• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/224

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

224 Cards in this Set

  • Front
  • Back

What's the difference between an exon and an intron?

Exon = protein coding sequence
Intron = non-coding sequence

What is an allele?

Different versions of the same gene

Explain the cell biology approach for studying cells

Direct observation (microscopy), label and observe cell structures

Explain the biochemical approach to studying cells

Isolating and describing proteins

Explain the genetics approach to studying cells

Looking at mutant genes and proteins and studying their effects

Explain the genomics approach to studying cells

Looking at all the genes at the same time

Explain the developmental biology approach to studying cells

Looking at the differential gene expression and signals that lead to the mature organism

What happens in G1?

Recovery from mitosis, growth

What happens in S phase?

DNA is replicated

What happens in G2

Pre-mitosis checkpoints

What happens in M phase?

Mitosis - chromosome segregation and division

What happens in G0 phase?

Permanent or temporary exit from the cell cycle (most cells are in this phase, fully differentiated, quiescent)

What is a chromatid?

One copy of a duplicated chromosome

What are sister chromatids?

identical copies of a chromosome joined by a centromere

What are homologous chromosomes?

Chromosome pairs, distinct, one from each parent

What is the product of mitotic division?

Two genetically identical daughter cells

What type of chromosome alignment happens in mitosis?

Independent alignment (not paired with its homologue)

Each sister chromatid separates and goes to a different cell making both of the daughters genetically identical

What is chromatin?

Proteins that bind DNA to help it condense

What are cohesins?

Proteins (like little rubber bands) that hold the chromosome together

What is the centromere?

repetitive DNA sequence that serves as a target for mitotic machinery (the bit in the middle of the chromosome 'X' shape)

What is a kinetochore?

Protein complex (one per chromatid) that links the centromere to the microtubules. Kinetochore proteins target the centromere to place itself

What are the stages of mitosis?

Interphase
Prophase
Prometaphase
Metaphase
Anaphase
Telophase
Cytokinesis

What happens during interphase?

chromosome duplication and cohesion
centrosome duplication

What happens during prophase?

interphase microtubule breakdown replaced by mitotic asters
Mitotic aster separation




Chromosome condensation

What happens during prometaphase?

Nuclear envelope breakdown
Chromosomes captured, orientated and brought to the equator


Microtubules contact the kinetochore

What happens during metaphase?

Chromosomes align at metaphase plate

What happens during anaphase?

Cohesins degrade
Chromosomes move to poles
Centrosome separation

What happens during telophase?

Assembly of contractile ring
Nuclear envelope assembly

What happens during cytokinesis?

Reformation of interphase microtubule array
Contractile ring forms cleavage burrow

What is the goal of meiosis?

To produce four genetically different daughter cells with half the number of chromosomes as the cell that undergoes the division (e.g. 2n cell --> 4 x 1n cells)

What is the form of chromosome alignment in meiosis?

Homologous chromosomes pair, recombination occurs and each duplicated chromosome separates.

What is the basic process of meiosis

duplication of chromosomes, pairing of homologous chromosomes, recombination, separation of homologous pairs (meiosis 1) (each daughter cell contains recombined version of either maternal or paternal chromosome), sister chromatids separate (meiosis 2), four haploid gametes form

What is a kinase?

An enzyme that adds a phosphate (activating or inhibiting) to a target

- serine, threonine and tyrosine are common targets

What is a phosphatase?

An enzyme that removes a phosphate group

What is a cyclin?

A family of proteins that control a cell's progression through the cell cycle by binding and activating cyclin dependent kinases

At what stages of the cell cycle are cyclin and CDKs expressed?

CDKs- present throughout cell cycle

Cyclin - only present in specific cell cycle stages

Does the cyclin or the kinase determine the specificity of the cyclin heterodimer?

The cyclin

What is ubiquitination?

Ubiquitin protein ligases attach Ub to target proteins. Repeating this process multiple times (polyubiquitination) marks a protein for degradation by the proteasome (only K63 ubiquitination)

What are the key complexes involved in the G1/S and the G2/M transition?

G1/S = SCF (removes skp1 inhibitor from S cyclin complex)

G2/M = APC/C (securin degradation to activate separase and remove cohesins so that chromatids can separate)

What is the difference between phosphorylation and ubiquitination in terms of permanence of effect?

Phosphorylation is temporary and reversible


Ubiquitination is permanent and irreversible

What are some functions of G1 CDKs?

Phosphorylate transcription factors


help prepare G1/S CDKs

Explain the basic transition at the G1/S boundary (in terms of the CDKs)

An inhibitor of S CDKs determines the G1/S boundary.
G1/S CDKs are involved in synthesising S CDKs, however they have an inhibitor (sic1) attached. Once the concentration of G1/S CDKs gets high enough, all of the inhibitors become phosphorylated at multiple sites, attracting a SCF ubiquitin ligase which degrades the inhibitor. This produces a sharp and defined boundary between G1 and S

Graph the level of protein activity of sic1, S CDKs and G1/S CDKs

What are some of the functions of S CDKs?

Phosphorylation of numerous proteins that go on to replicate DNA


Also help prepare cell for mitosis

What are some ways to differentiate cells based on their positions in the cell cycle?

- size (later in cycle = larger)


- growth factors


- drugs (can arrest cells at particular stages)

What is cdc2?

A cyclin dependent kinase that is key in the mitosis pathway

What is the phenotype of Cdc2+, Cdc2- and Cdc2 dominant cells?

Cdc2+ = wild type (normal cells)


Cdc2- = long cells (no mitosis)


Cdc2 dominant = wee cells (premature mitosis)

What is cdc13

Mitotic cyclin (binds to cdc2)

What is cdc25? Does it drive or inhibit mitosis?

CDC25 is a phosphatase --> DRIVES MITOSIS

What is wee1? Does it drive or inhibit mitosis?

WEE1 is a kinase --> INHIBITS MITOSIS

What mutations could cause a long phenotype in S. pombe?

Defecit of CDC25 (drives mitosis)
Overexpression of WEE1 (inhibits mitosis)

What mutations could cause a wee phenotype in S.pombe?

Overexpression of CDC25 (drives mitosis)
Defecit of WEE1 (inhibits mitosis)

Explain the basic process of wee1/cdc25 activation of the maturation promoting factor (MPF) (MPF = mitotic CDK complex)

Mitotic cyclin and CDK come together to form an inactive MPF. WEE1 phosphorylates Y15 (tyrosine) (inhibitory phosphorylation). CAK then phosphorylates T161 (threonine) with an activating phosphorylation. CDC25 then removes the inhibitory phosphorylation and the MPF becomes active

What are some of the functions of mitotic CDK complexes?

Activaiton of


- chromatin associated proteins
- nuclear envelope proteins


- microtubule associated proteins


- kinetochore proteins

What is a cell cycle checkpoint?

Cellular mechanisms that make sure a task has been completed before cell cycle progression

Give an example mechanism of a cell cycle checkpoint

ATR1
This proteins sits at the replication fork. It can bind and activate CHK1 which inhibits CDC25 by phosphorylation. Thus, if DNA replication hasn't finished, CDC25 is inhibited and the mitotic CDKs cannot be activated so mitosis won't progress with only partially replicated DNA.

How do mitotic CDKs breakdown the nuclear envelope?

The nuclear lamina supports the nuclear envelope. The lamina is made of lamins. Normally, lamins form a tetramer. Mitotic CDKs phosphorylate lamins, making them only able to form dimers. This results in the breakdown of the nuclear envelope.

What are two major roles of APC/C?

1. Metaphase-anaphase transition (securin-separase to remove cohesins)


2. Degrade mitotic cyclins in late anaphase (after chromatids have separated)

What is PP2A?

A phosphatase associated with the centromere that stops the cohesins close to it from being removed by phosphorylation --> meaning they have to be removed via the APC/C pathway

Explain the basic process of the removal of cohesins near the centromere by APC/C

Cohesins are composed of smc proteins and scc1. Separase is a protease that cleaves scc1. Securin binds to and inhibits separase. When all kinetochores bind to microtubules, CDC20 binds APC/C and polyubiquitinates securin leaving separase to be free to cleave the scc1 of the cohesin.

Explain how APC/C degrades mitotic CDKs

APC/C is an E3 ubiquitin ligase. It can either be bound to cdc20 or cdh1 to activate it. When bound to cdc20 it degrades securin.


APC/C binds to cdh1 (changes its specificity) in late anaphase. This E3 complex then polyubiquitinates mitotic cyclins (as well as remaining cdc20 bound APC/C) to ensure that the cell doesn't undergo mitosis again and can have a period of growth

What is necrosis?

Unplanned cell death involving membrane rupture and the spilling of the cellular contents into the surrounding tissue

What are some causes of necrosis?

Toxins


Burns


Lack of circulation
Infection


Inflammation

What is apoptosis?

Programmed cell death involving the cell splitting up into 'apoptotic blebs' to be consumed by macrophages

What are some causes of apoptosis?

DNA damage (e.g. UV exposure)
Structural removal of cells from tissue (eg. removing tadpole tail)
Lack of growth factor (e.g. keeping useful, well connected neural junctions because they secrete growth factor)


explain the basic apoptosis signalling pathway (C. elegans)

Apoptosis signal --> EGL 1 binds to and inhibits CED-9 (which normally represses apoptosis) --> stimulates CED4 dimer release --> CED4 becomes octamer and joins with CED 3 to form the CED4/CED3 caspase holoenzyme which then leads to cell death

What are the results of mutations in the apoptotic pathway?

EGL 1 mutant: no apoptosis
CED 9 mutant: all cells die
CED 4 mutant: no apoptosis


CED 3 mutant: no apoptosis



What is the result of a CED9/CED3 double mutant?

No apoptosis - becuase CED 3 is downstream of CED 9 (epistatic to it) and whatever is downstream is more important

How do tumor cells develop sustained proliferative signalling?

Mutations allowing growth factor receptors to fire independently of the GF ligand

How do tumor cells evade growth supressors?

Mutations e.g. the removal of the sic1 inhibitor on S CDKs which allow them to progress into S phase more quickly

How can cancer cells enable replicative immortality?

Telomeres have repeating terminal sequences. Replication of chromosomes shortens these sequences each time, limiting the amount of time a cell can divide. Telomerase extends the end of telomeres after replication. Telomerase is turned off in adult cells and cancer can turn it back on.

What is angiogenesis?

The stimulation of blood vessel formation within a tumor to avoid necrosis

What are the three different types of mutation in cancer cells?

Gain of function: e.g. in pathways that promote growth


Loss of function: e.g. in pathways that inhibit growth


Dominant negative: e.g in proteins that form complexes (p53), one mutant part of the complex can turn off the whole thing

What are proto-oncogenes?

Genes that normally promote cell growth. Once mutated (generally gain of function), they become oncogenes.


e.g. ras, myc, src

What are tumor supressor genes?

Genes that normally inhibit cell cycle progression. Generally loss of function mutations allow the cell to grow out of control e.g. p53, Rb, ATR

How can cancer mutations occur?

They can be hereditary (e.g. retinoblastoma), spontaneous or evironmental (e.g. carcinogens, UV)

What is the biggest cancer risk factor?

AGE


- multiple mutations are required for cancers to form


- DNA repair is less efficient with age

What is metastasis?

The spread of cancer cells from their site of origin and the start of secondary growth

(cancer cells use invadopodia to degrade the basement membrane, detect epidermal growth factor from blood vessels etc and then degrade and enter the blood vessels to travel)

What is retinoblastoma?

A heritable or spontaneous cancer in young children caused by a defect in the Rb gene

Children can inherit a defective Rb allele from one parent, and then a somatic mutation causes cancer or they can inherit two normal copies and have two mutations resulting in cancer.

What is the role of Rb in the cell cycle?

Rb fits in the G1 phase of the cell cycle and is a key restriction point - i.e. if it is passed then the cell is committed to another phase of replication.

Rb is a transcriptional repressor of E2F. While it is in place, G1 CDKs cannot phosphorylate and activate the E2F transcription factor (which drives the expression of S phase genes). Thus, removal of Rb commits the cell to another phase of replication.

What is hyperphosphorylation?

Some proteins (sic1, Rb) must be phosphorylated multiple times for an action to occur. This means that the action can only happen when the CDK concentration is very high

What is P16 and how does it relate to Rb?

P16 is a CDK inhibitor which prevents phosphorylation and removal of Rb. This inhibitor thus inhibits activation of the E2F transcription factor and cell cycle progression.

What is the expression of P16 driven by?

Cell stress




More stress = more P16 = no progression of cell cycle whilst conditions are bad

How do cancer cells affect P16?

Turn it off

How do cancer cells affect the Rb pathway?

Overexpress/amplify G1 CDKs


Delete or inactivate Rb (retinoblastoma)

What is p53?

p53 is a transcription factor that can cause cell cycle arrest, senescence (permanent arrest) and apoptosis

Explain the basic process of p53 activation in cells

ATR can inhibit Mdm2 (which normally ubiquitinates p53 to keep it unstable). This makes p53 stable allowing it to detect conflict within the cell and activate cell cycle arrest/senescence/apoptosis

(ATR also acts to inhibit CDC25 and hence MPF formation to further stop premature cell cycle progression)

Do p53 mutations cause cancer?

No, but they do predispose a cell to cancer through a loss of tumor suppressor function

What type of mutation is a p53 mutation?

A dominant negative - p53 forms a tetramer so if the mutation produces just one defective protein then the entire complex is defecctive

How do cancer cells target the p53 pathway?

Deletion or inactivation of ATR


Upregulation of Mdm2


Deletion or inactivation of p53

How does p53 inhibit cell growth?

Upregulates CKI --> inhibiton of G1/S and G2/M progression via CDKs

Acts via Bax and PUMA to stimulate apoptosis

Acts via CKI and PAI1 to stimulate senescence

What is meant by the term amphipathic?

Hydrophilic and hydrophobic elements to the same molecule

What are some of the functions of cell membranes?

Structural
- membrane components


- protein modification

Metabolic
- energy storage




Other


- cell signalling


- enzyme cofactors


- electron carriers


- pigments

How does hydrocarbon chain length affect melting point and solubility in water?

Increased chain length
- increases melting point


- decreases solubility in water

How does the number of double bonds affect the melting point?

Increased double bonds decreases the melting point

What is a sphingolipid?

A major membrane component, derivative of amino alcohol spingosine




example = ceramides

What are the different kinds of lipid aggregates?

Micelle - single phospholipid layer sphere

Bilayer - double layer of phospholipid

Vesicle - bilayer sphere

What are the different kinds of bonds that occur in a plasma membrane?

Ionic bonds between the head groups
Hydrogen bonds with water
Van der Waals bonds between the fatty acid tails

What are the two faces of a bilayer?

The cytosolic and the exoplasmic

What are the types of motion in lipids?

Spinning (without changing location)
Lateral diffusion (movement within same face) (10^7 times/sec)
Flip flop (very rare)

What is FRAP and how does it relate to lipids?

Fluorescence recovery after photobleaching

Shows that lipids can easily move laterally in a membrane

What are some factors that affect the fluidity of a membrane?

- temperature


- lipid composition


- chain length


- level of saturation
- cholesterol

How is the curvature of the membrane determined? Give two examples of phospholipids that would contribute

PC (phosphatidylcholine) - flat membrane

PE (phosphatidylethanolamine) - curved membrane

Do membranes differ in their lipid distribution?

Yes - most have asymmetric distribution

What lipids would you expect to see in a cytosolic leaflet vs the exoplasmic?

Cytosolic: rich in spingolipids/PC (less fluid)

Exoplasmic: rich in PE/PS/PI (more fluid)

How does asymmetry in the lipid composition of leaflets arise?

- leaflets do not spontaneously flip
- specific enzymes can catalyse translocations


- synthesis of lipids can promote asymmetry

What are the enzymes that catalyse transbilayer translocations?

Flippase (P-type ATPase)
Floppase (ABC transporter)
Scramblase

What are membrane microdomains and what do they do?

Stable associations of lipids within the membrane - lipid rafts

microdomains control lateral diffusion


What are some functions of membrane proteins?

Transporters
Receptors
Adhesion molecules
Lipid synthesis


Energy transduction

What are the three main types of membrane proteins?

Integral

Lipid anchored

Peripheral

What treatments releases each type of membrane protein?

Integral: harsh treatment e.g. detergents, organic solvents, denaturants

Lipid anchored: do not enter membrane bilayer- attached to lipids - can be released using phospholipase C

Peripheral: mild treatment: attached weakly and can be released with mild treatment e.g. carbonate at high pH, high salt)



What are some features of integral membrane proteins?

Goes fully across the membrane
Has both hydrophobic and hydrophilic interactions
Firmly associated to membrane- need harsh treatment e.g. detergents, organic solvents to remove

Give some features of lipid anchored membrane proteins

Protein covalently linked to one or more lipid molecules
Lipid is embedded in the leaflet but does not cross the whole bilayer
Can be released using phospholipase C

What are some features of peripheral membrane proteins?

Attached more weakly and can be released using milder treatments e.g. high salt, carbonate at high pH



Usually only attach via hydrophilic reactions

In order to span the bilayer, approximately how many residues must there be for an
a) alpha helix


b) beta sheet

a) 20-25


b) 7-9

What kind of reactions are involved in alpha helices?

- hydrophobic (amino acid side chains react with fatty acid tails)
- ionic interactions between protein and head groups (Hydrophilic)

What are some basic features of beta sheets?

Can be used to make barrel shape through the membrane - porins
12,16,18,22 strands
side chains facing out are hydrophobic and the ones facing in are hydrophilic

What are the three ways that water soluble proteins can be linked to the membrane (lipid anchored)?

Covalently linked to...
- Fatty acids via N terminus (acylation)
- Prenyls via two modifications of cys at or near C terminus (prenylation)
- GPI via PE and variable sugars at C terminus

Do proteins move more or less freely than lipids, on average?
What experiments can be used to show this?

Less freely - some are anchored
- FRAP, cell fusion experiments, treatment with 125I

What substances is the membrane permeable to?

Gases, ethanol, water (sort of), urea (sort of)

(gas + small uncharged molecules

What are the different types of membrane transport proteins and what are their relative speeds?

ATP powered pump, ion channel, transporters

What is the difference between an electrochemical and chemical gradient?

The electrochemical gradient applies to solutes that have a charge (which then affects their equilibrium). Neutral solutes just have a chemcial gradient based on concentration

What is the difference in terms of speed and specificity for carrier proteins and channel proteins?

Channels = faster, less specific

Carriers = slower, more specific

What is the delta G and how does it apply to the plasma membrane?

Delta G is the free energy change. Diffusing across the membrane requires an energy change (G). The magnitude of delta G determines whether a molecule will be able to passively diffuse or whether it will need a transporter

What is GLUT1?

Glucose transporter in erythrocytes implicated in diabetes. This transporter facilitates diffusion (50,000 x faster than diffusion alone). It is still passive transport and as such is dependent on the chemical gradient

What is the main CO2 transporter in erythrocytes?

Chloride bicarbonate exchanger
- antiporter of Cl- and HCO3- (this comes from the reaction of CO2 and H2O)


- antiporter of Cl- and HCO3-
- 12 TM helices

What is primary active transport?

Transport of a substance against its concentration gradient for which the energy comes from chemical reaction (ATP)

What is secondary active transport?

Transport of a substance against its concentration gradient via coupled transport. The ion gradients for secondary active transport are made by primary active transport.

How does vancomycin work?

Vancomycin is an ionophore that collapses the ion gradient of bacterial cells and kills them

What are the main classes of ATP pumps and give an example of each

P class pumps - include lots of the common ion pumps e.g. Na/K, H+, Ca2+ pumps

V class pumps - vacuolar membranes of plants and fungi

F class pumps - inner mitochondrial membrane

ABC superfamily - bacterial plasma membrane (transporters)

P type ATPases are mainly what kind of transporters?

Cation transporters
e.g. Na/K pump, Ca2+ pump

Explain the basic mechanism of the Na/K ATPase pump

Transporter binds 3Na+ from inside the cell (high affinity) --> phosphorylation via ATP (PEnzII) --> transporter opens up to extracellular space and releases the 3Na+, takes up 2K+ --> dephosphorylation of pump (EnzI) --> transporter releases 2K+ inside

What is the basic function of F and V type ATPases?

Proton transport

What is the basic function of ABC transporters?

Pumping amino acids, peptides, proteins, metal ions, lipids, compounds (drugs)

Give two examples of an ABC transporter

MDR1: multi drug transporter (resistance of bacteria to drugs)

CTFR: Cl- channel involved in cystic fibrosis

Give an example of a secondary transporter

H+/lactose co transporter




brings lactose into cell

What are aquaporins?

Water transporters (facilitated transport)

What are some of the specialised features of aquaporins to prevent H3O+ transport?

- Arg195 in aquaporin repels H3O+
- porin prevents chains of water being transported at a time which prevents 'proton hopping' between the water molecules

What are ion channels and how do they differ from transporters?

Channels straight through the membrane allowing passage of molecules that cannot naturally diffuse across

- Flux: unrestricted flow
- not saturable
- ligand or voltage gated
- open for only ms

What are some consequences of defective ion channels?

- defective Na+ channel: muscle paralysis/stiffness (tetrodotoxin)

- defective CFTR channel: cystic fibrosis

What is endocrine signalling?

hormone secretion into the blood to affect target tissue anywhere in the body

What is paracrine signalling?

cell secrete stimuli to an adjacent target cell e.g. immune cell secreting interferon

What is autocrine signalling?

Cell is stimulated by the substances that it secretes e.g. cancer cells producing their own growth factors

What is juxtacrine signalling?

signalling by plasma membrane proteins physically attached to the target cell e.g. killer t cells present an attached death ligand to infected cells

How can the magnitude of a cell's response to a stimuli be altered?

It can be limited by the number of receptors available to recognise the ligand

What are second messengers?

Transient chemicals that are released as a response to primary messengers (hormones/ligands etc) that mediate signalling e.g. cAMP, Ca2+

What are effectors?

Proteins that are stimulated in the signal transduction pathway that have some enzymatic function e.g. kinases, phosphatases

What is signal amplification?

Amplification of the signal (no way!) to vastly increase the cellular response to a single ligand/hormone. Enzyme activation and second messenger generation allows for rapid amplification

What is allosteric modification? Give an example

The ability of a molecule to alter the conformation of a protein when it binds non covalently to that protein

e.g. calmodulin changes its shape when Ca2+ binds to its EF hand motifs

What is covalent modification? give an example

A modification of the chemical structure of a target protein by covalent bonding. This process is reversible.

e.g. phosphorylation of a protein by kinases

How do allosteric/covalent modifications alter protein function?

1. activates enzyme activity


2. unmasks active sites


3. alters location of the protein
4. Alters stability of the protein
5. facilitates protein:protein interactions

What are kinases?

Proteins that phosphorylate other proteins to activate or inactivate them

What residues do kinases phosphorylate?

Serine, threonine, tyrosine

What do phosphorylation cascades allow for?

- signal amplification
- multiple regulation points
- divergence
- cross talk with other pathways

What is meant by cross talk between pathways?

Different receptors can stimulate the same signal transduction pathway

What does the EF hand motif do?

Binds calcium (e.g. calmodulin)

What does the SH2/PTB domains do?

Bind phosphotyrosine (tyrosine kinase receptor pathways)

What does the PH domain do?

Binds phosphoinosotides (PI-3 kinase pathway)

What are adaptor proteins?

Proteins that contain no functional activity but multiple interaction domains to function as a bridge between active proteins

What is a molecular switch?

Some proteins involved in signal transduction. Allosteric binding to other molecules results in a switch from off to on state

Give some examples of molecules that interact with GPCRs

- glucagon
- epinephrine
- vasopressin
- serotonin
- TSH

What are the majority of GPCRs involved in response to?

Olfactory signals

What is the basic structure of a GPCR?

7 TM alpha helical domains


4 cytosolic domains


4 extracellular domains

Explain the basic process of GPCR activation

Receptor binds hormone/ligand --> shape change in receptor allows it to interact with heterotrimeric G protein --> alpha G protein subunit exchanges GDP for GTP --> trimer dissociates and Ga goes on to activate receptor

How do G proteins act as molecular switches?

Exchange of GDP (off) for GTP (on)

What is the mechanism of the G alpha s protein?

Acts on adenylyl cyclase (excitatory)--> cAMP --> B adrenergic receptors

What is the mechanism of the G alpha i protein?

Acts on adenylyl cyclase (inhibitory) --> cAMP --> a2 adrenergic receptor

What is the mechanism of the G alpha q protein?

Acts on phospholipase C --> increased IP3 and DAG --> alpha 1 adrenergic receptor

What is the mechanism of the G alpha o protein?

Acts of phospholipase C --> Increased IP3 and DAG --> ACh receptor in endothelial cells

How does cAMP interact with PKA?

PKA has two regulatory subunits and two active subunits. cAMP induces the release of the regulatory units forming an active dimer

Explain the effect of epinephrine on adipose, liver, heart and GI/Kidney tissue

Adipose, liver, heart = stimulates B-AR, upregulates cAMP, via G alpha s --> fatty acid release, glucose release, increased contraction

GI tract/kidney = stimulates a2-AR to downregulate cAMP via G alpha i to induce vasoconstriction

When epinephrine stimulates the liver, what two enzymes does PKA then go on to effect?

Inhibitory phosphorylation on glycogen synthase
(stops glycogen synthesis)




Excitatory phosphorylation on glycogen phosphorylase (increases glucose)

What G alpha subunits are used for GPCRs to interact with phospholipase C?

o and q



In a GPCR process, what does phospholipase C do?

Cleaves PIP2 into IP3 and DAG

In a GPCR process, what does IP3 do?

Stimulates calcium release

In a GPCR process, what does DAG do?

(Along with calcium) activates PKC

How does calmodulin act as a molecular switch?

Binds 4 Ca2+ to its EF hand domains and undergoes a conformational change

What are some functions of calmodulin?

Activates calmodulin kinase
Mysosin light chain kinase
Adenylyl cyclase
Glycogen synthase kinase

What are three ways that a GPCR signalling pathway can be turned off?

Turn off the GPCR

Turn off G protein

Turn off second messenger

How can the GPCR be turned off?

Activated GPCRs are phosphorylated by GPCRkinase, which prevents G protein interaction and induces arrestin binding. Arrestin stimulates the internalisation of the receptor where it is then either degraded or recycled back to the surface

What is receptor exhaustion?

Repeated exposure to the same ligand over time can cause the mechanisms that turn signalling off to reduce the number of available receptors on the cell surface --> leads to the insensitivity of a tissue to a particular ligand

How are G proteins turned off?

G alpha proteins have intrinsic GTPase activity. This activity hydrolyses GTP. GTPase activity increases upon Ga binding to its receptor

How cAMP turned off?

cAMP is converted to AMP by phosphodieserases

What are the biding domains that recognise phosphorylated tyrosine?

SH2


PTB

Explain the basics of tyrosine kinase receptor activation

Ligand binding causes inactive monomers to dimerise and become active or inactive dimers to change shape and activate --> brings protein tyrosine kinases close together --> these can phosphorylate each other --> phosphoryation of tyrosine residues in the tail provide docking ports for new proteins

Explain the MAPK signalling pathway

Stimulated receptor binds GRB2 via a SH2 domain. GRB2 has an SH3 domain which it uses to bind SOS. SOS is a GEF which stimulates RAS to exchange GTP for GDP and become active. RAS binds RAF and stimulates the removal of the inhibitory 14-3-3 subunit. Active RAF phosphorylates MEK which then phosphorylates MAPK on tyrosine and threonine, forming an active dimer. MAPK active dimer travels to the nucleus and controls genes and TFs involved in cell proliferation and survival.

Explain the PI-3 kinase signalling pathway

RTKs recruit PI3 kinase via their SH2 domain. Activated PI3 kinase phosphorylates the 3rd position on the inositol ring of PIP/PIP2. This generates PI-3 phosphates, which bind AKT via its PH domain. Whilst bound, AKT (can also be called PKB) needs PDK1 (PI3 bound) and PDK2 to phosphorylate it and activate it. AKT then goes on to regulate processes such as cell survival and glucose uptake

Explain the basic phospholipase C pathway in tyrosine kinase receptors

PLC binds to the RTK via a SH2 domain. PLC then goes on to cleave PIP2 into IP3 and DAG, which go on to eventually activate PKC

What's the difference between a cytokine receptor and a tyrosine kinase receptor?

Both are structurally similar however cytokine receptors lack endogenous kinase activity and must bind an external kinase to signal

What is a common signal pathway through which cytokine receptors act?

JAK/STAT pathway

Explain the basics of the JAK/STAT pathway

ligand binds to cytokine receptor and stimulates dimerisation. Receptor binds JAK kinases which phosphorylate and activate each other and then other tyrosine residues on the receptor tail. STAT then binds to JAK via an SH2 domain, becomes active and dissociates forming a dimer with the other STAT molecule. This dimer then goes to the nucleus and binds DNA, activating transcription

What are some inhibitors of RTKs and cytokine receptors?

- phosphotyrosine phosphatases
- receptor internalisation by endocytosis


- other inhibitors (e.g. SOCs proteins)

What is SHP1 and how does it work?

SHP1 is an example of a SOC protein that inhibits cytokine receptor signalling. Inactive SHP1 binds to tyrosine residues (blocking them) in the receptor tail via an SH2 domain. An intrinsic phosphatase then dephosphorylates and inactivates JAK. The SOCs box then recruits an E3 ubiquitin ligase to stimulate the removal of the receptor.

SHP1 knockout mice have an extreme inflammatory phenotype

What's the difference between transcriptional regulators and transcription factors?

Transcriptional regulators = co activators, co repressors which modulate gene accessibility


Transcription factors = modulate gene expression initiation by RNA polymerase

What is NF-kB, what is it activated by and what covalent modification is central to its action?

An immune function regulator protein that provides a rapid transcriptional response to cellular stress

Activated by infection (e.g. TLRs) or inflammatory cytokines (IL-1, TNF)

Ubiquitination

What kind of ubiquitination is involved in the NF-kB signalling pathway?

K48 (structural scaffold rather than the degradation-inducing K63)

Explain the Nf-kB pathway

After the receptor (e.g. IL-1) activates ikB kinase, it then phosphorylates the inhibitor attached to the NF-kB heterodimer (p65 + p50). This phosphorylation attracts an E3 ubiquitin ligase which polyubiquitinates and removes the inhibitor. Free NF-kB can then travel to the nucleus and influence transcription

What kinds of things does the NF-kB pathway stimulate the production of?

Adhesion proteins


Cytokines
Enzymes
Chemokines
NF-kB inhibitor


IL-1RA

Explain the process of the IL receptor activating NF-kB

IL-1B binds to IL-1 receptor. Receptor activates and recruits MyD88 (adaptor), IRAK (kinase) and TRAF-6 (Ub ligase). TRAF-6 creates a K63 Ub scaffold to which TAK-1 (kinase) joins. The ikB kinase also joins (via the NEMO subunit) and is phosphorylated by TAK-1. The ikB can then dissociate and remove the NF-kB inhibitor

How is negative feedback used in the NF-kB pathway?

Both the ikB inhibitor and IL1RA are produced as a direct result of NF-kB activation which turns off the pathway

What was the new second messenger identified recently in the regulation of NF-kB and IFN?

cGAMP

What does TREX-1 do?

Stops human DNA leaked from the nucleus from stimulating the cGAMP pathway and triggering an immune/inflammatory response

Draw the cGAMP pathway

What are nuclear hormone receptors?

Transcription factors that are also receptors

What is an example of a homodimeric nuclear hormone receptor?

Heat shock proteins can act as an inhibitor to the nuclear receptors in the cytosol. Ligand binding kicks off the HSP, allowing it to translocate to the nucleus and bind to the response element section of DNA

Where are heterodimeric nuclear receptors often found?

In the nucleus, often already bound to DNA

(homodimeric are often found in the cytosol)

How do heterodimeric nuclear receptors change with ligand binding?

No ligand = bound to co repressors --> gene expression OFF

Ligand = bound to co activators --> gene expression ON

Binding of co regulators changes the chromatin structure giving access to genes (or not)

What does ligand binding do to the helix 12 position of nuclear receptors?

Ligand binding causes retraction of H12 and space for a co activator to join

No ligand attached means that H12 is extended leaving a big enough space for co repressors to bind

What is APO-LBD?

Nuclear hormone receptor with extended H12 and attached co repressors

What is HOLO-LBD?

Nuclear hormone receptor with H12 retracted with a small enough space that only co activators can bind

What enzyme do co repressors recruit and what does it do?

HDAC (histone deacetylase) which removes acetyl groups from chromatin, closing them off and inhibiting transcription

What enzyme do co activators recruit and what does it do?

recruit HAT (histone acetyl transferase) which acetylate chromatin, opening it up and allowing transcription to occur

What are the two broad classes of caspases?

Apoptotic and inflammatory

Caspases are what type of protease?

Cytesine

What are the two types of apoptotic caspases and how do they differ?

Initiator caspases: require clustering on a signalling hub in order to dimerise and activate




Executioner caspases: already dimeric, require cleavage to activate

What is apoptosis?

A 'neat and tidy' form of programmed cell death that involves the budding off of 'apoptotic blebs'- little bits of cell that can be easily phagocytosed by macrophages

What are the two types of apoptosis?

Intrinsic- signal coming from within e.g. cytochrome C found in the cytosol

Extrinsic - signal coming from outside e.g. death ligand presented by macrophage

How does the apoptosome form and how does it lead to cell death?

The apoptosome forms an oligomeric complex made up of APAF-1 after it binds to its ligand, cytochrome C. This apoptosome has a CARD (caspase activation and recruitment domain) to attract and activate caspase 9. Caspase 9 can then activate caspase 3 which goes on to cleave substrates and initiate apoptosis.

What is pyroptosis?

A form of planned necrosis that involves the rupture of the plasma membrane and spilling of cellular contents (as well as the release of cytokines) leading to an inflammatory response.

What mediates apoptosis?

Apoptotic caspases which are activated via the intrinsic or extrinsic pathway

What mediates pyroptosis?

Inflammatory caspases activated by the inflammasome

What is the ASC protein and what does it do?

It is an adaptor protein involved in the activation of capases by the inflammasome. It has a PYD domain to attach to the inflammasome nucleating protein and a CARD domain to attach to the caspase, serving as the connector between the two.

How is the ASC protein prion-like? What is the ASC speck?

The ASC protein is prion like because it can exist in two states. When it is in its prion like state, the ASC protein can induce other ASC proteins to adopt this same state.

In a resting cell, ASC is spread throughout the cytosol. When activated via the inflammasome, ASC condenses and forms long, pyrin domain fibrillates. This is the scaffold for caspase 1 attachment and activation, also known as the ASC speck.

Explain runaway inflammasome signalling

Receptor activation in response to infection --> ASC speck formation --> cytokine release and cell death (pyroptosis) --> ASC release --> extracellular activation of caspase 1 and IL-1B --> phagocytosis of ASC speck --> perpetuation of signalling in neighbouring cell