing different things show different results
31
Who looked at animals and plants?
History of animals - Aristotle in 400 BC Enquiry into plants - Theophrastus in 200 BC
32
What is the major differences between prokaryotic and eukaryotic cells with transmission electron microscopy?
Nucleus vs no nucleus - compartmentalisation Size Membrane bound organelles in eukaryotes
33
What is the nucleus like in eukaryotic cells?
- Contains chromatin = DNA with histones - histones only found in eukaryotes - Site of mRNA, tRNA and rRNA synthesis - Has euchromatin and heterochromatin (denser)
34
What is the ER like in eukaryotic cells?
RER: membrane network with ribosomes, site of proteinsynthesis, signals allow import into RER SER: no proteinsynthesis, lipid synthesis, steroids and toxin breakdown - important for protein trafficking
35
What is the golgi like in eukaryotic cells?
- Carbohydrate synthesis (plasma synthesis) - Modify proteins for specific targeting - receive vesicles from the RER
36
What are lysosomes and peroxisomes in eukaryotic cells?
Lysosomes: originate from golgi, contain digestive enzymes Peroxisomes: from ER, encorporate lipids and proteins from cytoplasm, oxidise alcohol and fatty acids
37
What is the mitochondria like in eukaryotic cells?
- Respiration/ox phos/kreb cycle --> ATP and reducing power Has own genome (15kB and 37 genes) - multiply by division Transcription can occur here
38
What are chloroplasts like in eukaryotic cells?
Made of thylakoids - form grana in stroma Converts light into organic compounds Similar to mitochondria - own DNA (150 kB) and multiply by division Transcription can occur here
39
What is the flagella/cilia like in eukaryotic cells?
- Bundles of 9 pairs of microtubules surrounding central pair - axoneme. - Microtubules attached together by nexin molecules. - ATP is used and motion is carried out by dynein. - Slide past eachother - whiplike.
40
What is the nucleoid in prokaryotic cells?
Usually one circular chromosomes - ds DNA - less that 10 Mbp Has histone like proteins - but not HISTONES Can have plasmids
41
What is the cytoplasm like in prokaryotic cells?
Some contain organelles (compartment enclosed with membrane with dedicated physiological function) e.g. magnetosomes (have magnetite - Fe3O4), photosynthetic membranes, internal membranes (planctomycetes) Contains proteins, tRNA, mRNA and ribosomes. Can contain inclusion bodies (protein bound): carboxysomes (CO2 reduction), storage granules (S, PO4, N2), gas vesicles
42
What is the envelope like in prokaryotic cells?
Cytoplasmic membrane (no carbohydrates or sterols) --> peptidoglycan --> polymers covalently bound to peptidoglycan --> outer membrane (not always got outer membrane) Pg + polymers = cell wall
43
What are the appendages in prokaryotic cells?
Pilus: appendage dedicated to conjugation (plasmid exchange) - long Fimbriae (pili): involved in adhering to host cells/surfaces - antigenic structures are made of 1 major protein Flagella: supramolecular assembly involved in motility
44
What are the four types of flagella in terms of number of flagella?
Monotrichous - 1 flagella Lophotrichous - many on one side Amphitrichous - one on each side Peritrichous - loads on all sides
45
What is the structure of the flagella in prokaryotes?
Simple - two protein subunits, in eukaryotes: complex microtubules Driven by ATP hydrolysis like eukaryotes
46
How do prokaryotes vs eukaryotes divide?
Eu: mitosis except for mitochondria and chloroplasts - binary fission Pro: binary fission
47
What are the ribosomes in prokaryotes vs eukaryotes?
Eu: 80s except for mitochondria and ribosomes - 70s Pro: 70s
48
What is endosymbiosis?
All cells from LUCA Incorporation of bacteria led to mitochondria and chloroplasts. Nucleus appeared before both. Chloroplasts came after mitochondria Doesn't account for the fact that prokaryotic and eukaryotic cells have similar lipid composition.
49
What is the evidence for endosymbiosis theory?
Both mitochondria and chloroplasts have inner and outer membranes with some signatures of prokaryotes - cardiolipin/peptidoglycan. They contain mitoribosomes and chlororibosomes - not identical to bacterial ribosomes. Semi autonomous organelles that can do binary fission.
50
What is the hydrogen hypothesis?
Mitochondria originated from engulfment of a H2 producing bacterium by a H2 consuming archaea Genes for lipid synthesis were transferred to host - gave rise to nucleus Chloroplasts were acquired later by endosymbiosis - first phototropic eukaryotes
51
What is a mesosome in prokaryotic cells?
Invagination of the cell membrane
52
What are the unicellular eukaryotes?
Fungi (can be multi) Protists: Algae - plant-like Protozoa - animal like Slime molds
53
What are the eukarya kingdoms?
Animalia Fungi Plantae Protista
54
Name three types of fungi
Molds - filamentous Yeasts - unicellular Basidiomycetes - mushrooms
55
Why are fungi important?
Ecological role: decomposers in carbon cycle Economical role: key in biotechnology and can be plant pathogens Human health: >1.5 million deaths from fungal infections
56
What are the common properties of fungi?
Morphology: most form multicellular filaments (hyphae) and are pleiomorphic (can have different forms) Cell walls: carbohydrates - often chitin, mannans or glucans Life cycle: 2 phases involving asexual/sexual reproduction to form spores - undergo transitions between uni/multicellular forms
57
What are filamentous fungi?
Molds: bread mold, aspergillus, morel Vegetative form = hyphae - not all form it Mycelium (body) is a section with nuclei, cell wall, pore and septum (branched) - some have just nuclei and cell wall - coenocytic hyphae - unseptated Absorption of nutrient and growth occur at tip of hyphae Can have wide range of morphologies due to complex life cycle - doesn't have to be the same for each species
58
What is the life cycle of filamentous fungi?
Alternate diploid/haploid phases - can form spores which help fungi survive harsh conditions Can spend most of life cycle as haploid or diploid - different between species
59
What are types of basidiomycetes?
Mushrooms: death cap, starfish stinkhorn, magic mushroom, enoki mushroom, oyster mushroom, spiny puffball Ectomycorrhizal basidomycetes are found near tree roots - help plants obtain minerals and in return receive sugars
60
What is the lifecycle of basidiomycetes?
They grow a mycelium from spores during vegetative growth - the cytoplasm of gametes fuse (plasmogamy) which forms fruiting bodies (mushrooms) - nuclear fusion will form basidium (true zygote)
61
What are yeasts?
Single cell fungi: saccharomyces (baker's yeast), crytococcus, candida Economic: S.cerevisiae used in brewery, baking Health: cryptococcus and candida cause infection, some used as probiotics Used as a model organism
62
Where can we use yeast as model organism?
Protein folding Lipid biology Vesicular trafficking and fusion Lysosomal/peroxisomal function Apoptosis Cell cycle Mitochondria
63
How do yeast reproduce?
Cell division by budding/binary fission Some form hyphae under specific conditions (dimorphic) Can be haploid or diploid Haploid (mitosis) -> mate -> cell fusion -> nuclear fusion -> Diploid -> make ascus by meiosis -> ascospores (haploid) -> germination
64
What are the types of algae?
Primary endosymbiotic: green and red from ancestral endosymbiosis (autotrophs) - 2 MEMBRANES Secondary endosymbiotic (diatoms): centric (circular) and pennate (long). From protists that engulfed primary - mixotrophs - 3/4 MEMBRANES Predatory algae - dinoflagellates - secondary - From protists that engulfed primary - mixotrophs - 3/4 MEMBRANES
65
What are the properties and importance of algae?
Ecological importance: component of phytoplankton - produce half of atmosphere's O2 and is a key food item (zooplankton eats phytoplankton) Algae are photosynthetic organisms and have chloroplasts - diatoms are more diverse metabolism
66
What algae is a model organism?
Primary endosymbiotic - Chlamydomonas reinhardtii Model for photosynthesis, motility, cell cycle - doubling time is 6-8 hours at 25 degrees
67
What is the structure of the primary endosymbiotic algae, Chlamydomonas reinhardtii?
Swims with 2 flagella (breast stroke) Contains: - a huge chloroplasts (2 membranes) with chlorophyll a and b - no phycoerythrin - a pyrenoid to stock bicarb which can be converted to CO2 - a contractile vacuole for osmoregulation - cell wall made of hydroxyproline rich glycoproteins - NO CELLULOSE
68
What is the life cycle of the primary endosymbiotic algae, Chlamydomonas reinhardtii?
Mostly found as haploid dividing by binary fission (asexual) Haploid cells from opposite mating types can fuse to form a zygote - loses flagella and grows protective coat The zygote undergoes meiosis to regenerate haploid Some primary endosymbiotic algae have colonial lifestyle: Volvox carteri made of generative or somatic cells - generative cells make colonies
69
What are secondary endosymbiotic algae?
Diatoms Most abundant organisms in phytoplankton - produces 20% of O2 Mobile but no appendages - use actin and myosin on mucus they produce Used in toothpaste Same biomass as rainforests Large morphology diversity and unusual division mechanism
70
What is the structure of the chloroplast in diatoms compared to plants?
Much more complex Has a pyrenoid, encircling lamellae, periplastid reticulum, osmiophilic globules, multiple membranes
71
What is the diatoms structure?
Cell wall called frustule made of silica Two types of diatoms: Radical symmetry: centric Bilateral symmetry: pennate Between 1-2000 micrometres Frustules sediment at bottom of ocean - have usual division due to rigid cell wall Large range of morphologies
72
How do diatoms divide?
Frustules are made of two valves - one overlaps the other like a petri dish Each daughter cell inherits one half of frustule - smaller than parental When size is reached - undergoes meiosis to form gametes After gamete fusion - zygote forms a frustule same size as parental
73
What are diatoms used for?
Treatment against fleas/red mites - dries them out Diatomaceous earth
74
What are coccolithophores?
Secondary endosymbiotic algae Frustules made of calcium carbonate - key role in carbon cycle
75
What are the protists: alveolates and parasitic protozoa?
Alveolates: Ciliates - predatory protists Apicomplexans - parasites Dinoflagellates - predatory protists Parasitic protozoa: Metamonads - symbionts/parasites Trypanosomes - parasites
76
What are the properties and importances of alveolates?
- contain alveoli (cytoplasmic sacs of fluid surrounded by lipid membrane - unknown role) - motile (cilia/gliding) - mostly aquatic Ecological importance: - ciliates play a role in food web (zooplankton) - apicomplexans impact health (malaria) - dinoflagellates are key in carbon cycle
77
What are ciliates?
Alveolates - predatory protists - alveoli under cytoplasmic membrane - cilia and trichocyst (protrusible filaments) - use cilia for motility and to catch prey - enclosed in phagocytic vacuole and dissolved by enzymes - contractile vacuole to regulate water exchanges - digestive vacuoles - 3 nuclei: 2 micro, 1 macro Goes through binary fission and/or conjugation (starts with fusion of 2 micronuclei) - sexual and asexual
78
What are apicomplexa?
Alveolates - parasites Spore-forming parasitic protozoan - no flagella/cilia/pseudopods Contain an apical complex involved in entry into host cell Contains an organelle called apicoplast - degenerate chloroplast (no chlorophyll) which carries out fatty acid metabolism All are endoparasites causing diseases: malaria (Plasmodium falciparum), cryptosporidiosis (Crytosporidium parvum), toxoplasmosis (Toxoplasma gondii)
79
What is the apicomplexa life cycle?
Sporozoite is infectious form Multiply in a structure called schizont which ruptures to release merozoites The merozoites can differentiate into male and female gametocytes that are transmitted to mosquito In mosquito host - gametes fuse - form zygote and invade vector cells to produce sporozoites Diploid in mosquito, haploid in human
80
What are dinoflagellates?
Alveolates - predatory algae Photosynthetic organisms Mixotrophs - use non light/carbon sources except CO2 Responsible for algal blooms which can be toxic Mobile - feed on bacteria, algae, small dinoflagellates, diatoms, ciliates and other dinoflagellates Involved in complex symbiotic/parasitic interactions Life cycle is unknown but mostly vegetative reproduction and can exist as dormant (cysts)
81
What is the structure of dinoflagellates?
2 flagella - 1 wrapped around cell Contain a chloroplast with complex membranes Cell walls made of cellulose plates called thecae Contain an organelle called extrusome - all ciliates have this
82
What are metamonads and trypanosomes?
Parasitic protozoa Mobile parasites that cause disease (diarrhea, sleeping sickness, chagas disease) Mostly harmful but sometimes symbionts (metamonads) Can be transmitted via a vector or direct contact
83
What is a metamonad?
Parasitic protozoa Mixotrichia: Found in gut of specific termites, lives in symbiosis with other bacteria: 1 endosymbiotic bacteria which degrades cellulose, 3 ectosymbiotic bacteria (short/long spirochetes and bacteriodes) providing motility, possess 4 non-functional flagella Giardia lamblia: Human parasite - diarrhea 2 nuclei Adhere to epithelial cells using ventral adhesive disk Divide by binary fission and forms cysts when conditions are unfavourable
84
How is the metamonad typanosome spread?
Through fly (vector) Sleeping disease 2 phases: Acute) fever, headaches, lymph node inflammation Chronic) if not treated - invades CNS and disrupts sleep cycle
85
What are slime molds and amoebas?
Slime molds (pleiomorphic) and amoeba (parasite) - Common predatory organisms in damp environments - Both have similar morphology and share a developmental stage - Differentiate to multicellular reproductive stage when food supply is low - Amoebas are responsible for human diseases
86
How do amoebas move?
Shape is variable Mobility is conferred by pseudopods created by actin polymerisation/disassembly - actin pushes the membrane to form pseudopods Same mechanism as human phagocytes
87
How does slime mold dictyostelium discoideum grow?
Dictyostelium discoideum - cellular slime mold Grows as amoeboid unicellular organism that divides by binary fission Under starvation - produces cAMP - forms slug by plasmogamy - undergoes differentiation into a fruiting body (sporangium and stalk) Spores (cysts) are released to initiate a new cycle
88
How does physarum polycephalum slime mold grow?
Plasmodial slime mold Grows and divides by binary fission as an amoeboid single cell Individual cells can aggregate to form plasmodium - single cell Plasmodium undergoes differentiation into fruiting body (sporangium) Spores (cysts) are released to initiate a new cycle Slime mold cell walls are not made of cellulose
89
What are the different phenotypic diversities between bacteria?
Morphology Size Colour Smell
90
What are the different morphologies of bacteria?
Cocci e.g. staphylococcus aureus and streptococcus pneumoniae (circular) Rods e.g. escherichia coli (round cylinder like a pill) Curved e.g. vibrio cholerae (like a boomerang) Spiral e.g. titanospirillium velox and borellia burgdorferi (like a hair curl) Exotic e.g. stella humosa (like 2 stars attached together) and aloquadratum walsbyi (square)
91
Can bacteria morphology change?
Yes! - during the cell cycle e.g. Caulobacter crescentus - unequally divides into a swarmer and stalked
92
Is the bacteria size important?
Yes! Thiomargata magnifica - 1cm Mycoplasma pneumoniae - 0.2 micrometres Means a large SA:V - nutrient exchange and growth rate, higher intracellular nutrient conc, rapid evolution due to a high selection rate of mutations
93
Can bacteria be coloured?
Yes! - produce pigments Serratia marcescens: pigment = prodigiosin - pink -> immunosuppressant Staphylococcus aureus: pigment = staphyloxanthin - yellow -> antitoxin and detoxify ROS Chromobacterium violaceum: pigment = violacein - purple -> antitoxin and detoxify ROS Pseudomonas aeruginosa: pigment = pyocyanin - green/blue -> cytotoxicity, neutrophil apoptosis, ciliary dysmotility, proinflammatory
94
Do bacteria produce smell?
Yes! e.g. degrade apocrine secretions - human odors. Leucine -> isovaleric acid (short FA) by staph, propanoic production by propionibacteria e.g. decarboxylation of amino acids to produce polyamines - putrescine, spermidine, cadaverine - role in bacterial physiology - ROS scavenger and signalling
95
What is the gram stain?
Stain: Crystal violet Fix: Iodine Wash: alcohol Counter stain: Safranin Can show differences in gram positive vs negative
96
What is the difference in the staining of gram positive vs negative?
Positive - no outer membrane, thick cell wall thin, really thick Negative - outer membrane, thin cell wall - thin, thick, thin Some exceptions e.g. mycobacteria
97
What are s-layers?
Facultative structures (not in most model organisms) Non-covalently bound to cell surface Proteinaceous crystalline arrays: self assembly
98
What are capsules in bacteria?
Most are made of polysaccharides Some made of AA's (poly-y-D-glutamate) Covalently bound to cell wall or outer membrane Confer resistance to host phagocytes/bacteriophages and keep environment hydrated Can be seen with indian ink stain - halo
99
What are exopolysaccharides?
Homo/hetero polysaccharides - non covalently attached to cell surface Important for biofilm formation, some economically important e.g. xanthan gum - ice cream, toothpaste, salad dressing
100
What is the outer membrane in gram negative bacteria?
Asymmetric membrane Made of: - phospholipids - proteins e.g. porin - lipoproteins (Lpos/braun lipoprotein) covalently linked to peptidoglycan - LPS (endotoxin) - activates immune system (septic shock) LPS - O-specific polysaccharide, core polysaccharide and lipid A
101
What is peptidoglycan (murein)?
Makes the cell wall - also has teichoic acid and lipoteichoic acid Roles: exoskeleton (resistance to osmotic stress), cell shape and scaffold for display of polymers and proteins Protective role and acts as a sieve to regulate exchanges - not rigid (elastic)
102
What is the composition of peptidoglycan?
Glycan chains of alternating N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid (MurNAc) - bound to pentapeptide stem This is substituted by short peptides (L and D aas) - contains high proportion of proteins Involves glycosyl transferase and transpeptidase activity Not found in fungi Composition doesn't change Multiple layers in gram positive Assemble via peptide bonds
103
What is the cytoplasmic membrane in bacteria?
Key components: - phospholipids - hopanoids (equivalent to sterols in eukaryotes) - protein (transporters/signals - for ion, protein and nutrient transport - selective permeability)
104
What are the phospholipids in the cytoplasmic membrane of bacteria?
Polar head (alcohol and phosphate) -- glycerol derivative -- fatty acid Phosphatidyl ethanolamine Phosphatidyl choline Unsaturated fatty acids modulate fluidity and permeability
105
What are hopanoids?
Pentacyclic lipids in bacterial cytoplasmic membrane which modulate membrane fluidity and permeability Equivalent to cholesterol in eukaryotes
106
What are the chromosomes in bacteria?
ALWAYS dsDNA Single circular chromosomes in most bacteria - can be 0.5 Mbp -14.8 Mbp Can have more than one Can be linear Organised as a nucleoid (histone-like protein - supercoiling)
107
What are the plasmids in bacteria?
Always dsDNA - usually circular but can be linear Variable copy number (1 to hundreds) - number of copies in a cell Between 2 kbp - 600 kbp Can be self-transferable (horizontal transfer) - conjugation Carry resistance genes
108
Do bacteria have introns?
NO! Continuous coding sequence = open reading frame (ORF)
109
What is an operon in bacteria?
One promotor - multiple open reading frames
110
Are genes bigger in eukaryotes?
Yes! E.g. human dystrophin = 2.2 Mbp -3500 residues, E.coli Ihr = 4.6 kbp - 1538 residues
111
What does gene expression (transcription) work in bacteria?
1) RNA polymerase haloenzyme scans DNA forming loose complex 2) The sigma factor binds to 2 specific sequences upstream from start codon - closed complex 3) The DNA is unwinded - allows formation of open complex - transcription starts and sigma factor released 4) Termination requires either: Rho independant - palindromic GC rich region upstream from AT rich sequence - one GC rich region is transcribed, forms hairpin that makes RNA polymerase fall apart - helped by AT rich region - few H bonds Or: Rho proteins (275 kDa hexamer) recognises and binds 72 residues GC rich It has RNA dependent ATPase activity - wraps the RNA around itself - one it reaches polymerase, Rho unwinds RNA-DNA duplex in the polymerase and releases RNA polymerase
112
How does gene expression (translation) work in bacteria?
Similar to eukaryotes but the ribosomes are different. Prokaryotes (70s) = 50s (31 proteins) + 30s (21 proteins) Eukaryotes (80s) = 60s (50 proteins) + 40s (33 proteins) 80s - mRNA bind in absence of tRNA, 40s subunit is guided by 5' cap 70s - interact better with mRNA in presence of tRNA, 30s recognises shine-delgarno sequence Transcription and translation are coupled in prokaryotes Translation is inhibited by cycloheximide in eukaryotes
113
How is transcription different in eukaryotes than prokaryotes?
Eukaryotes - in nucleus, prokaryotes - in cytoplasm Eukaryotes - 3 RNA pol, Prokaryotes - 1 Eukaryotes termination involves AAUAAA (mRNA cleavage) mRNA is modified in eukaryotes
114
What is needed for metabolism in bacteria?
Energy source + source of electron + carbon source Energy = light/ preformed molecules Electrons = organic (oxidise organic molecules), inorganic (oxidise inorganic e.g. Fe2+ (ferrous iron) --> Fe3+ (ferric iron), NO2- (nitrite) --> NO3 (nitrate), S (sulfur) --> SO42- (sulfate)) Carbon = organic (aas, carbohydrates), inorganic (CO2)
115
How do we name bacteria in terms of metabolism?
Energy source: Light (photo), preformed molecules (chemo) Electrons: organic (organo), inorganic (litho) Carbon: organic (heterotroph), inorganic (autotroph)
116
How can we measure the growth of microorganisms?
Counting cells or measuring cell biomass. More conveniently - monitoring the turbidity/optical density (we can use a spectrophotometer - cells scatter the light, more cells = more scattering = increases optical density)
117
How can we test the susceptibility of bacteria to antibiotics?
Disc diffusion assay Etest Minimal inhibitory concentration assay Antimicrobial assay
118
What is the disc diffusion assay?
Bacteria spread on plate - form a lawn of cell On lawn - place thin paper disks with antibiotics If cells are susceptible - zone of inhibition - diameter can be measured
119
What is etest?
Spread bacteria on agar = lawn of cells Place a strip with gradient of antibiotic concentrations on surface The minimum inhibitory conc can then be read
120
What is the minimal inhibitory concentration assay?
Time consuming and uses liquid culture. A growth media with various conc of antibiotics is inoculated with 5 x 10^5 colony forming units per ml. Growth is then monitored for 24/48 hours.
121
What is an antimicrobial assay?
Semi quantitative - not used in hospitals. Bacteria are serially diluted - each dilution is spotted on the surface of agar containing an antimicrobial compound at a given conc
122
What are the physical antimicrobial controls?
Heat Irradiation Filtration
123
How methods use heat kill bacteria?
Moist heat (boiling water/autoclave) - 15 minutes at 121 degrees under pressure is required for spores Dry heat (oven) - direct flaming, incineration, - 150 degrees for 2 hours Pasteurization (mild heat/HTST/UHT) - Can't kill spores - temperature below 100 degrees to avoid casein aggregates, HTST = 72 degrees for 15 seconds - kills 99.999% of microorganisms in milk, UHT = 140 degrees, 2-5 seconds
124
What is the thermal death point?
Thermal death point - minimum temperature where all organisms are killed in 10 minutes
125
What is the thermal death time?
Minimal time required to kill all bacteria in a liquid at a given temperature
126
How does irradiation kill bacteria?
Ionizing radiations (x rays and gamma rays) - food industry/ medical and lab equipment - destroys DNA via double strand breaks + ROS Non ionizing radiations (UV) - surface decontamination, DNA damage (breaks/dimers) Energy is inversely proportionate to wavelength
127
How does filtration kill bacteria?
Used to sterilize gases or liquids that can be damaged by heat The porosity of filters can be chosen for specific applications (1mm - 0.01um) e.g. nucleopore, membrane filter
128
Which are the different effects of chemicals on bacteria growth?
Bacteriostatic - stops cells from reproducing Bacteriocidal - kill bacteria - number of cells stay the same Bacteriolytic - kill bacteria - both number of cells and viable cell count decrease
129
What are the different classes of chemical agents?
Destroy on objects Sterilants: completely destroy all forms of microorganism, including spores. e.g. ethylene oxide Disinfectants: kill microorganisms but not endospores e.g. alcohol Destroy on living tissue: Antiseptics/germicides: inhibit/kill microorganisms e.g. handwash
130
How do we measure antimicrobial activity?
Disc diffusion technique: Inocculate plate with liquid culture - discs containing antimicrobial agents are placed on surface - shows susceptability if there are zones of inhibition or can do etest Spot assays: Bacteria are serially diluted - each dilution is spotted on the surface of agar containing an antimicrobial compound at a given conc Minimum inhibitory concentration: Lowest conc of a drug inhibiting the visible growth of a test organism after overnight incubation - use liquid cultures and add different conc of agent - see which is clear Minimum bactericidal concentration: Lowest conc of a drug killing >99.9% of a test organism after overnight - better indicator
131
What is MIC and MBC?
Minimum inhibitory concentration: Lowest conc of a drug inhibiting the visible growth of a test organism after overnight incubation - MEASURES GROWTH INHIBITION Minimum bactericidal concentration: Lowest conc of a drug killing >99.9% of a test organism after overnight incubation - MEASURES DEATH - better indicator
132
Which chemicals are used for antimicrobial control?
Phenolic: can be local anaesthetic at low concs, high conc - antimicrobial - disrupts cytoplasmic membrane and denatures proteins e.g. chlorhexidine (mouthwash), triclosan Alcohols: denature proteins, lipid solvent, disrupts membrane - active conc = 60-85% Aldehydes: alkylating agents which modify proteins and DNA - cell death e.g. formalin/formaldehyde, glutaraldehyde Quaternary ammonium compounds 'quat': interact with phospholipids of cytoplasmic membrane Halogen releasing agents: can be chlorine/iodine releasing
133
Is temperature required for growth?
YES Temperature: cardinal temperatures - minimum (enzymatic reactions at increasing rate - low = membrane gelling), optimum, maximum (protein denaturing) Different classes have different cardinal temps: psychrophile (4 degrees), mesophile (E.coli 37 degrees), thermophile (TAQ 70 degrees), extreme thermophile (P.fumarii - 106 degrees)
134
How are psychrophiles adapted to cold temperatures?
- Increased membrane fluidity - high amount of unsaturated/polyunsaturated and methyl branched fatty acids - shorter acyl-chain length - Production of Anti-Freeze proteins (AFPs) - these bind to small ice crystals to inhibit their growth by covering water accessible surfaces of ice - Produce cyroprotectants - trehalose and exopolysaccharides - Produce cold adapted enzymes - more a helices, less weak bonds and interdomain interactions - greater flexibility
135
What is an example of a psychrophile?
Polaromonas vacuolata in Lake Bonney, Antarctica
136
How are thermophiles adapted to high temperatures?
- Protects genome: stabilises DNA by DNA-binding proteins, DNA gyrases introduce supercoils, have high GC% - resistant to denaturation - Membrane composition: ether linked phospholipids, single lipid layer (glycerol tetraethers) - Produces thermostable proteins: increases ionic interactions and hydrophobic interactions - Thermostable chaperonins: thermosome in Pyrodictium abyssi
137
What is an example of a thermophile?
Thermus aquaticus in Yellowstone
138
Is pH needed for growth?
Yes! Can be a neutrophile, acidophile (volcanoes, fruit, gastric juices) or alkaliphile (limes, seawater, ammonia)
139
What are acidophiles metabolic adaptations?
Membrane is impermeable - tetraether lipids There is a H+ motive force which powers motility, substrate symport and ATP synthesis H+/Na+ antiporters help maintain pH lower than external K+/H+ antiporters excrete H+ Respiratory chains pump H+ Reverse membrane potential
140
What are alkaliphiles metabolic adaptations?
Na+ motive force drives motility, ATP synthesis and substrate symport Decarboxylases secrete Na+ Respiratory chains pump Na+ Na+ driven ATPases export Na+ H+/Na+ antiporters scavenge proteins
141
Is osmotic pressure needed for growth?
Yes! Extreme halophile (2M<) e.g. Halobacterium salinarum - high salt conc Halophiles (0-2.5 M - optimum is 1) e.g. Aliivibrio fischeri - requires high salt concentrations and thrive in salt lakes Halotolerant (high at 0 - decreases to 2.1M) e.g. Staphylococus aureus - can survive in salty environments but don't need salt to grow Nonhalophile e.g. E.coli
142
How are bacterial cells adapted to osmotic conditions?
Response to osmotic stress: - regulate water movement by passive transport and aquaporins - produce compatible solutes (proline, glutamic acid) - release solutes by mechano-sensitive channels Salt requirement in halophiles - needed for stabilization of S-layer glycoprotein by Na+, accumulation of k+ as a compatible solute (>4M in cell)
143
Do bacteria need nutrients to grow?
Yes! Needs: nitrogen, sulfur, phosphorus, vitamins, K+/Ca2+/Mg2+ (cofactors), trace elements (Fe/Cu/Zn)
144
Do bacteria need oxygen to grow?
Yes! - for metabolism There are toxic forms of ROS that may form: superoxide (O2-), Hydrogen peroxide and hydroxyl radical Catalase/peroxidase: convert H2O2 into H2O Superoxide dismutase/reductase + catalase: convert O2- into H2O2 then H2O
145
What are the requirements for oxygen?
Obligate aerobes - use exclusively O2 - need catalase and superoxide dismutase Facultative aerobes (E.Coli) - can use O2 - need catalase and superoxide dismutase Microaerophiles - require O2 Anaerobes aerotolerant - no O2 - need superoxide dismutase Obligate anaerobes - die if used oxygen
146
How can we measure bacterial growth?
Direct measuring: microscopic counting, viable counting (serial dilution so can count), flow cytometry (light scattering - cell density - can distinguish between living and dead) Indirect: optical density (measures unscattered light - needs high cell densities, doesn't distinguish live vs dead cells, OD differs between organisms, doesn't work with molds and filamentous bacteria), dry weight, metabolic activity Bacterial growth curve: exponential growth
147
What are the 4 phases of bacterial growth?
1) Lag phase - metabolism starts but NO DIVISION 2) Log phase - exponential increase in population 3) Stationary phase - microbial death balances new cells 4) Death phase - population decreases
148
How are halogen releasing agents used as an antimicrobial agent?
Two types: Chlorine releasing agents - sodium hypochlorite (bleach) - forms chlorinated bases in DNA and oxidises proteins. Hypochlorite ion OCl- is in equilibrium with hypochlorite acid HOCl. Iodine releasing agents - very powerful but stain, targets DNA and proteins
149
What are some major pandemics?
Black plague - 1328-1350 - killed 30%-50% of the population in europe 1918 flu pandemic - killed 3-5% of world population (50-100 million) 2020 COVID pandemic - 400 million cases, 6 million death
150
What are the two therapeutic strategies for pandemics?
Antibiotics (infections) and vaccinations
151
Who did germ theory?
Louis Pasteur in the 1860s - the concept of antibiotics emerged
152
What did Robert Koch do?
He established that relationship with microbes and disease - 1890. 1) The microorganism must be found in all suffering organisms - not in healthy. 2) Must be isolated from diseased organism and grown in pure culture. 3) This should cause disease when introduced in healthy organism. 4) The microorganism should be reisolated and identified as being identical to original.
153
What are some historical points in the development of antibiotics?
1888: Emmerich and Low - Pyocyanase showed antibacterial activity - unstable, toxic. 1910: Ehrlich: Salvarsan (dye linked to arsenic - magic bullet) - treated syphilis - very painful and toxic side effects 1928: Fleming - penicillin formed from fungi. 1932: Domagk - protosil red
154
What are the major classes of antibiotics?
Can effect cell wall synthesis, folic acid metabolism, DNA gyrase, RNA elongation, Proteinsynthesis (30s/50s subunits), lipid biosynthesis
155
What are the four modes of action of resisting antibiotics?
- Drug inactivation - Target modification - Efflux/impermeability - Bypass
156
Is antibiotic resistance effecting the world?
Yes! 1.4 million died with TB in 2019 700,000 deaths a year from resistant bugs - by 2050 ~ 10 million a year - cost economy £75 trillion
157
How is antibiotic resistance effecting Europe and the US?
European Centre for Disease Prevention and Control (2015 - 2019) - 670,000 infections due to antibiotic resistant bacteria - 33,000 deaths - annual loss of £1 billion US WHO data 2019 - 3 million infections due to antibiotic resistant bacteria - 50,000 deaths (700,000 worldwide) from drug-resistance pathogens
158
When were antibiotics used and when was a resistant strain found?
Some have some really soon, some takes years but always has a resistant bacteria eventually
159
What causes antibiotic resistance?
1) Antibiotic misuse in human therapeutics 2) Farming: started in the 40's, animals are fed with antibiotics - 4 x more than humans, the dose required has increased by 10-20x 3) Agriculture: treatment of diseases in trees and plants (oxytetracycline/streptomycin) 4) Aquaculture: 2g/ton (Norway) to 150g/ton (Canada) 5) Pets
160
What is the mass of antibiotics that humans intake?
10g
161
What are the properties of an ideal antibiotic?
1) Target - selective toxicity and must inhibit an essential process/inhibit virulence 2) Stability and effectiveness - pharmacokinetics - what does the body do to the antibiotics, pharmacodynamics - what does the antibiotic do to the body 3) Cost
162
What do beta lactams do?
Inhibit peptidoglycan polymerisation mediated by D,D-transpeptidases (penicillin binding proteins) Cheap, target both gram +/-
163
What are the structures of B-lactams?
Structural analogs of D-Ala-D-Ala C-terminal residues in the peptide stem They are used by Penicillin binding proteins as substrates and inactivate these enzymes irreversibly - form covalent complex penicillin-PBP
164
How are bacteria resisting B-lactams?
1) Drug inactivation - B-lactamases 2) Target modification - low affinity PBP/overexpression of PBP targeted by B-lactams (so not all are inhibited and peptidoglycan can still be formed) 3) Efflux - efflux systems (Gram negative) 4) Bypass - alternative pathway
165
What is the mechanism of inactivation by B-lactamases?
Nucleophilic attack by catalytic serine on B lactamase This forms a covalent complex penicillin-B-lactamase Water is added - penicillin hydrolysis
166
How do gram negative bacteria secrete antibiotics?
E.g. P.aeruginosa Overproduction of MexAB-OprM system - carbapenam resistance Overproduction of MexEF-OprN system - imipenem resistance
167
How do bacteria modify the synthetic pathway targeted by B-lactams?
L,D-transpeptidation is B-lactam insensitive - no D-ala-D-ala 3- crosslink and not 3-4 crosslink This causes resistance
168
What is a antibiogram?
Diffusion disk assay In a hospital - tests antibiotics on patients blood
169
How do we name metabolic types?
Energy: Preformed molecules (redox) - chemo Sunlight - photo Carbon: Organic (aas/carbohydrates) - heterotroph Inorganic (CO2 - NO C-C/C-H bonds) - autotroph
170
What are examples of the metabolic types - just energy and carbon?
Chemoheterotroph - animals and fungi Chemoautotroph - extremophiles Photoheterotrophs - purple and green non-sulphur bacteria Photoautotrophs - plants, algae, cyanobacteria
171
What are the two sources of electrons for respiration in metabolism?
Organo: Organic - e.g. CH2O - have C-H/C-C bonds Litho: Inorganic - e.g. Fe2+ (ferrous ion) -> Fe3+ (ferric ion) NO2- (nitrite) -> NO3- (nitrate) S (sulfur) -> SO42- (sulphate) 2H20 -> 4H+ + 4e + O2
172
What are examples of all metabolic types?
- Chemoorganoheterotroph = animals - Chemoorganoautotroph = some archaea - Chemolithoheterotroph = sulfur bacteria - Chemolithoautotroph = bradyrhizobiaceae - Photoorganoheterotroph = heliobacteriaceae - Photoorganoautotroph = x - Photolithoheterotroph = rhodobacteraceae - Photolithoautotroph = algae and plants - often shortened to photoautotroph
173
What happens to the sources in metabolism?
Chemoorganotrophy (reduced organic molecules) Chemolithotrophy (reduced inorganic) Phototrophy (sunlight) These are broken down - catabolism and form ATP This is coupled to anabolism - long term energy storage, biosynthesis, C/N2/H2O
174
What is the photosynthesis and respiration equations?
Co2 + H2O -> O2 + sugars -> H2O + CO2
175
What are the electron donors in metabolism?
Chemoorganotrophy: organic Chemolithotrophy: inorganic Phototrophy: use light energy to reduce compounds, then use these as an electron donor
176
What are the electron acceptors in metabolism?
Respiration: inorganic or organic molecules Fermentation: organic molecules
177
What is the electron transfer system?
NADH donates electron to cytochrome C - this conserves energy (E) in the form of a transmembrane PMF used for ATP synthesis From low to high redox potential O2 is the electron acceptor
178
What is the change in reduction potential?
From -340 mV - 800 mV Delta g = -nF x change in energy n = number of e F = faraday constant The PMF is 180 mV
179
What sources of energy does metabolism form?
Reducing power (NADH/NADPH/FADH2) ATP
180
What is the compounds produced and used in chemoorganotrophy?
Used: organic compounds e.g. carbohydrates, lipids, peptides, aromatic compounds 2 metabolites produced: Acetyl-CoA and pyruvate Distinct pathways (Glycolysis, Entner-Doudoroff, Pentose Phosphate Pathway) lead to key metabolites and energy currencies: ATP/NADH/FADH2 Example of glucose metabolism
181
What is the different between anaerobic respiration and fermentation?
Both have no oxygen Anaerobic is where there's an alternative inorganic electron acceptor or organic via a membrane bound respiratory chain. ATP produced by oxphos via PMF Fermentation is where there's an organic acceptor but NO RESPIRATORY CHAIN - ATP is produced by substrate level phosphorylation in cytoplasm
182
What are the three major metabolic types depending on the electron acceptor in chemoorganotrophs?
- oxygenic respiration - anaerobic respiration - fermentation
183
What is oxygenic respiration?
High energetic output Glycolysis Link reaction Kreb cycle Oxidative phosphorylation
184
What is anaerobic respiration?
A wide range of inorganic compounds used as electron acceptors E transport occurs via cytochromes, quinones and iron-sulfur proteins Bacteria usually use anaerobic when no O2 is available Depending on redox potential of acceptor - different amounts of energy are generated
185
Why is anaerobic respiration important?
To exploit a wide range of ecological niches e.g. aquatic water quality Use different electron acceptors at different points in the ocean
186
What is denitrification?
Respiration: Nitrate -> nitrite -> nitric oxide -> nitrous oxide -> N2
187
What is methanogenesis?
Acetate CH3-COO + H+ -> CH4 + CO2 Methanol 4CH3OH -> 3CH4 + CO2 + 2H2O
188
What is fermentation?
Under anaerobic conditions when no electron acceptor is available ATP produced by SLP - not oxphos Energy yields are low - cells grow more slowly
189
What are the major sources of electrons in chemolithotrophs?
H2 - hydrogenotrophy, sulfate reduction, methanogenesis Fe2+ - iron oxidation NH3 - anammox, ammonia oxidation NO2 - nitrification HS - sulfide oxidation S - sulfur oxidation
190
What do chemolithotrophs use?
Use CO2 as a carbon source to produce organic molecules via calvin cycle Also produce more complex molecules - acetate To fix carbon they require NADH, this needs the consumption of H+ for a reverse electron flow process
191
What is CFU?
Colony forming unit Number of viable microorganisms
192
What metabolic types do chemoorganotrophs?
Oxygenic respiration and fermentation
193
Can chemolithotrophs use H2 as an electron donor?
Yes - hydrogenotrophy H2 is a great electron donor that uses a wide range of acceptors O2 as acceptor: H2 + 1/2O2 --> H2O SO42- as acceptor: 4H2 + SO42- + H+ --> HS- + 4H2O CO2 as acceptor (methanogenesis): 4H2 + CO2 --> CH4 + 2H2O Chlorinated compounds as acceptors (dehalorespiration): Dehalobacter (tetrachloroethene -> trichloroethene -> dichloro ethene) to Dehalococcoides (dichloroethene -> vinyl chloride -> ethene) Formate and acetate can also be used - yields CO2 and CH4
194
Can chemolithotrophs do iron oxidation?
Yes! Fe2+ can be oxidised to Fe3+ at a low pH 2Fe2+ + 1/2O2 + 2H+ --> 2Fe3+H2O Ferric ions (Fe3+) can form insoluble ferric hydroxide as pH gets lower: Fe3+ + 3H2O --> Fe(OH)3 + 3H+ Acceptors other than Oxygen can be used e.g. nitrates to nitrites
195
Can chemolithotrophs do sulfur oxidation?
Some sulfur derivatives can be used as electron donors to form sulfuric acid. H2S (hydrogen sulfide) -> S (elemental sulfur) -> S2O32- (thiosulfate) -> H2SO4 Acid producing microbes can be used in biomining e.g. Acidothiobacillus ferrooxidans - oxidise iron and copper - Cu+ and acid dissolves the metal from rocks
195
Can chemolithotrophs do nitrogen oxidation?
Yes! Ammonia and nitrites can be used as electron donors to produce nitrates Nitrification: NH4+ -> NH2OH (this bit by nitrosomonas) -> NO2- -> NO3- (nitrosbacter) Occurs in aerobic conditions Anammox: NH4+ + NO2- -> N2 + 2H2O (planctomycetes) Occurs in anaerobic conditions
195
What are the two types of photosynthesis?
Oxygenic: cyanobacteria and plants - PSI + PSII Anoxygenic: bacteriorhodopsin, green sulfur bacteria, purple bacteria - BR, PSI (green), PSII (purple)
196
What is bacteriorhodopsin?
A primitive photosynthetic system Very abundant light-driven proton pump in archaeal membrane Contains a pigment (retinal) that undergoes conformational change when excited by light The movement of H+ generates gradient used to produce ATP
197
How does bacteriorhodopsin work?
Excitation of retinal induces conformational change: trans -> cis This triggers transfer of proton to Asp85 Deprotonated retinal pushes against helix F - opens cytoplasmic channel - induces reprotonation of retinal from Asp96 Asp96 is reprotonated Asp 85 transfers H+ outside through hydrogen bonding with water
198
What is cyanobacteria?
Oxygenic photosynthesis No chloroplasts - are primitive chloroplasts Often made of thylakoids - sometimes does not
199
How does cyanobacteria work?
Light is captured by light harvesting complexes (contain chlorophyll, carotenoids, bilins) - channel energy to reaction centre 'Z pathway' - 2 distinct photosystems are excited by light - light provides energy to strip e from H20 - forms H+ - electron flow is used to pump H+ outside to form NADPH - H+ gradient forms ATP - NADPH + ATP are used to fix CO2
200
What are green sulfur bacteria?
Anoxygenic photosynthesis - light captures by antenna complexes in organelles called chlorosomes - photon energy is transferred to PSI - PSI donates e to ETC - electron transport pumps H+ outside the cell and reduce NADP+ via ferredoxin - H+ gradient forms ATP -PSI receives electrons from inorganic sulfur derivatives (H2, H2S)
201
What is purple bacteria?
Anoxygenic photosynthesis - light captured by antenna complexes called chromatophores - photon energy is transferred to PSII reaction centre - PSII donates e to a cyclic ETC - e transport pumps H+ outside cell - generates ATP - cyclic phosphorylation - NADH is produced by reverse electron flow - this is where e is transferred from reduced components with higher redox potential - not energetically favourable - needs energy - e transferred to NAD by ETC - replenished by inorganic/organic compounds (H2S/succinate)
202
What are the two major archaea phyla?
Euryarcheota Crenarcheaota (part of the TACK superphyla) Most are adapted to extreme conditions - extremophiles
203
What is the ultrastructure of archaea?
S layer Pseudomurein cell wall (not always) Other unusual cell envelope structure/organisation Cytoplasmic membrane
204
What is the role of the s layer in archaea?
Cell shape - Can be glycosylated
205
What is pseudomurein?
In archaea cell wall - not always present Heteropolymer - disaccharide-peptides - has N-acetyltalosaminuronic acid Similar to bacteria peptidoglycan Resistant to: - lysosomes - most antibiotics targeting peptidoglycan synthesis (penicillin)
206
What unusual cell envelope structures may archaea have?
Sulfolobus -> Archaellum - like a flagellum Pyrodictium abyssi -> Cannulae Altiarcheum hamiconexum -> Hooks The organisation of s layer and pseudomurein can be different - some don't have pseudomurein, some have pseudomurein above/below s layer
207
What is the cytoplasmic membrane of archaea?
Phospholipids contain no fatty acids - instead have isoprenes These are ether-linked - NOT ESTER They are more stable than bacterial cytoplasmic membranes Some contain diglycerol tetraether lipids Can be present as mono and/or bilayers - always present
208
What is the chromosome organisation and replication in archaea?
- circular chromosome and plasmids - histones - multiple replication origins - encode polymerase B (eukarya) and polymerase D (specific to archaea)
209
What are the polymerases?
Polymerase B - eukarya Polymerase B & D - archaea Polymerase C - bacteria
210
What is transcription like in archaea?
Has a single RNA polymerase - similar to eukaryotic RNA pol II Has introns Genes are organised in operons - polycistronic (multiple proteins from one mRNA)
211
What is translation in archaea?
Coupled to transcription Involved several translation factors like eukarya Ribosomes are 70 S particles
212
Where are archaea found?
Lots of habitats - Antarctica, volcanoes. hot springs, hydrothermal vents
213
What are archaea hyperthermophiles?
Include Crenarchaeota and Euryarchaeota High growth temp (80-120) Most require elemental sulfur for growth Often acidophiles (pH 1-3)
214
What are Acidianus infernus?
Hyperthermophile - crenarchaeota Isolated in geothermic hot spring - optimal growth at 75 and pH 2.5-3 Grows anaerobically (produces H2S) or aerobically (produces H2SO4)
215
What is the role of sulfate-reducing archaea?
Produces H2S which is highly soluble in oil. Increases sulfur emissions when oil is burned and the cost of refining oil. Sulfur also attacks metal in pipelines - causes leaks and corrosion.
216
What is Thermococcus barossi?
Archaeal hyperthermophile Isolated from a hydrothermal vent - optimal growth at 82 and pH 2.5-3 Grows anaerobically - requires sulfur
217
What are halophiles?
Mostly Euryarchaeota Found in evaporating ponds, dead sea, great salt lake. Require up to 5M NaCl for growth
218
What is halobacterium halobium?
Archaeal Halophile Prevalent species in Great Black Lake (4M salt) Optimal growth at 75/pH 2.5-3 Uses light as energy source to make H+ gradient - make ATP Other transporters (halorhodopsin) ensure ion transport
219
What are methanogens?
Archaeal Euryarchaeota Found in anaerobic environments Use acetate/formiate/CO2 as electron acceptor e.g. CO2 + 4H2 --> CH4 + H2O
220
What are examples of methanogens?
Methanopyrus kandleri - isolated in deep ocean on hydrothermal vent - optimal growth at 105-115 - growth conditions = high pressure. We can use OD/production of methane to see growth Methanobrevibacter smithii - prevalent species in human gut, contributes to removal of bacteria end products of fermentation.
221
What are archaea most related to: bacteria or eukarya?
Eukarya Some studies say eukarya evolved from them
222
How many viruses are in marine sediment, seawater and the human gut?
Marine sediment - may be over one million Seawater - may be ~ 5000 types Human gut - may be over 1000
223
What are the general properties of viruses?
- Viruses are obligatory particles: need to hijack host metabolic machineries to replicate - Small - Made of a nucleic acid genome surrounded by a protein coat (capsid) and facultative lipid membrane (envelope) - Viruses exist which infect all living organisms: archaea, bacteria (bacteriophages), animals, plants, fungi, protists, viruses (virophages)
224
What are the nucleic acids in viruses?
Genome composition: DNA - ss or ds, RNA - ss(+/-) or ds Size: usually between 2-20kb (1.2Mb for megaviruses) - minimal info required - only specific genes needed for hijacking. DS>SS, DNA>RNA Organisation: usually 1 molecule - can be fragmented
225
What is the capsid like in viruses?
Surrounds nucleic acid (both together = nucleocapsid) Composition: made of proteins (1 or more) called capsomers Structure: self assembly products - highly ordered - can be - icosahedral symmetry (spherical viruses like mastdenovirus) - helical symmetry (rod shaped viruses like ebola)
226
What is the envelope like in viruses?
Surrounds the nucleocapsid Composition: made of lipid bilayer with glycoproteins from host or virus encoded Role: allows entry into host cell (fusion/endocytosis) E.g. SARS-Cov2 spike protein binds to ACE-2 - modulates angiotensin II activity - increases BP and inflammation
227
What is a bacteriophage?
Virus Contains a mixture of icosahedral/filamentous structures Head Collar Tail Tail pins Endplate Tail fibers
228
What is an archaea phage?
Virus Lots of different morphologies
229
What is a non symmetrical virus?
Complex virus e.g. poxyviridae (smallpox)
230
What is the problem with naming viruses?
Named in lots of different ways - After disease: measles - After place reported: ebola - After host/signs: tobacco mosaic - After shape: coronaviridae (crown) - After discover: Epstein-Barr - After supposed transmission: Influenza - influence of bad air - Combination of above
231
What is the Baltimore classification?
Group I, II and VII - DNA viruses Group III, IV, V, VI - RNA viruses Takes into account: nature of genome RNA/DNA, type of RNA/DNA (ds/ss/polarity), genome replication mechanism By Baltimore, Dulbecco and Temin
232
What are the Baltimore classification group genome replications?
Needs host to replicate I - classical semiconservative - transcribes minus strand to form (+) mRNA II - classical semiconservative, discard negative strand (make itself ds then transcribe to make a + mRNA) VII - transcribed into RNA - forms mRNA (+) - followed by reverse transcription III - make ssRNA (+) then transcribe from this to give ssRNA (-) partner - forms (+) mRNA IV - use ssRNA+ directly as mRNA (+) - make ssRNA (-) and transcribe from this to give ssRNA genome (+) V - make ssRNA (+) and transcribe from this to give ssRNA (-) genome VI - reverse transcribe into dsDNA then make ssRNA (+) genome by transcribing (-) strand of dsDNA ALL FORM mRNA (+)
233
What are some examples of the Baltimore classification groups?
I - Herpesvirus, HBV, Poxviruses, Adenoviruses, Papillomaviruses II - Parvovirus B19 III - Rotavirus IV - HCV, Poliovirus, Rhinovirus, Coronavirus, West-Nile-Virus V - Influenzavirus, Rabies Virus, Measles Virus, Respiratory Synctial Virus VI - HIV, Human T-cell Leukemia Virus VII - Hepatitis B Virus
234
What is the summary of the classes of the Baltimore classification?
7 classes: 1 ssDNA, 2 dsDNA, 2 ssRNA(+), 1 ssRNA, 1 dsRNA
235
What does the lifecycle of viruses require?
A positive strand of RNA
236
What is an example of a class I virus?
dsDNA - no envelope - 5.3-8 kb Papillomavirus - warts and cervical cancer 19 million new cancer cases, 9.9 million deaths (2020) - 15% due to infectious agents ~ 600,000 cases of cervical cancer in 2020 - 95% due to HPV - 342,000 deaths Vaccination campaign in the UK (9-14 year old girls) - offered to Y8 boys in 2019 - 4 vaccines
237
What is an example of a class II virus?
ssDNA - no envelope - 5 kb Adeno-associated virus - harmless
238
What is an example of a class III virus?
dsRNA - no envelope - 18-31 kb Reovirus - causes rotavirus (severe gastroenteritis) 2 million hospitalization (1986-2000), 530,000 deaths in 2000, 215,000 deaths in 2013 In the UK - rarely fatal but has economic burden There is a vaccine - protects against 90% of strains - 1st dose before 8 weeks - 2nd dose 1 month later
239
What is an example of a class IV virus?
(+) ssRNA Foot and mouth (no envelope - 7.5kb) - blisters in cloven hoofed animals - aphtovirus 2001 outbreak - 2000 cases - 4 million cattle died - EU imposed worldwide ban on british exports of lifestock - Cost >3 billion to public sector, >5 billion to private Coronavirus (has an envelope - 26-32kb)
240
What is an example of a class V virus?
(-) ssRNA - has an envelope and 12 - 15 kb Influenza - killed 20-100 million in 1918 - killed 3-5% of the world 3-5 million cases a year
241
What is an example of a class VI virus?
reverse RNA - has an envelope - 9.7 kb HIV - can cause AIDS (progressive failure of immune system) - body more susceptible to opportunistic infections HIV has a tropism for immune cells: macrophages, dendritic cells, CD4+ T cells 78 million deaths to date In 2016 - 38 million living with it, 1.5 million contaminations and 0.5 million deaths
242
What is an example of a class VII virus?
reverse DNA - has an envelope - 3.1 kb Hepatitis B - cirrhosis and liver cancer
243
What is the genome of each virus class?
I - dsDNA II - ssDNA (+) VII - dsDNA III - dsRNA IV - ssRNA + V - ssRNA - VI - ssRNA +
244
Do bacteriophages, animals and plant viruses have diversity?
YES! There are lots of different forms Bacteriophages - different sizes, dsDNA Animal/plant - nonenveloped, enveloped
245
What are the five major steps in the virus life cycle?
1) Attachment 2) Genome injection (entry) 3) Production of nucleic acid and protein 4) Maturation (assembly of viral particles) 5) Release
246
What is a virulent phages life cycle?
1) Attachment 2) Genome injection 3) Phage DNA circulates - could enter lysogenic 4) Phage DNA and proteins are synthesised and assembled into virions. 5) Cell lyses - releases phage virions Lytic cycle - infectious
247
What is the temperate phages life cycle?
1) Attachment 2) Genome injection (entry) 3) Phage DNA circulates 4) Phage DNA integrates within the bacterial chromosome by recombination = prophage 5) Cell division 6) The prophage may excise from the chromosome initiating a lytic cycle. Lysogenic cycle
248
What is the DNA viruses life cycle in animal/plants?
1) Attachment (virion attaches to host) 2) Entry (virion enters cell, DNA is uncoated) 3) Viral DNA is transferred to nucleus and the DNA is replicated - viral proteins are produced 4) Maturation - Virions assemble 5) Release - virions are released
249
What is the RNA virus life cycle in animals/plants?
1) Attachment 2) Entry 3) Maturation: - group IV - make proteins directly from ssRNA (+), make genome via ssRNA (-), - group V - make ssRNA (+) first and use this to produce proteins and ssRNA (-) genome, - group III - make ssRNA (+) and use it to make proteins and ssRNA (-) genome 4) Maturation - capsid forms 5) Release
250
What is the life cycle of group VI retroviruses viruses?
1) Retrovirus enters by fusion between attachment spikes and host receptors 2) Viral RNA genome and 3 viral enzymes are released: reverse transcriptase, integrase, protease. 3) Reverse transcriptase copies viral RNA to dsDNA in the cytoplasm 4) dsDNA is transported to nucleus and integrated into host genome by integrase - the provirus is replicated when host replicates 5) Transcription of the provirus produces new genomes and RNA encoding capsid, enzymes and envelope proteins 6) Viral proteins are processed by viral proteases - some go to plasma membrane 7) Mature retrovirus buds out of host cell with envelope and attachment spikes
251
How are viral particles packaged in animals/plants?
dsDNA viruses: encapsidation is coupled with maturation of replicative genome ssRNA virus (inc. retroviruses): electrostatic interactions between basic capsid proteins and RNA genome (acidic), promote cooperative assembly of capsomers Fragmented genomes: genetic fragments in complex with proteins are paired before encapsidation
252
What is a pathogen?
Any agent that can cause disease
253
What is pathogenesis?
Survival strategy One organism using resources of another in a way which is not beneficial to the host Tick on a dog = parasite Common cold = virus = pathogen
254
Can archaea be a pathogen?
No - there is none that cause disease to any pathogen
255
What are some examples of pathogens?
Bacterial - yersinia pestis - plague Viral - polio virus - polio Fungal - cryptococcus - meningitis Protozoa ('eukaryotic) - plasmodium falciparum - malaria Prions (infectious proteins) -vCJD - human 'mad cow' disease NO ARCHAEA
256
What makes a successful pathogen?
1) gain access to host 2) locate a nutritionally compatible niche 3) avoid/subvert/circumvent the host innate and adaptive immune responses 4) access host's resources and replicate (THIS IS WHERE INFECTION HAS OCCURED - not all exposure results in disease) 5) exit and spread to new host - transmission - air (droplets), water, food, vector
257
What is the host's innate and adaptive immune responses?
Innate: - non-specific - rapid Adaptive: - highly specific - slower
258
What is virulence?
Measure of disease severity Mortality - number of deaths from a disease Morbidity - number of cases of a disease Ebola (EBOV strain) - ~ 90% mortality Plague (pneumonic), rabies (preventable) and vCJD have mortality rates near 100%
259
How do we measure the successfulness of a pathogen?
See if the pathogen is still there
260
What is the infectious dose?
Number of individual particles/cells required for infection High virulence: e.g. Shingella dysenteriae - diarrhoeal disease - 10 cells Low virulence: e.g. salmonella - food poisoning - 1,000,000 cells
261
What are virulence factors?
These enable a pathogen to colonise the host - DO NOT CAUSE DISEASE Adhesins 'anchors' - find niche and colonise Capsules/s-layers 'armour' - immune evasion and survival in host Digestive enzymes - finding niche, colonising and finding host resources Toxins - reprogram host biology to benefit pathogen Stealth mode - absence of outer surface structures - immune evasion
262
What is the difference between a pathogen and a parasite?
You can see a parasite with the naked eye
263
Where do we find reliable information on pathogens?
World: WHO UK: UK health security agency (humans), DEFRA (animals/plants) USA: CDC (humans), USDA (animals/plants)
264
What percent of world deaths are caused by infectious disease?
26% lower resp - 2.2 million in 2019 diarrhoeal diseases - 1.5 million TB - 1.2 million HIV/AIDS - 700,000 Heart disease, stroke, COPD, resp infections, child birth, lung cancer, dementia, diarrhoeal diseases, diabetes, kidney disease = 55% of all deaths worldwide
265
What is germ theory?
Founding principle States many diseases are caused by microbes
266
What did Robert Koch do?
Established a scientific basis linking microbes and disease. Used pure cultures to understand infectious diseases. Used the bacterium Bacillus anthracis (Anthrax). He studied this disease because it would kill sheep and cattle.
267
What did Koch postulate?
He proved that a specific pathogen causes a specific disease. 1) The microbe is found in all cases of disease and absent from healthy individuals. 2) The microbe is isolated from the diseased host and grown in pure culture. 3) When the microbe is introduced into a healthy susceptible host, the same disease occurs. 4) The same strain of microbe is obtained from the newly diseased host. Host + pathogen = disease
268
When did humans start to try to combat disease?
In the last 200 years - better diet and clean drinking water. Improved sanitation and less overcrowding in urban areas and better living conditions.
269
What are vaccines?
Chemical agents which prime the adaptive immune system to repel a pathogen. After vaccination, if the subject can be exposed to the pathogen and does not develop the disease - they are now IMMUNE
270
What is smallpox?
Very common disease in 1800's with mortality rate of 30%. Between 1900-1979, 300-500 million died of smallpox. Lady Montagu (1717) introduced an early form of vaccination by directly adding pus from smallpox into the open vein of the patient. Edward Jenner (1796) inoculated a person with cowpox virus (less virulent than smallpox) - they were then protected against smallpox - cross protection
271
Is smallpox eradicated?
Yes! As the smallpox vaccine worked - smallpox is extinct from 1979.
272
What do we use for vaccines?
It varies for disease Chickenpox - attenuated strain Hepatitis A - inactivated virus Hepatitis B - viral antigen Influenza - inactivated virus MMR - attenuated viruses Polio - inactivated Rabies - inactivated Yellow fever - attenuated virus
273
Do vaccines work for every pathogen?
NO - but work well with viruses
274
What are antibiotics?
Chemicals produced by bacteria and fungi that inhibit or kill other microbes Last therapeutic options. Compound which controls pathogen infections without harming patient - magic bullet
275
What is the most impactful antibiotic?
Penicillin - the first - Alexander Fleming noticed that the penicillium mold was killing staphylococcus Penicillium mold
276
What were the challenges of making penicillin?
Florey and Chain's challenges: - purification - production - prove penicillin was non-toxic - 'first' human and animal tests Saved millions of lives - 80-200
277
How are antibiotics divided?
Classes or family e.g. penicillins Injection - Penicillin G Orally - Amoxycillin Narrow spectrum - Oxacillin Broad spectrum - Ampicillin Extended spectrum - Carbenicillin Resistant to B-lactamase enzymes - Methicillin
278
What may antibiotics target?
Fundamental processes in a bacterial cell - not human Cell wall synthesis - penicillins Proteinsynthesis Cell membrane integrity Nucleic acid function Intermediary metabolism
279
What is the difference between symptom, sign and syndrome?
Symptom: a change in body function felt by a patient as result of disease e.g. feeling tired Sign: a change in a body which can be measured or observed as a result of disease e.g. high temp Syndrome: a specific group of signs and symptoms that accompany a disease
280
Can one pathogen cause multiple diseases?
Yes! Streptococcus pneumoniae (gram positive bacteria) - pneumonia - sepsis - meningitis
281
How severe is pneumonia?
920,136 children under 5 in 2015 died - accounts for 16% of deaths for under 5 The most common cause of infection-related deaths in UK and USA
282
Can diseases be caused by multiple pathogens?
YES Pneumonia: Bacterial - Streptococcus pneumoniae, Haemophilus influenzae Viral - respiratory syncytial virus Fungi - Cryptococcus neoformans (usually in immunocompromised individuals) The symptoms and signs are identical
283
What is pneumonia?
A reaction of the body to colonisation or damage to the airways
284
What does our microbiota provide?
It benefits our health and wellbeing: - helps with digestion - healthy metabolism - immune function - mood and behaviour - obesity/diabetes/heart disease
285
What is commensalism?
Relationship between bacteria and organisms One organism benefits, the other is unaffected - friendly colonisers e.g. Staphylococcus epidermidis on the skin
286
What is mutualism?
Both organisms benefit e.g. E.coli in large intestine - makes vitamin K12 - blood clotting
287
What is parasitism 'pathogenesis'?
One organism benefits at the expense of the other Human viruses
288
What is C.difficile and what does it cause?
Clostridium difficile - spore-forming gram positive bacteria Causes Clostridium difficile infection: diarrhoea colonisation and inflammation of the colon, abdominal pain, fever
289
How is Clostridium difficile transmitted and what are the virulence factors?
Faecal-oral route Mortality = 9% In USA - 500,000 infections, 29,000 deaths Virulence factors = TcdA and TcdB, S-layer armour The spores allow them to survive penicillin - allows c.diff to colonise (is it a pathogen or a survivor?) Treatment: antibiotics, infection control, faecal transplantation
290
What is an opportunistic pathogen?
Microbes which aren't normally pathogenic but can cause infection/disease in a compromised host Sepsis - treatment requires total annihilation of the organism
291
What is a hospital acquired infection?
Infections acquired as a result of a hospital stay Usually the patient themselves which are the source of their own infections e.g. C.diff and MRSA
292
What is urbanisation?
Urban areas 2007 - >50% of the world's population live in urban areas - Africa and Asia expect to rise Drives potential routes of transmission
293
What is Vibrio cholerae?
Causes Cholera Gram negative bacteria
294
What is Cholera?
By Vibrio cholerae Symptoms: large amounts of watery diarrhoea (rice-water stool), abdominal pain, vomiting Virulence: death through severe dehydration - 1.3/4 million cases and 21,000 - 143,000 deaths - mortality can be 1% if treated rapidly Virulence factors: cholera toxin
295
How is cholera treated and transmitted?
Transmitted: faecal-oral route (water/food) Treatment: oral rehydration and antibiotics
296
Who is John Snow?
Investigated the 1854 cholera outbreak - the broad street pump
297
What is epidemiology?
Study of where and when diseases occur to control spread of disease
298
What are the principles of epidemiology?
- identify first person to have disease 'patient zero' - identify anyone who had contact with that person - identify reservoir for pathogen - blocking or containing it (difficult for new pathogen, disease must first be recognised, many already infected by then)
299
Are disease patterns used for epidemiology?
Yes! 'Tracking and surveillance of disease patterns is an essential tool to control disease' - WHO
300
What is the difference between epidemic, endemic and pandemic?
Epidemic: disease acquired by many hosts in a given area in a short time Endemic: disease constantly present e.g. common cold Pandemic: worldwide epidemic e.g. HIV
301
What is Salmonella enterica serovar Typhi?
Gram negative bacteria Causes Typhoid fever
302
What is typhoid fever?
Caused by Salmonella enterica serovar Typhi Symptoms/signs: rash (rose spots), malaise (feeling ill), delirium, fever, pain , high BP, diarrhoea Symptoms are non-specific and extremely variable e.g. rose spots only in 50% of cases
303
What is the virulence of typhoid fever?
Mortality rate - 30% with no treatment, 1% with treatment Virulence factors: toxins, capsule
304
How is typhoid fever transmitted and treated?
Transmission/reservoir: human carriers (colonised gall bladder), 1-3% of exposed will permanently carry disease, faecal oral route Treatment: antibiotics, vaccine (doesn't offer full protection)
305
What is polio (poliomyelitis) caused by?
Caused by polio virus (+) strand RNA virus picornavirus
306
What are the symptoms and virulence of polio?
95% have mild symptoms 1/200 - irreversible paralysis Muscle weakness Atrophy Deformities Twisted feet/legs Virulence: 15% of symptomatic cases = death as breathing muscles become immobilised
307
How is polio transmitted and treated?
Transmitted: humans (some chimps), faecal oral route usually by contaminated water source Treatment: vaccine - attenuated(weakened) virus, few treatment options
308
Does the polio vaccine work?
Uses attenuated version (weakened) In rare cases the virus can revert to a form which causes disease and infects others In extremely rare cases some immunocompromised individuals become healthy carriers - excrete large amounts of active polio for long amount of time - OPV paradox
309
What is a healthy carrier?
Individuals capable of transmitting disease to others but so not have signs of symptoms of infection e.g. Typhoid mary, Birmingham man (excreted polio for the last 31 years)
310
What is Yersinia pestis?
A gram negative bacteria causing bubonic and pneumonic plague Virulence factors: many toxins. V, W, YopB, YopD - avoid phagocytosis, F1 - makes flea hungry
311
What is the plague?
Symptoms: buboes (swollen lymph glands), flu symptoms (chills, fever), sepsis, pneumonia Virulence: very high mortality if untreated - bubonic = 50%, pneumonic = 90-100%, treatable if <24 hours since first symptoms
312
How is the plague transmitted and treated?
Transmitted: rodents/prairie dogs - rodent fleas, human respiratory aerosol (pneumonic) Treatment: antibiotics and vaccine
313
Is Yersinia pestis a zoonotic pathogen?
Yes! It can be equally as devastating to rodent Epizootics - animal equivalent of epidemic - have animal reservoir
314
What is Phytophthora infestans?
Spore forming oomycete (water mold) which causes potato blight
315
What are the symptoms and virulence of potato blight?
Symptoms/signs: white mycelium growth over surface of leaves and tubers (potatoes) Virulence: approaching 100% of crops - especially if soil is pre-contaminated and the potato variety is susceptible
316
How is potato blight transmitted and treated?
Transmitted: very stable spore structures Treated: fungicides (limited use), copper sulphate (could cause heavy metal toxicity)
317
Do pathogens share same geographical distribution to host?
Yes! They usually co-evolve with them Tight population bottleneck reduced genetic diversity in potatoes - greater disease
318
What is influenza?
Caused by influenza virus - Orthomyxoviridae (- ssRNA) Virulence factors: changed in the H (hemagglutinin) and N (neuraminidse) proteins
319
What are the symptoms and virulence of influenza?
Symptoms: fever, chills, cough, chest pain, sore throat, muscle pain Virulence: 0.01-50% mortality - newer strains have higher fatality
320
How is influenza transmitted and treated?
Transmission: human reservoir, respiratory droplets, can infect other animals Treatment: antiviral drugs, vaccine - varied efficacy due to differing strain types
321
What forms new influenza virus particles?
Re-assortment of genetic material E.g. bird virus + human virus -> swine (pig) cell - makes a new reassortant virus - this can infect humans
322
What have pathogen caused throughout history?
Economic changes Social changes Scientific changes
323
What is Zika virus disease?
Identified in 1947 and named after the Zika forest in Uganda Symptoms: mild fever, rash, muscle and joint pain, headache, symptoms last for 2-7 days Virulence: extremely low, most people won't have symptoms at all - associated with birth defects - microcephaly, Guillain-Barre syndrome Transmitted: Aedes species mosquito, sexually transmitted - men can infect for 6 months, women = 8 weeks Treatment = control of mosquito population, condoms
324
How is a disease identified?
A disease is first identified as a geographically clustered pattern of symptoms and signs
325
How was Zika first noticed?
August 2015 - Dr van der Linden noticed an increase in microcephaly in newborns in Brazil October 2015 - She alerted the state health secretary At the same time and location - outbreak of mosquito associated diseases 2016 - link between Zika and microcephaly was established by 'scientific consensus'
326
What is the Aedes mosquito?
Vector for more than one pathogen Dengue virus - high virulence Zika virus - low virulence
327
What is epidemiological surveillance?
Collection, analysis and dissemination of public health data - takes time - can be inaccurate - correlation does not mean causation
328
What are Zika-associated birth defects?
Brain abnormalities Microcephaly Eye defects Hearing loss Damage to: nerves, muscles and bones
329
What is the global distribution of Zika?
Reported cases in south and north america, south asia, some africa and france Zika is no longer strictly limited to areas where mosquitos are found
330
What is the surveillance of Zika?
In the USA - the Zika Birth Defects Surveillance system monitors birth defects potentially due to Zika virus infection e.g. 2,394 births, 116 with defects, 9 died due to defects The correlation between Zika virus infection during pregnancy and birth defects has a mild effect size - 2 to 2.4 cases per 1000 births
331
What is the risk that Zika has on birth rate?
Zika-associated birth defect rate = 5% Any pregnancy having a birth defect = 3%
332
What is the Ebola virus?
Filovirus - (-) ssRNA Causes Ebola virus disease and Ebola hemorrhagic fever Named after the Ebola river in the democratic republic of congo
333
What are the symptoms and virulence of Ebola?
Symptoms: fever, sore throat, muscle pain, headaches, bleeding (vomiting blood, eyes, nose, mouth, anus) Virulence: highly - 40-90% mortality rate
334
How is Ebola transmitted and treated?
Transmission: by direct contact, sexual transmission (unclear), reservoir not known but believed to be fruit bats - zoonotic Treatment: currently untreatable - isolation, potential antibody therapies, vaccine under development
335
What is the biggest challenge to global public health?
Antimicrobial resistance - superbugs
336
Why is antimicrobial resistance a big challenge?
Modern medicine relies on antibiotics Without them: minor infections become life threatening, surgery, child birth, chemotherapy (immunocompromised), infectious diseases would claim more lives
337
What is an antibiotic and its resistance?
Antibiotics are chemicals produced by bacteria and fungi that inhibit or kill other microbes - but these bacteria can fight back 1943 penicillin: - 1940 Penicillin-R Staphylococcus - 1965 - Penicillin-R pneumococcus 1950 tetracycline - 1959 tetracycline-R Shingella The rate of drug discovery has sharply declined - continued use of antibiotics has driven selection for resistant strains
338
Are there deaths due to antimicrobial resistance?
Yes - we estimate it to kill 10 million in 2050 Now - 700,000
339
What are some antibiotics and their pathogens?
Penicillin - clostridium difficile Tetracycline - staphylococcus aureus Erythromycin - streptococcus pneumoniae Vancomycin - neisseria gonorrhoeae Chloramphenicol - mycobacterium tuberculosis Polymyxin - salmonella serotype typhi Trimethoprim - shingella
340
What are resistance mechanisms?
Chemical modification of antibiotic Biochemical changes in target protein Multidrug resistance pumps
341
What are three relationships between organisms?
Mutualism: both benefit e.g. E.coli in the intestine produce vitamins beneficial for the host, in return they use nutrients to sustain growth Commensalism: microbe benefits from the interaction, no impact to host e.g. bacteria on the skin - can be sometimes beneficial to host Parasitism/predation/competition: microbe benefits at the expense of the host e.g. some intracellular pathogens - chlamydiae, apicomplexans
342
What is the difference between endosymbiosis and ectosymbiosis?
Ecto - lives on the surface of another species Endo - one organism inside another Not mutually exclusive
343
What is Mixotrichia paradoxa?
Found in the gut of specific microbes Lives in symbiosis with other bacteria: - 2 endosymbiotic bacteria which degrade cellulose - replace mitochondria - 3 ectosymbiotic bacteria (outside the mixotrichia) (short spirochetes, long spirochetes and bacteroides) which provide motility - possess 4 non functioning flagella
344
What are the symbiotic associations in Hemipteran insects?
Several bacterial species are found: intracellular (A and B) and extracellular (C and D) These are found in specialised structures called bacteriocytes which can aggregate to form organs called bacteriomes
345
What is Legionella pneumophila?
Parasite of amoebae than can replicate within alveolar macrophages Found in freshwater
346
What is bdellovibrio bacteriovorus?
Parasite which attacks gram-negative bacteria Invades the periplasm and feeds on the host - multiply then release
347
What is nodulation?
Plants can't use atmospheric N2 (can only use ammonium or NO3) Some legumes (Fabaceae) can fix N2 via a symbiotic interaction with soil bacteria - Rhizobia This is nodulation - increases growth rate
348
What is a Rhizobia?
Alphaproteobacteria (gram negative) Soil dwelling bacteria part of rhizosphere Complex genome - 5-10Mbp, several plasmids Help some legumes
349
What is the first step of nodulation?
1) Bacterial attraction: flavonoids (root exsudates) attract bacteria Acidic: Rhizobium has glucomannan which touches Lectin on a root hair, cellulose then traps lots of others Basic: They bind with Rhicadhesin Form biofilm Lectin act as plant surface receptors
350
What is the second step of nodulation?
2) Nodulation genes are activated in the Rhizobium - Nod factors produced Nod factors are short oligosaccharides
351
What is the third step of nodulation?
3) Root curls around rhizobium
352
What is step four of nodulation?
4) Formation of infection threads - like tunnels where rhizobium can enter the root hair Symbiosomes (membrane vesicles) contain rhizobium cells
353
What is step five of nodulation?
5) Bacterial differentiation into bacteroids - NCR transforms them They invade cortical cells in symbiosomes Viable = determinate nodules Dead = indeterminate nodules
354
What is step six of nodulation?
6) N2 fixing
355
What is metagenomics?
Allow us to identify bacterial species which can't be grown in lab conditions Axenic = independently of any other living organism
356
What is unusual about Mycoplasma genitalium?
Its genome is 580 kbp - within the circle there are 5 organisms genetic material The five are likely to be due to endosymbiosis - have at least half the size of genome
357
What is the origin of small genomes in symbionts?
Phylogenetic studies indicate that these organism do not have an independent origin Genome size is a result of gene loss
358
What are Mealybugs?
Can grow them on potatoes In symbiosis with Moranella and Trembaya organisms - nested symbiosis This is where an organism has an endosymbiosis relationship with a bacteria (Trembaya) which itself has an endosymbiosis relationship with another bacteria (Moranella)
359
What are Mosaic pathways?
Underpins amino acid biosynthesis The mealybug combines genes from all three organisms - this symbiotic relationship has been selected as all three organisms contribute to the biosynthetic pathway of some amino acids The mealybug also contributes specific components and genes for peptidoglycan synthesis in Moranella
360
Can protists eat protists?
Yes some do Some can also eat bacteria
363
What is a disease state?
Product of a relationship change/conflict between host and pathogen
364
What is immunology?
Study of the immune system - integrated system of cells and molecules that defend against disease Reacts against infectious diseases
365
What are some malfunctions of the immune system?
Immunodeficiency Allergy Autoimmune disease Graft rejection
366
What do we use some immunological techniques?
Research Diagnostics and therapeutics
367
What are the features of the innate immune system?
- Broad specificity - Not improved by repeat infections - no memory - Rapid (hours) - First form of immunity to evolve - Helps initiate and mediate adaptive immune responses
368
What are the leucocytes and soluble factors in the innate immune system?
Leucocytes: phagocytes, natural killer cells (NK) Soluble factors: lysozymes, complement, interferons
369
What are features of the adaptive immune systems?
- Highly specific - Improved by repeat infection (memory) - Slower response (days) - Foreign material (antigen) is recognised by specific receptors on T and B lymphocytes - Immune system is tolerant of own body's cells and molecules
370
What are the leucocytes and soluble factors in the adaptive immune system?
Leucocytes: B and T lymphocytes Soluble factors: antibodies
371
Where are leukocytes (WBC) derived and what do they give rise to?
Derived from pluripotent stem cells in bone marrow. Gives rise to two main lineages: one for myeloid (polymorphonuclear leukocytes from myeloblast - neutro/eosino/basophils, monoblasts - macrophages, monocytes dendritic) cells, one for lymphoid (lymphocytes - natural killer cells, B/T) cells
372
What are some external barriers to infection?
Keratinised skin - keratin Secretions: sebum, FAs, lactic acid, lysozyme Mucous - cilia in resp tract Low pH - stomach pH is 2.5 Commensals
373
What are the types of phagocytes?
Elie Metchnikoff 1883 saw them firstly Neutrophils: main phagocyte, short-lived, fast-moving, lysosomes release enzymes, H2O2 ect. Mononuclear phagocytes: long lived (months), help initiate adaptive responses Monocyte (blood) --> macrophage (tissues)
374
What are natural killer cells?
Type of lymphocyte - kill virally infected cells non-specifically - important in self/non-self recognition - may kill cancer cells
375
How do phagocytes detect pathogens?
Have general pathogen-recognition receptors (PRRs) that recognise microbe-associated molecular patterns (MAMPs) which are shared by many microbes and essential e.g. Toll-like receptor 4 (TLR4) recognises lipopolysaccharide
376
How do natural killer cells detect pathogens?
Kill targets unless they recognise self-proteins (MHC) which are present on all nucleated cells
377
What do the soluble factors do in the innate immune system?
Complement system: ~ 20 in blood which are activated during infection - bacterial cell lysis Defensins: positively charged peptides made by neutrophils, disrupt bacterial membranes Interferons: produced by virally infected cells - protect uninfected cells and activate macrophages and NK cells
378
What are some of the soluble factors in the innate immune system which are involved in cell:cell communication?
Cytokines: small, secreted proteins that bind to cells and regulate immune response e.g. interleukins - produced by cells of innate and adaptive IS Inflammatory mediators: histamine, prostaglandins
379
What is inflammation?
Heat, redness, swelling, pain Localised response to infection/damage Dilation of BV, increased capillary permeability, phagocytes migrate into tissues Infection/damage induces release of inflammatory mediators (PG, histamine) and production of cytokines
380
What is the temperature response (fever)?
Macrophages may release cytokines when there's infection e.g. interleukin 1 This acts on hypothalamus --> raise temp --> stims phagocytosis --> reduces level of iron in blood (as bacteria use the iron to grow)
381
What manipulates the immune system to provide protection?
Vaccination
382
What are some examples of pathogens?
Bacteria (1-5 micrometres) - Staphylococcus aureus (MRSA) = Boils, septicaemia Viruses (20-400 nm) - HIV = AIDS, influenza = flu Fungi (2-20 micrometres) - Candida albicans = thrush, Epidermophyton flocosum = ringworm Parasites (1 micrometres - 10m) - Schistosoma mansoni = schistosomiasis, Typanasome brucei = sleeping sickness
383
What are the differences between T and B lymphocytes?
T - mature in thymus, has a T cell receptor B - mature in bone marrow , has an antibody Primary lymphoid tissue - antigen independent differentiation
384
What is the peripheral lymphoid tissue?
Antigen dependent differentiation B and T lymphocytes go to lymph nodes and spleen Causes humoral and cell mediated immunity Humoral - B cells - secrete antibody, for extracellular bacteria and secondary viruses Cell mediated - T cells - kill infected host cells and make cytokines, for viral, intracellular bacteria and intracellular parasite
385
What are antibodies?
Produces in response to antigen (antibody generating material)
386
What is the clonal selection hypothesis?
Macfarlane Burnett Those B cells with the antigen are chosen for division- form plasma cells to produce antibodies and form memory cells Lymphocytes that recognise 'self' are deleted early in development
387
What cells can T cells recognise?
Only antigen bound to host cells
388
How does the cell mediated immune response work?
T cells Clonal expansion --> differentiation --> memory
389
What are the differences between primary and secondary lymphoid tissue?
Primary: lymphocytes reach maturity and acquire their specific receptors Secondary: mature lymphocytes are stimulated by antigen
390
What are the primary and secondary lymphoid tissues?
Primary: thymus and bone marrow Secondary: Tonsils, adenoids, lymph nodes, spleen, peyer's patches, appendix, lymphatic vessels
391
What is the basic antibody structure?
Belong to class of soluble glycoproteins known as immunoglobulins - act as labels for infectious material Dual role Fab regions - variable and binds different antigens specifically - antigen recognition FC region: constant in sequence, binds to complement, Fc receptors on phagocytes, NK cells ect. - antigen elimination
392
What are the chains in antibodies?
Light chain: 25kD Heavy chain: 50kD L2H2 = 150kD Bound together by disulphide bridges
393
What keeps the Fab and Fc regions together?
Fab = fragment antigen binding Fc = fragment crystallizable Fab are two arms, Fc is bottom Papain cleavage cuts the hinge region - 2 Fab, 1 Fc
394
What are the classes of immunoglobulins?
IgG = γ - main class in serum (blood without cells and clotting factors) and tissues important in secondary responses - can cross placenta IgM - µ - important in primary responses IgA - α - in serum and secretions, protects mucosal surfaces IgD - δ - ? IgE - ε - low levels in serum, involved in allergy, protective against extracellular parasites Also two light chain types - kappa (κ) and lambda (λ) - not class restricted - e.g. can have IgGλ antibodies
395
What do primary and secondary responses involve?
IgG and IgM in primary (IgM first) and secondary - IgG higher in secondary IgA and IgE in secondary depending on type of pathogen and site of infection
396
What are the two regions in an antibody?
Variable - bind to antigen - differ between antibodies Constant - same for antibodies of a given H or L chain type Encoded by different exons Multiple V region exons in genome can recombine and mutate giving B cell differentiation to give different specificities
397
What do the five classes of immunoglobulins differ on?
Amino acid sequence of heavy chain
398
Which immunoglobulins look different?
IgA - can be monomer (serum) or dimer (secretions) IgM - pentamer
399
What are the two functions of antibodies?
Specific binding/multivalency (FAB): - Neutralise - IgG, IgA - Immobilise motile microbes - IgM - Prevent binding to, and infection of, host cells - Form complexes Effector functions (Fc): - Activate complement - IgG, IgM - Bind Fc receptors - phagocytes (IgG, IgA), NK cells (IgG), mast cells (IgE)
400
What is complement?
Immune defence against bacteria (viruses) and inducer of inflammation - can cause pathology ~ 20 serum proteins activated via enzyme cascade e.g. proenzyme 1 makes enzyme 1 which makes proenzymes 2 ect. Activated specifically by antigen/antibody complexes (CLASSICAL pathway) or non-specifically e.g. by certain bacteria (MB-LECTIN or ALTERNATIVE pathway)
401
What are the main proteins of the classical complement pathway?
C1, 4, 2, 3, 5, 6, 7, 8, 9 Many components have enzymatic (protease) activity - generate fragments with biological activity e.g. C3 --> C3a + C3b
402
What does the classical pathway require?
C1, C2, C4 Require 1 antigen and 2 antibodies C1 must interact with 2 Fc regions to be activated IgM is more potent activator of complement as it's a pentamer
403
What does the activation of C1, C4 and C2 lead to in the classical pathway?
Leads to generation of C3 convertase C3b joins C3 convertase to make C5 convertase
404
What are the three major biological activities of complement?
1) Activation (phagocyte recruitment and induce inflammation) - C5a (C3a), chemoattractants and anaphylatoxins 2) Opsonification - C3b, increased binding and phagocytosis 3) Cell lysis - Membrane Attack Complex: C5-9 - hollow cylinders which form 'pores' in bacterial membranes
405
Which bacteria are not susceptible to membrane attack complex?
Gram positive
406
Which antibodies can act as opsonins?
IgG and IgA Fc receptors bind to Fc region - help phagocytes engulf using pseudopods Pseudopods then fuse - phagosome - lysosomes fuse with phagosome - phagolysosome - enzymes (lysozymes), competitors (lactoferrin), reactive oxygen species (H2O2) - bacteria killed
407
Are there Fc receptors on Natural Killer cells?
Yes! Mediate antibody dependent cell-mediated cytotoxicity (ADCC) IgG Secretion (perforin) - Target then undergoes apoptosis
408
Are there Fc receptors on mast cells?
Yes! Mediate allergy/defence against large parasites IgE Degranulation releases inflammatory mediators e.g. histamine - local inflammation - beneficial in response to parasites
409
How do we use antibodies in research?
Producing antisera (ready made immunity from animals/humans already exposed) - gives antibodies directly Conventional antisera = polyclonal antisera (mix of antibodies targeting same antigen at different sites (epitome))
410
How does a polyclonal B cell respond to antigen?
Mixture of antibodies to different epitome (shape antibody binds to) May lack fine specificity and difficult to standardise
411
What are monoclonal antibodies?
Single specificity derived from single B lymphocytes B cell from animal immunized with antigen A + Tumour cell line (divides indefinitely) = hybrid cells making anti-A antibody and divide indefinitely
412
How are antibodies used in research, diagnostics and therapy?
1) identify and label molecules in complex mixtures 2) identify pathogens 3) characterise cell surface proteins, identify cell types 4) humanised antibodies are used in therapy
413
What are the two major subpopulations of T lymphocytes?
T helper cells (CD4 +ve): - help B cells make antibodies - activate macrophages and NK cells - help development of cytotoxic T cells - do all this using cytokines T cytotoxic cells (CD8 +ve) - recognises and kills infected host cells
414
What is the structure of the T lymphocyte receptor?
Has an alpha and beta chain which both have control and variable regions Chains help by disulphide bridges TCR structure is similar to Fab arm of antibody
415
What do the B and T cells recognise?
B cells - 'soluble', free, native antigens T cells - 'cell-associated' processed antigens on MHC molecules
416
What are MHC proteins?
Major histocompatibility proteins On body cells - can tell immune cells that the cell is infected
417
What are properties of MHC proteins?
Encoded by Major Histocompatibility gene Complex on chromosome 6 Important in graft rejection Very polymorphic - lots of different alleles Major role in initiating T cell responses
418
What are the two MHC proteins?
MHC I - expressed by all nucleated cells - display antigen to CD8+ve T cells (cytotoxic) MHC II - expressed by macrophages, dendritic cells and B cells - display antigen to CD4+ve T cells (helper)
419
How does the cytotoxic T cell recognise infected cells?
Recognises peptide bound to MHCI Virus infected cell --> viral proteins broken down in cytosol (proteosomes) --> peptides transported to ER and bind MHCI --> cell surface Activated cytotoxic T cells kill infected cell (perforins) by inducing apoptosis
420
How do T cells recognise MHCII?
Macrophage/dendritic/B cell internalises and breaks down foreign material ---> peptides bind to MHCII in endosomes --> cell surface T helper cells bind and help BB cells make antibodies and produce cytokines that activate/regulate other leucocytes
421
What is thymic selection?
T cells acquire their receptors and are 'educated' in thymus Only T cells that recognise self-MHC and not self-peptides survive 5% go to peripheral lymphoid tissue
422
What are cytokines?
'Hormones' of immune response Small (5-20 kD) secreted proteins involved in the communication between cells of immune response Usually produced and act locally Act by binding to specific receptors on the surface of target cells (cytokine receptors) Biological effects: changes in cell behaviour e.g. movement, secretion, changes in gene expression
423
What are some main cytokines?
Interleukins (IL-1, IL38?) -often made by T cells Interferons - viral infections e.g. IFNalpha, IFNbeta; cell activation (IFNgamma) Chemokines - cell movement or chemotaxis e.g. IL-8 (CXCL8) Tumour necrosis factor - proinflammatory, can kill some cells Colony stimulating factors - leukocyte production e.g. M-CSF
424
How is the adaptive immune system activated by draining of lymph node?
In lymphatic vessel: Lymph draining from infected tissue --> dendritic cell with bacterial antigen --> T cells activated --> plasma cells and antibodies made --> effector T cells Naive T cells in blood
425
How do innate and adaptive immune responses interact?
Co-evolved Innate initiate adaptive Adaptive augment innate
426
What did Edward Jenner do?
1796 Showed that infection with cowpox was protective against smallpox This is because they share antigens Smallpox is eradicated (1979)
427
What are the types of vaccine?
Attenuated strains e.g. polio Killed pathogen e.g. polio Subunit e.g. toxoid (from toxin), virus spike protein Engineered virus e.g. Astrazeneca SARS CoV-2 RNA e.g. Moderna CoV-2; melanoma vaccine mRNA-4157
428
Which diseases have no effective vaccines?
Malaria Schistosomiasis TB HIV/AIDS
429
When was AIDS first recognised?
1981 The HIV virus was identified in 1983
430
What type of virus is HIV?
Retrovirus - tumour virus (association with cancer shown in 1911 by Peyton Rous) Lentivirus - "slow virus"
431
What is the genome of HIV?
2 copies of + ssRNA
432
How is HIV replicated and transcribed in a cell?
It's RNA (can act as mRNA) is converted to DNA by reverse transcriptase The DNA then can be transcribed to RNA then protein Life cycle of retroviruses determined in 1973
433
What is the HIV virion?
Has viral receptors - gp120 which is bound to the gp41 in the membrane Nucleocapsid - has reverse transcriptase, protease, integrase, RNA
434
How does HIV enter a cell?
1) virus binds to cell receptor 2) envelope fuses with plasma membrane 3) nucleocapsid enters cytoplasm 4) viral RNA reverse transcribed into dsDNA 5) viral DNA is transported to nucleus and integrates into host genome = provirus 6) new viral genome RNA and mRNA --> cytoplasm 7) viral mRNA --> viral protein 8) new nucleocapsids form and virus buds from cell - acquires lipid envelope
435
What are the three parts of retroviral infection?
Latent Permissive Lytic
436
What does HIV infect?
CD4 +ve cells (T helper) susceptible to infection by Gp120 T helper cells - virus initially cleared but pool of infected cells gradually increase Latent ----> activated - caused by T cell stimulation activating HIV provirus transcription Number of T cells infected increases with each round of viral replication
437
How are monocytes/macrophages/dendritic cells involved with HIV?
Dendritic cells present antigen to T cells in lymphoid tissue Infection is 'permissive' - acts as reservoir of virus Monocytes may cross the blood brain barrier - CNS involvement
438
Is CD4 expression enough for HIV infection?
Co-receptors required - identified in 1996 Co-receptors are chemokine receptors e.g. CCR5 - required for virus fusion with host cell May influence susceptibility to infection/disease progression
439
What is the immune response to HIV?
Initially high levels of virus in the blood are cleared Antibodies to HIV may not be detected for 3 months (seroconversion) Immune system mounts a strong response - CD8 +ve cytotoxic cells are important BUT virus mutates to avoid immune detection
440
What does T helper cell depletion lead to?
AIDS - direct lysis by virus - syncytium formation - killed by cytotoxic T cells or other immune mechanism - apoptosis Infected cells are in lymphoid tissue
441
What level of T helper cells is AIDS?
Blood count CD4+ve T cells < 200/mm3 = AIDS
442
What are the symptoms of AIDS?
- memory T cells lost - all adaptive immune responses are compromised - opportunistic - reactivation of latent viruses - rare cancers - CNS involvement - Dementia
443
What is the epidemiology of HIV?
HIV-1 - central Africa Likely source - related virus infecting chimpanzees Phylogenetic studies suggest cross-species transmission 1910-1930 HIV-2 - west Africa - pre-dates HIV-1 Likely source - related virus affecting sooty mangabees Less virulent and less easily transmitted
444
What is HIV's prevalence?
Claimed over 32.7 million lives so far ~ 38 million people currently living with it New infections decreasing by 1 million over last 10 years
445
How is HIV transmitted?
Unprotected Sexual intercourse ~ 70% Blood/blood products ~ 28% Breast feeding Mother to foetus
446
How do we prevent HIV transmission with behaviour?
Change behaviour: blood testing, safe sex, less needle sharing, treat HIV+ve pregnant women
447
Is there a HIV vaccine?
Currently focus on: vaccines inducing 'broadly neutralising antibodies' - self-assembling nanoparticles and mRNA. Also engineered virus vaccines which induce cytotoxic T cells Problems: high mutation rate (60x higher than flu), 'humoral' immunity may not be protective, need to induce cytotoxic T cells 9 vaccine trials to date - only one (RV144) showed protection (31%) - a combination of two vaccines that didn't work
448
Can we use drug therapy to treat HIV?
Combination therapy: - cocktail of drugs directed at different viral targets e.g. AZT (nucleoside analogue), inhibitors of reverse transcriptase, HIV protease inhibitors, fusion inhibitors, capsid inhibitors. > 25 licenced drugs which block HIV replication Preventative drugs: twice yearly injections of Lenecapavir (multistage HIV-1 capsid inhibitor with long half-life)
449
What are problems of drug therapy on HIV?
- high mutation rate - toxicity - viral latency - cost
450
How many people are on HIV treatment?
At end of 2019: 67% of HIV +ve people had antiretroviral therapy and 59% had suppression of virus 2020 - drop in new people starting treatment e.g. due to COVID 2025 - US cuts funding to science and aid
451
What are some future treatments of HIV?
Stem cell therapy (bone marrow cells lacking CCR5) "Kick and kill - reactivate latent virus and immunotherapy Passive immunisation - using monoclonal antibodies or engineered T cells Gene editing with CRISPR/Cas9 Engineered T cell receptors recognising reservoirs of infected CD4+ve T cells