• 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

1/234

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;

234 Cards in this Set

  • Front
  • Back

Give some basic features of microfilaments

Smallest of the cytoskeletal features (7-9nm)
composed of actin which interacts with myosin and other proteins
can occur in bundles
Important in cell motility and membrane shape

Give some basic features of intermediate filaments

medium size (10 nm)
made up of desmin, cytokeratin, neural filaments, lamin
provide tensile strength and support

Give some basic features of microtubules

Largest cytoskeletal element (25 nm)


alpha beta tubulin dimers
extend throughout the cell


make up structure of mitotic spindle
transport and support (cilia, flagella)

What is G actin?

Globular, monomeric actin

What is F actin?

Filamentous, polymeric actin

Give some examples of cellular structures which utilise actin

microvilli, filopodia, lamellopodia, contractile ring

What are the three stages of actin polymerisation?

1. nucleation (assembly of actin monomers to >3)
2. Elongation (addition of actin monomers to both ends)
3. Steady state (addition of monomers to (+) end and removal from (-) end)

What is the rate limiting step in actin polymerisation?

Nucleation

Explain the idea of actin polarity and treadmilling

Actin is constantly polymerising and depolymerising in the cell. ATP-G actin is always added to (+) end, then ATP is hydrolysed to ADP + P. ADP-G actin is then removed at (-) end causing treadmilling of the actin fibre

What other proteins are involved in the treadmilling process and what do they do?

Profillin: promotes ATP-G actin formation for the (+) end, regulates formins

Cofillin: Destabilises ADP-G actin enhancing disassembly at (-) end

Thymosin B4: sequesters and acts as a ATP-G actin buffer for the (+) end

What are the two different classes of actin nucleating proteins?

Formins = assemble unbranched filaments

Arp 2/3 = assemble branched filaments

What are formins and how do they work?

Proteins that regulate actin assembly. Have an FH2 domain that forms a dimer, which then binds two actin subunits. By rocking back and forth, additional actin monomers are added. Regulated by profillin. Produces unbranched filaments

How does WASp and Arp2/3 regulate branched actin assembly?

Cdc42 activates WASp (takes it out of its autoinhibited state). Active WASp then binds to and activates Arp2/3 by inducing a conformational change. Arp2/3 then forms a weak association with the actin filament. WASp also binds to G actin which binds to and strengthens the Arp2/3-actin connection. Branch nucleation and polymerisation then occurs

How is actin used in endocytosis?

Endocytosis factors (e.g. clathrin) recruit WASp. WASp promotes Arp2/3 assembly of branched actin filaments which help transport the endocytosed vesicle into the cell

How is muscular dystrophy linked with actin?

Dystrophin is a protein that links actin to the cell membrane

- mutation in the dystrophin gene means that cycles of muscle contraction weakens the plasma membrane resulting in death of the myofibril and muscular atrophy

What are microfilament motors?

Actin acts as a track for myosin motor motility

Tropomyosin - heterodimer that blocks myosin biding sites on actin

Troponin (T/I/C) - binds Ca2+, changes shape, removes tropomyosin from myosin binding sites

Where are type I myosins found?

Found in cell periphery

Where are type II myosins found and what do they do?

Found in muscle and non-muscle cells. required for cytokinesis and focal adhesion

What is myosin V?

A type of myosin required for organelle transport

Explain the power stroke?

ATP bound to mysosin (low energy) --> ATP hydrolysis (high energy state) --> ATP bound myosin binds actin --> Pi dissociates --> ADP dissociates --> power stroke --> ATP binds and starts the process again

What determines the rate of myosin movement?

Length of neck

What is the sliding filament model?

The idea that during sarcomere contraction, there is no actual change in the length of the fibres - just varying levels of overlap

Where is myosin II found and what does it do?

Found in stress fibres and required for cytokinesis

What does myosin I do?

Found in cell periphery

Single headed motors that operate via ATP hydrolysis - required for cell motility


Type I myosins are required to take membranous organelles to the membrane - eg vesicle travelling down actin (via myosin I) fuse with the membrane to allow the cell to extend out and move in one direction.

Explain the basic process of cell migration

1. Lamellipodia extension (actin crosslinking)
2. Formation of new focal adhesions
3. Bulk of cytoplasm moves forward due to contraction at rear of cell
4. Detachment of rear by specific proteolytic enzymes

What does Rho do?

Regulates stress fibre formation

What does Rac do?

Regulates lamellipodia formation

What does cdc42 do?

Regulates filopodia formation


(and WASp activation)

In a moving cell, where would Rho, Rac and cdc42 be active?

Rear: Rho activation --> stress fibre formation and mysoin II (cytokinesis) activation

Front: Rac activation --> Arp 2/3 activation (inhibited by Rho), cdc42 activation

What is chemotaxis and how does it lead to cell movement?

The sensing and movement towards chemical gradients by motile cells. Chemokines bind to GPCRs which results in PIP3 formation. PIP3 activates Rac to induce actin configuration changes.


To detect gradients, cells can redistribute receptors

In cell migration, various receptors, signalling molecules and cytoskeletal elements are moved around. What are some examples of elements that would be found at the rear and the front of the cell?

Rear = myosin II, lipid phosphatase

Front = lipid kinase, actin, PI(3,4,5)P3

What are some structural features of microtubules?

Heterodimers of alpha and beta tubulin

Microtubules are made of 13 laterally associated protofilaments to form a tube


What is the microtubule organising centre?

An anchor for the (-) end of microtubules, found near the nucleus, which extends MTs out into the cell

also called the centrosome

How does the MTOC change during mitosis?

during mitosis, cells completely reassemble their microtubules to form bipolar spindles


Spindle assembles from two MTOCs, called spindle poles

Explain the idea of microtubule treadmilling

alpha/beta tubulin dimers bind GTP and add to (+) end of microtubules.
After dimer is incorporated into the microtubule, GTP hydrolysis occurs (beta subunit). Dimers bound to GTP tend to assemble into microtubules whilst those bound to GDP tend to fall apart.

What is anterograde transport?

Transport towards the periphery (+)

KINESIN

What is retrograde transport?

Transport towards the MTOC (-)

DYNEIN

Explain how kinesin movement occurs

1. Leading head binds, releases ADP, causing strong leading head connection and weak lagging head connection
2. Leading head binds ATP
3. Conformational change causes lagging head to move forward
4. Lagging head becomes leading head, binds tubulin, releases ADP and binds strongly. Trailing leg hydrolyses ATP to ADP
5. Lagging head then moves forward and the cycle repeats

Kinesin movement is highly processive- what does this mean?

That it involves catalysing consecutive reactions without releasing the substrate

What governs the volocity of kinesin movement?

ATP hydrolysis

(dependent on ATP/ADP levels in both heads)

How do kinesin and dynein connect to cargo vesicles?

Both kinesin and dynein require adaptor proteins to join to the cargo vesicles

What is kinesin's adaptor protein to connect to cargo vesicles?

Kinectin

What is dynein's adaptor protein to connect to cargo vesicles?

Dynactin complex

What is the basic structure of cilia?

Consist of a singlet pair of microtubules surrounded by 9 doublets (9 + 2 arrangement)

Held together by nexins, radial spokes and dynein (motor protein)

Why do cilia and flagella bend?

Cilia and flagella bend due to dynein activation- dynein on A subunit of one doublet walks along the B subunit on the adjacent doublet. Because they're connected by nexin, the cilia bends

How are microtubules used during mitosis?

Spindle microtubules attach to the kinetochore and pull chromosomes apart via a combination of sliding and treadmilling

The cytoskeleton is a common target for the treatment of what disease?

Cancer

What is vinblastine? How does it work?

A plant alkaloid used as a cancer treatment
- used as chemotherapy for Hodgkin's disease, non-hodgkin's lymphoma, breast and other cancers

Inhibits microtubule assembly by binding to B tubulin subunits at the (+) end --> disrupts assembly and spindle formation in M phase

What is Paclitaxel (taxol)? How does it work?

An anti-cancer drug used in ovarian, breast, lung, bladder and other solid tumor cancers

Binds to B tubulin subunits (like vinblastine) however it results in stabilisation of microtubule and blocks metaphase spindle formation

Do IFs have polarity?

No - they are not motors

no treadmilling


Although they are normally stable, IFs do have to depolymerise for cell division

Give some examples of different types of IFs

Keratins (epithelial cells), vimentin (mesenchymal cells), desmin (muscle cells), lamins (nucleus)

What is an example of an intermediate filament disease?

Epidermylosis bullosa simplex
- abnormal blistering (dermis-epidermis separation)


- due to mutation in keratin 14

Why aren't there many microtubule/microfilament diseases?

Because these tend to be embryonic lethal

What is fibronectin, and how does its expression differ between normal cells and cancer cells?

it is a component of the extracellular matrix

normal cells produce a lot of fibronectin whilst cancer cells produce less- in general, cancer cells are less strongly adhered to one another which may facilitate metastasis

Antibodies against fibronectin have what effect if present during development?

Inhibit branching morphogenesis in salivary glands

What occurs if collagen II is deleted?

Dwarfism

How do cells attach to the ECM?

Receptors for ECM components = integrins. Integrins are heterodimers of different alpha and beta chains (different combinations give different specificity for what they bind to)

Alpha and beta subunit bind ECM component, beta subunit binds talin in the cytoplasm. Vinculin binds and connects talin and actin. Cell-ECM links are adherens junctions

How do cell-ECM links change as the cell moves?

Cell-ECM adhesion plaques stay in the same spot as the cell moves over the top of them

When the cell moves all the way over them, they break off the tail end

Explain cell-cell adherens junctions

Cadherins are the transmembrane proteins, which are linked to actin inside the cell by alpha and beta catenins (alpha joins actin, beta joins cadherin).

P120 catenins, vinculin and VASP also assist actin-cadherin connection. Alpha actinin joins actin segments together

What do cadherins do?

They are transmembrane proteins that link cells to other cells via adherens junctions

Mediate homotypic interactions - calcium dependent

Linked to actin filaments via alpha, beta and P120 catenins, as well as vinculin and VASP

What are podosomes and how are they relevant to cancer?

Adhesion structures in cells containing Arp2/3. WASp and cortactin. Pososomes, named invadopodia, break down intracellular matrix and direct secretion of matrix metalloproteases

What is the fixation step for light microscopy?

Specimen is fixed in a chemical (e.g. formaldehyde) causing cross linking of protein in the tissue to prevent distortion

What is the embedding step for light microscopy?

Tissue is dehydrated (ethanol) then 'cleared' with solvent (e.g. chloroform), placed into melted wax and left to cool and solidify

What is the sectioning step for light microscopy?

Block of tissue cut into 5 um sections using a knife held in a microtome

What is a HE stain and what colours does it produce in what parts of tissue?

Haematoxylin = basic stain, stains acidic structures (DNA) purple


Eosin = acidic stain, stains basic structures (cytoplamic proteins etc) purple

What would a normal cell look like with a H&E stain?

Purple nucleus, pink cytoplasm

What is IHC useful for?

Analysing spatial expression of target antigens, not so much the quantative expression levels (western blots/ELISA are better)

What are the major steps of IHC?

Fixation and antigen retrieval
Quenching/blocking endogenous activity
Blocking non specific binding
Immunodetection


Visualisation of antibody
Counterstaining

What is the fixation step of IHC?

Formaldehyde generates methylene bridges that crosslink proteins in tissue sample to protect morphology. However these can hide antigenic sites so heat or enzymes are used to partially break the bridges

How is endogenous target activity blocked during IHC

Immunodetection that relies of enzymes such as peroxidase needs to have endogenous activity blocked (eg by incubation with hydrogen peroxide) to prevent excessive background staining

How is non specific protein binding prevented in IHC?

Tissue sections are incubated with buffer (skim milk, BSA) which should bind and block proteins to which antibodies may weakly bind, preventing false positives. Detergents are often inhibit non-specific hydrophobic reactions

What are the two types of immunodetection in IHC?

Direct - reporter is conjugated to an antibody (primary)

Indirect - primary antibody against antigen + anti-antibody with reporter attached

Does direct or indirect immunodetection have higher sensitivity?

Indirect

How does visualisation of the antibody occur in IHC?

Through chromogenic or fluorescent means

Chromogenic - conversion of soluble substrate into coloured, insoluble precipitate by enzymes
(HRP catalyses oxidation of DAB to form brown precipitates)

Fluorescent detection simply has fluorescent reporters

What is counterstaining in IHC?

A different stain is added to the tissue to contrast the primary IHC stain e.g. haematoxylin to stain nuclei purple

What is the primary structure of a protein?

Amino acid sequence

What is the secondary structure of a protein?

Alpha helix and beta sheets

What is the tertiary structure of a protein?

The protein's 3D structure

What is the quaternary structure of a protein?

Dimerisation, oligomerisation

What are structural motifs?

Combinations of secondary structures e.g. ring finger, zinc finger motifs. For non-covalent interactions (commonly divalent)

What are protein domains?

Distinct regions of tertiary structure which can have its own structural and functional properties (enzymatic etc).

What is a Svedberg unit?

The rate of sedimentation on a centrifuge


Related to size, shape and density. Non standard, non-linear (50S might not be twice the size of 25S)

What is the benefit of multi subunit complexes vs modular domains in a protein?

Modular domains can't be swapped out

Multi subunit complexes provide flexibility and diversity through different combinations

What are post translational modifications?

covalent modifications that change a protein's structure
(changed activity, target for degradation, change cellular location)

What is phosphorylation?

The addition of a phosphate group to a serine, tyrosine or histidine by kinases

What is ubiquitination?

The addition of one or more ubiquitin peptides to lysine residues

What is allosteric regulation?

The change in protein structure/function due to non-covalent binding of a ligand

What is calmodulin?

An example of allosteric regulation where Ca binding changes the tertiary structure allowing it to bind to other proteins

What does a GEF do?

switches GDP out for GTP

What does a GAP do?

switches GTP out for GDP

What are chaperonins?

Barrel shaped folding machine, with a lid homoheptamer, which together fold proteins powered by ATP

What are the major features of the nuclear membrane?

The nucleus has an outer and inner membrane. The inner membrane defines the nucleus and the outer is continuous with the rough ER. Has nuclear pores for key protein exchange


What is the nuclear lamina?

A meshwork of intermediate filaments (lamins) which interconnect with nuclear pores - provides structure to the nucleus

What is the nucleolus?

A suborganelle within the nucleus, with no membrane, the site of ribosome biogenesis - hotspot of transcriptional activity

What are nuclear bodies?

Membraneless sub compartments, concentrated regions of protein and RNA transcription and processing. Formation may enhance process efficiency

What is chromatin?

A complex of DNA and histone proteins (nucleosomes) which has a dynamic structure that determines gene expression

How it chromatin structure regulated?

Histone tails extending from a nucleosome can be targets for post translational modification

What is chromatin acetylation?

PTM that makes the chromatin less condensed and more transcriptionally active


Euchromatin


done by histone acetylase transferase (HAT)

What is chromatin deacetylation?

A highly condensed form of chromatin that is less transcriptionally active

Heterochromatin

Done by histone deacetylase (HDAC)

What is the nuclear pore complex?

A huge structure in the nuclear membrane made up of ~30 different nucleoporin (Nup) proteins


Cytoplasmic and nuclear asymmetry

Different nups for scaffold, linker, transmembrane anchor, barrier and transport nups.

What are nuclear localisation sequences?

Amino acid sequences that target a protien to the nucleus, generally internal.

Import = nuclear localisation sequence (NLS)
Export = nuclear export sequence (NES)


What are importins and exportins?

Cargo carrying complexes that overcome the size limit of the NPC, allowing proteins to be brought in/out. They are receptors for FG nups in the NPC, and recognise the nuclear import/export sequences on the cargo

How is nuclear import/export controlled?

Uses a GTPase swtich - RanGTP/GDP

What are the steps of nuclear import?

Importin binds NLE on cargo and moves into the nucleus, along with RanGDP.
Chromatin-bound GEF changes RanGDP to RanGTP
RanGTP binds to importins and causes them to release the cargo
RanGTP-Importin moves out to the cytoplasm
Cytoplasmic nups on NPC are GAPs which change RanGTP to RanGDP. Ran-importin dissociate and the process starts again.

Show nuclear import with a diagram

What drives the movement of the importing complex across the nuclear membrane?

More GTP inside nucleus (drives GTP out)

More GDP outside nucleus (drives GDP in)

Explain the nuclear export mechanism

Exportin and RanGDP diffuse into nucleus
Chromatin GEF changes RanGDP to RanGTP
Cargo, exportin and RanGTP form complex and travel out
RanGTP hydrolysis causes dissociation of cargo and exportin, cycle starts again

What are laminopathies?

Genetic mutations that affect lamins, nuclear proteins connected to lamins or proteins involved in lamin maturation

Give some examples of laminopathies

Muscular dystrophies and cardiomyopathies
Familial partial lipodystrophy
Peripheral and sensory neuropathies
Premature ageing (preogeria)

What is Hutchinson-Gilford progeria syndrome?

Accelerated ageing, premature hair loss, muscle wasting, fat loss, reduced bone density

Mutations cause accumulation of unprocessed lamins (prelamins) or misprocessed lamins (progerins)


Inability to repair DNA damage due to damaged lamins causes premature ageing

What are the morphological differences between the smooth and rough ER?

Rough = sheetlike cisternae - flattened membrane, covered with ribosomes

Smooth - branched, tubular morphology

How does the ER membrane curve?

reticulons inserted into the membrane are responsible for curvature

How is 3 way branching acheived in the ER?

Comes about via membrane fusion between GTPase alastins in the membrane. When two membranes come close, alastins dimerise and fuse

Where is the ER targeting sequence on proteins?

Signal located at the N terminal of the growing peptide

What is cotranslational translocation?

ER targeting must happen at the same time as protein synthesis

Explain the basic process of cotranslational translocation

Signal recognition particle recognises the ER targeting sequence on the growing peptide in the ribosome. SRP moves to the SRP receptor near the translocon on the ER membrane. GTP hydrolysis in both the SRP and the SRP receptor. Translocon opens and signal sequence is cleaved by signal peptidase. Translation stops, ribosome dissociates and protein folds in the ER.

Explain cotranslational translocation with a diagram

Show the different types of ER membrane proteins conformations

What is a type 1 ER membrane protein?

A protein which is inserted into the ER membrane, with the C terminal outside and the N terminal inside. Majority of protein is inside. Needs a stop transfer anchor. Has a cleaved terminal ER targeting sequence

What is a type 2 ER membrane protein?

Does not have a cleavable N terminal signal sequence - internal targeting signal anchor sequence instead.
Translation initially occurs in the cytoplasm, until the targeting sequence is recognised by the SRP and brought to the translocon.

N terminal outside, most of protein inside

What is a type 3 ER membrane protein?

No cleavable N terminal sequence - signal anchor sequence close to N terminal.

Signal anchor sequence recognised by SRP and brought to translocon

How do type 2 and 3 ER membrane proteins have different conformations?

The location of +ve charged residues --> these prefer to stay on the cytosolic side


Type 2 = before SA sequence = N out
Type 3 = after SA sequence = C out

What kind of modifications occur in the ER?

Glycosylation
Disulfide bond formation
Proteolysis
Assemble quaternary structure

What is hereditary spastic paraplegia?

Progressive stiffness, contraction (spasticity), loss of coordination and lower limb weakness
peripheral neuropathy

Caused by hereditary autosomal mutations --> mainly affecting reticulons or alastins causing defective ER branching and architecture

ER has to span the entire length of axons so any defect can cause neuronal dysfunction

Does the golgi have quality control mechanisms?

No - unlike the ER.

Deleting golgi enzymes may not stop secretion

What is the basic structure of the golgi apparatus?

Parallel stacks of flattened membrane discs connected to form ribbons. Transitions from cis golgi network to cis golgi to medial golgi to trans golgi to trans golgi network

What are the cis and trans golgi network?

Fenestrated tubular netowrks that receive and secreted vesicles respectively

What two proteins hold the golgi together?

GRASPs = golgi reassembly and stacking proteins that dimerise/oligomerise to facilitate stacking


Golgins = coiled, rod-like proteins that facilitate tethering


Microtubules
are required to maintain the golgi structure

What is anterograde golgi transport and what facilitates it?

Transport from ER to Golgi

COPII

What is retrograde golgi transport and what facilitates it?

Transport from golgi to ER

COPI

How is cargo sorted into vesicles to be transported to the golgi?

Membrane cargo proteins = sorting signal on the cytoplasmic domain recognised by coat proteins

Soluble cargo proteins = require recognition by membrane bound receptors which themselves have cytoplasmic signals for coat proteins

What is retrograde retrieval? Give an example of a protein that has to be recovered in this way?

Sorting signals are required to maintain residency within a compartment

ER residents randomly moved to the golgi require retrieval by retrograde COPI transport

Sorting signal = KDEL sequence
slight pH difference in golgi facilitates KDEL sequence binding to receptor and retrieval

What is cisternal maturation?

Each membrane compartment over time matures to become the 'next' on e.g. cis golgi matures to become the medial golgi.

Retrograde transport maintains resident enzymes

How are the different golgi compartments funtionally different?

Each compartment has its own resident enzymes.

Cis golgi glycans become the substrate for medial golgi enzymes and so on

What are congenital defects of glycosylation?

A complex and highly heterogenous disease resulting from deficiencies in glycan modifying enzymes in golgi compartments --> reduced branching and glycan extension

Clinical presentation includes: psychomotor retardation, cutis laxa (wrinkled, inelastic skin), dysmorphic features, autoimmune disease, MD

What is significant about the way that mitochondrial DNA is inherited?

Inherited cytoplasmically and maternally

What does mitochondrial DNA code for?

Always codes for mitochondrial proteins, but most of the coding is done by the nucleus

What does the mitochondrial matrix contain?

mtDNA and mitochondrial ribosomes

What are the different shapes that mitochondria can take on?

Can be individual spheroids (M phase) to long elongated networks (G1/S)

How does mitochondrial fission occur?

Mitochondrial fission factors recruit G proteins that hydrolyse GTP to constrict membranes and separate mitochondria

RECRUIT G PROTEINS

How does mitochondrial fusion occur?

Mitofusins are G proteins that hydrolyse GTP to fuse membranes
Different mitofusins on outer and inner membranes - outer membrane mitofusins must meet first

THESE ARE G PROTEINS

What are the features of the mitochondrial targeting sequence?

20-50aa N terminal cleaved sequence.

Amphipathic, alpha helix sequence

Compare the ER, mitochondrial and nucleus targeting sequences

ER = N terminus, cleaved, short hydrophobic

Mitochondria = N terminus, cleaved, long, amphipathic hydrophobic alpha helix

Nucleus = location varies, not cleaved, lys/arg rich

What is significant about the cargo for mitochondrial transport?

Protein synthesis occurs in the cytoplasm but proteins are kept unfolded by chaperones (cytosolic Hsc70)

Requires ATP

Folded proteins cannot undergo mitochondrial transport

What translocons are involved in mitochondrial transport?

To get to the matrix (if that's where they're directed to go) proteins must go through the outer membrane translocon (TOM) and the inner membrane translocon (TIM) SIMULTANEOUSLY

For this to happen, the membranes must be in close proximity

Explain the basic process of mitochondrial transport

Protein synthesis in cytoplasm, protein is kept unfolded by Hsc70 (no ribosome directly involved - no cotranslational translocation)

Targeting sequence recognised by import receptor and directed to TOM complex --> passes through TOM and TIM simuntaneously

During translocation, protein is bound by matrix Hsc70 which hydrolyses ATP to pull cargo through

Targeting sequence cleaved by protease
and cargo is folded into mature state

What is oxidative phosphorylation?

Coupling a series of oxidation/reduction reactions with phosphorylation of ADP to generate ATP

What are NAD and FAD?

High energy electron carriers

What is stage one of oxidative phosphorylation?

Sugar and fat metabolism

Sugars --> pyruvate
Fatty acids --> Fatty acyl coA

What is stage two of oxidative phosphorylation?

Giving up electrons

Fatty acyl coA/pyruvate --> Acetyl CoA
Electrons donated to NAD and FAD to form NADH and FADH2

Acetyl CoA --> citric acid cycle to generate more FADH2 and NADH

Sugars and lipids eventually oxidised to CO2

What is stage three of oxidative phosphorylation?

Pumping protons

FADH2 & NADH donate their electrons to a series of transmembrane inner membrane proteins - electrons shuttled between complexes by mobile carriers.
Energy from e- transport is used to pump H+ out of the matrix, making a voltage gradient across the inner membrane

What is stage four of oxidative phosphorylation?

Generating ATP

Electrochemical gradient provides the proton motive force for ATP synthase which, through chemoosmosis converts H+ movement to ATP generation

How is mitochondrial fission/fusion used for quality control?

Damaged components are segregated and removed through fission and targeted for degradation

What is PINK and Parkin and how are they related to Parkinson's disease?

PINK is a kinase and Parkin is a Ub ligase - they are required to prevent fusion of damaged mitochondria back into the healthy pool

PINK accumulates in damaged cells and recruits Parkin to the outer membrane. Parkin ubiquitinates mitofusins which prevents fusion and targets the damaged parts for degradation

How was the secretory pathway first visualised?

Using a temperature sensitive mutant of vesicular stomatitis virus G
Restrictive temp - protein was unfolded and stuck in ER
Permissive temp - protein went from ER to golgi to plasma membrane

What is the basic process of vesicular transport?

Cargo in donor cell (soluble/membrane bound)
Protein coat + budding
Scission
Uncoating
Translocation via cytoskeleton
Tethering
SNARE assembly
Fusion

What is a class A sec mutant?

Accumulation of protein in cytosol

Defective transport to ER

What is a class B sec mutant?

Accumulation of protein in rough ER

Defective release from ER

What is a class C sec mutant?

Accumulation of protein in ER --> golgi vesicles

Defective golgi fusion

What is a class D sec mutant?

Accumulation of protein in golgi

Defective transport to secretory vesicles

What is a class E sec mutant?

Accumulation of protein in secretory vesicles

Defective transport to cell surface

What are the two major types of cargo involved in secretion?

Passengers to be moved (e.g. newly synthesised proteins)

Crew to enable transport to occur (e.g. SNAREs) which must be regulated and retrieved via COPI

Where does COPII transport go to and what major proteins are involved?

ER --> cis golgi transport

Coat proteins = sec23/24 and sec13/31
GEF = sec12
GTPase = sar1
GAP = sec23

Where does COPI transport go to and what major proteins are involved?

cis golgi --> ER

Coat proteins = coatamers with 7 different COP subunits
GTPase = Arf

What provides the selectivity for COP transport?

Different coat proteins/GTPase provide specificity and selectivity

Why will some cargo bind to a receptor in the golgi and not in the ER?

The capacity for soluble cargo to engage receptors and be transported depends on the binding properties which differ between compartments



E.g. comparment differences = pH, PTMs

exmple = KDEL receptor binds KDEL signal on ER resident proteins only in the golgi because of the golgi's lower pH

Give an example of a type of soluble protein that undergoes retrograde transport

ER resident proteins with a KDEL (lys-asp-glu-leu) binds to the KDEL receptor in the lower pH of the golgi and is returned to the ER via a COPI vesicle

How is the KKXX signal sorted and transported?

KKXX is a sorting signal on ER resident membrane proteins
It is transported back to the ER via a COPI vesicle (COPI alpha and beta subunits recognise the cytoplasmic KKXX domain of the protein)

How is the X-Arg-Arg-X signal sorted and transported?

X-Arg-Arg-X is a sorting signal on ER resident membrane proteins
It is transported back to the ER via a COPI vesicle (COPI alpha and beta subunits recognise the cytoplasmic X-Arg-Arg-X domain of the protein)

Explain the basic process of COPII vesicle formation

Sar1 in its cytoplasmic GDP form interacts with sec12 (a membrane bound GEF), and is transformed into its GTP, ER-membrane-bound form. Sar1 GTP, in addition to sec23/24 interact with the cytosolic signalling domains of particular cargo (which is sorted to go to the golgi) forming the prebudding complex. Sec 13/31 is then recruited, and polymerisation of these proteins occurs until a bud is formed. The bud then undergoes scission by dynamin to become a free-floating, coated vesicle. The coat is then removed when sar1 hydrolyses GTP (sec23 is the GAP). Coat disassembly produces a free floating, uncoated vesicle which travels via the microtubule system to the golgi

Explain the mechanism of vesicle fusion

Free floating vesicle has VAMPs and RabGTP
RabGTP docks on the Rab effector on the acceptor membrane, GTP hydrolysis stabilises binding.
VAMP interacts with SNAP25 and syntaxin to form the SNARE complex (2 x SNAP25, 1 x VAMP, 1 x syntaxin).
Fusion occurs
NSF/alpha SNAP disassemble SNARE complex by ATP hydrolysis - VAMPs are returned

What are the GTPases involved in COPII, COPI and vesicle fusion?

COPII = Sar1
COPI = Arf
fusion = Rab

What are the components of a SNARE complex in vesicle fusion?

2 x SNAP25
1 x syntaxin


1 x VAMP (vesicle)

What is the segregation sorting mechanism?

Having different proteins in the cytoplasm and lumen that physically will not meet

What is an active sorting signal?

Sorting signals on proteins that will include them in transport vesicles - can be just the protein (membrane bound) or through protein-protein interactions (soluble protein)

How does active retention work as a sorting mechanism?

Protein retained in cell due to interactions e.g. with another complex too big to bud off

How does retrieval work as a sorting mechanism?

A specific sorting mechanism on the protein allows it to be retrieved e.g. KDEL

What are the two major types of secretion?

Constitutive
Regulated

Why does the golgi have different compartments?

So the various processes (e.g. glycosylation) can be completed correctly and in the correct order (if all the proteins and enzymes were in one big compartment there would be many errors)

What is the evidence for cisternal maturation?

- live cell imaging (fluorescent tagging of yeas cell golgi shows transition from cis (green) to trans (red))

- big molecules (e.g. laminin) could not fit in secretory vesicle and hence must just stay in the maturing golgi cisternae

What is constitutive secretion? Give an example

Sorting signal independent - used when protein needs to be constantly secreted (bulk flow). Soluble and transmembrane proteins included - level of secreted protein depends on the level produced by individual cells

e.g. albumin (blood protein)

What is regulated secretion? Give an example

Sorting signal dependent pathway that involves protein being accumulated in vesicles and released in response to a stimulus.




e.g. insulin (seen in its proinsulin form in new secretory vesicles and mature form in vesicles near the membrane)

Explain the regulated secretion of insulin

Munc molecules are key regulators - bind syntaxin and prevent SNARE complex formation

Uncoated transport vesicle
Rab-receptor activation
Munc binding
Vesicular - target SNARE interaction is stimulated when release signal (e.g. Ca2+ influx) deactivates the munc blocking)
Fusion and release

What is I cell disease?

A disease characterised by a deficiency of lysosomal acid hydrolase proteins leading to build up of substances in the cell that can not be degraded as they should (inclusions). A deficiency of a phosphatase in the golgi means that lysosome-bound proteins are secreted normally because they don't have a mannose-6-phosphate signal (to be recognised by the M6P signal and sent to the golgi). This results in growth and physical defects.

Explain the mannose-6-phosphate pathway

The M6PR in the golgi recognises the M6P signal on lysosyme-bound proteins (acid hydrolases). When bound in the TGN, this complex stimualtes clathrin-coated vesicle (via adaptins) transport to the late endosome. Here, lower pH stimulates receptor-ligand uncoupling - receptor is transported via retromer back to the TGN or to the cell surface. Any acid hydrolyase that was randomly secreted via constitutive secretion can be brought back via this surface receptor (endocytosis). As the late endosome matures to become a lysosome, the acid hydrolase becomes active

What are tethers?

Peripheral membrane proteins that reach out into the cytoplasm to sense and bind particular vesicles. They don't have quite the same specificity as SNAREs, and are more about increasing the probability of successful SNARE interaction. They often involve a small GTPase (Rab).

What are the major functions of plasma membrane proteins?

Transport
Attachment to cytoskeleton
Cell adhesion
Identification markers
Enzyme
Surface receptor

Why does a cell need endocytosis?

Uptake of nutrients
General housekeeping
Regulation of surface proteins
Pathogen entry
Immunity
Control of cell signalling events

What is LDL?

Low density lipoprotein - a structure through which cholesterol can be absorbed in our bodies

Explain the mechanisms behind LDL binding/uncoupling from its receptor

LDL-R is on the cell membrane and has an internal cytoplasmic sorting signal. Signal = NPXY (Asn-Pro-Val-Tyr). Adaptin binds NPXY signal and initiates endocytosis by recruiting clathrin.
Different adaptins bind different sorting signals - specificity.
LDL binds at high pH (extracellular) and disassociates at low pH (endosome)

What sorting signal is on LDL?

NPXY

How does LDL bind to its receptor?

At high pH (extracellular) the ligand binding arm binds apoprotein B

How does LDL unbind from its receptor?

At low pH (endosome) the B propellor domain becomes +vely charged and binds the ligand binding arm, releasing LDL

Explain the basic LDL pathway

LDL-R N terminus binds LDL, C terminus binds adaptin. Adaptin recruits clathrin which stimulates endocytosis bud formation. Bud undergoes scission when fully formed, by dynamin. Clathrin coat disassembly occurs. Uncoated vesicle duses with endosome, where low pH causes receptor and LDL to dissociate. Receptor is brought back to surface by recycling endocytosis, LDL stays in maturing endosome and is eventually broken down into cholesterol/fatty acids etc

What is the functional unit of clathrin?

Triskellion

What is dynamin?

A GTPase that creates a ring, hydrolyses GTP causing a conformational change that pinches off a membrane bud

How is actin polymerisation used in endocytosis?

Actin polymerisation (Arp2/3, WASp etc) is used to drive the vesicle away from the membrane (particularly bigger vesicles).

What is coincidence detection?

Recruitment of a protein to a particular point can depend on a number of factors e.g. lipids and proteins that must both be present for binding to occur

What is familial hypercholerterolemia?

excess LDL in blood - recessive single gene mutations in LDL receptor
(N domain = LDL doesn't bind receptor, C domain = LDLR not internalised)

Severe atherosclerosis at an early age

What is macropinocytosis?

bulk fluid phase endocytosis - not selective for cargo. Regulated by growth factor receptor signalling. Leads to extensive remodelling of plasma membrane. Involved in many processes - immune system, tumor progression.

Can be used for cell motility by taking up receptors from cell surface and moving them to the leading edge.

How do pathogens modulate macropinocytosis?

- induction of cell signalling (act on receptor)
- mimic apoptotic material
- directed actin remodelling
- secreted factors

What are endosomes?

A sorting site which sorts proteins away from the lysosome, and a major site of protein degradation. Endosomes become acidified via a proton pump (lowers pH)


Explain the process of transferrin uptake into a cell

Ferrotransferrin (with Fe3+) binds to receptor and stimulates clathrin coated endocytosis. Scission and uncoating of vesicle in cytoplasm. Fusion of uncoated vesicle with endosome. Low pH causes release of Fe3+ from ligand but NOT THE RELEASE OF THE LIGAND. Receptor + ligand is recycled back to cell surface. Low pH causes ligand release from the receptor (when there is no Fe3+ attached - apotransferrin)

What happens to membrane proteins and intraluminal proteins in the late endosome/lysosome?

Membrane proteins tend to survive whereas material within the intraluminal vesicles is degraded

Explain the basic process of EGF receptor

An example of receptor/ligand degradation

EGF binds to the EGF receptor which dimerises, stimulating phosphorylation of the receptor tail. This stimualtes internalisation and ubiquitination of the receptor/ligand. Internalised EGFR retained in the endosome and taken into MVBs. After this point, both the receptor and ligand are degraded in the lysosome. Some receptor can be rescued and recycled back to the cell surface (depends on the signals on the tail).

How can the endosome act as a signalling platform?

Signalling can occur from a receptor + ligand on the cell surface. Signalling can also occur from an internalised receptor + ligand in the endosome. Differential outcomes of signalling cascades due to different initiation points (different signalling cascades from the same endosome/different signalling from early vs late endosome).

What are multivesicular bodies?

Specialised endosomes that transit from early (sorting) endosome to late endosome/lysosome.
Internalised vesicles are sorted- some are taken in and some are secreted. MVBs mature into lysosomes and their content is degraded.

Explain the process of the internalisation of a vesicle into a MVB.

Ubiquitination of HRS proteins and cargo proteins on the endosome surface stimulates the formation of a bud. As bud grows, ESCRT proteins bind to ubiquitinated HRS proteins which stimulates the internalisation of the vesicle. ESCRT proteins are still bound to the Ub HRS proteins, and Vsp4 is required to use ATP to separate them.

Explain how HIV uses the MVB pathway to exit cells

HIV gag protein on cell surface is ubiquitinated and bud containing HIV core particle is formed. ESCRT proteins then bind to the Ub Gag, stimualting the release of the vesicle (an enveloped HIV virus). Vsp4 then uses ATP to disassemble the ESCRT proteins

What is autophagy?

The recycling of worn out organelles and degradation of specific cargo. Used in response to nutrient starvation, infection or apoptosis

Which would appear more dense during electron microscopy - an endosome or a lysosome?

A lysosome - full of enzymes and degradative material

How do lysosomes lower their pH?

A proton pump

How is the cell protected from the degradative enzymes it synthesises? (Why do they only work in the lysosome?)

Degradative enzymes may only work at the low pH of the lysosome and/or may be synthesised as precursors which require cleavage before they become active

How are nutrients retrieved from the lysosome to the cytoplasm?

Nutrient pump

What are some intracellular tracking pathways that involve recycling back to the plasma membrane?

LDL
Transferrin

Give an example of a receptor pathway that requires retrograde transport?

M6P pathway - recovered from late endosome to the golgi

Give an example of a receptor pathway in which both the ligand and the receptor are degraded

EGF pathway

How is the maturation of the endosome defined? (i.e what differentiates an early from a late endosome?)

As endosomes mature, the outside components change- maturation is defined by presence of specific Rab molecules and phosphoinositide content


This allows different trafficing machinery to be recruited and different cargo to be sorted

How can early/late endosomes have different functions?

The different Rab/PI content allows different trafficking machinery to be recruited and different cargo to be sorted

What are the different Rab effector functions?

Sorting = Rab can activate a sorting adaptor to sort a receptor into a budding vesicle
Uncoating = recruitment of PI kinases or phosphatases can cause change in the PI content of vesicles leading to uncoating (if coat proteins bind PI)
Transport = Rabs can recruit microtubule motor adaptor proteins or bind the motors themselves
Tethering = Rab can recruit tethering factors that interact with acceptor membrane to cause fusion

What is myosin V

Actin associated motor that moves vesicles around

What are Lewy Bodies?

Intracellular protein aggregates found in parkinson's disease- still debated whether these are a cause of the disease or a symptom

What is alpha synuclein?

The most abundant protein found in lewy bodies, and is directly linked to the activation of apoptotic pathways

How is immunofluorescence microscopy prepared?

Sample prepared on slide
Incubate with primary antibody
Wash away unbound antibody
Incubate with fluorochrome conjugated secondary antibody
Wash away unbound antibody
Mount and observe

How are monoclonal antibodies made?

Mouse injected with antigen X
Harvest spleen cells (some make Ab against Ag X)
Spleen cells fused with mutant mouse myeloma which is unable to grow on selective medium
Cells transferred to selective medium (unfused cells die)
Single cells cultured in separate wells
Each well is tested as an Ab to Ag X

What is the Stokes shift?

The difference between the peak absorption and peak emission of the fluorescent protein used in IF microscopy.

What is confocal microscopy?

Relies on elimination of out-of-focus light to create optimal sections
Pinhole in front of camera only allows light from a focus plane to enter the detector

What is two photon microscopy?

Used for imaging live tissue
Uses near infrared light to excite dyes, because of this, two photons are absorbed.
Due to the multiphoton absorption, the background signalling is strongly reduced, also allows deeper tissue penetration and more efficient light detection

What is electron microscopy?

A higher resolution/magnification microcopy technique than light microscopy. Samples are always processed. Live imaging is possible however processing artifacts can occur. Samples are stained with heavy metal prior to being imaged.

Why is yeast a good model organism?

- unicellular but a model of multicellular organisms
- easy/quick to grow
- contains homologs of most mammalian genes
- easy to knock out genes
- completely sequenced genome
- similar cytoskeletal organisation and signalling pathways to human cells

What is VRP1?

The yeast homolog of human WASp

What is the phenotype of a VRP1 mutant?

Changes in morphology, proliferation rate and temperature sensitivity

Explain the basic process of PCR

Repeated heating cycles to make multiple copies of a target section of DNA

1. 94'C = denaturation temperature
2. 55'C = annealing temperature for primers
3. 72'C = elongation using Taq polymerase

What happens if the annealing temperature is too high/too low?

Too high = primer may not bind
Too low = primer might bind imperfectly or form primer dimers