Sie sind auf Seite 1von 25

Chemical communication

• understand how protein modifications play a key role in determining


biological responses;

• understand the concept of receptors and their specificity for different


ligands;

• understand the principal elements of the major signal transduction


cascades;

• describe the roles of the major protein kinase classes and other second
messengers in signal transduction;

• describe the basic signal transduction schemes for G-protein coupled


receptors, growth factor receptors (tyrosine kinase receptors), cytokine
receptors, steroid (nuclear) receptors and integrins;

• relate at a molecular level how the selected chemical mediators produce


their major physiological/regulatory roles;

• understand the principles of endocrine regulation and the key histological


features of selected endocrine tissues.

How do cells communicate? • Extracellular signalling molecules bind


to specific receptors
• When bound to a ligand, receptors
initiate a response within the cell. They
may be on the cell surface or
intracellular
• Intracellular signalling proteins
activate a signalling pathway leading
to interaction with a target protein and
a biological response
• Removal of a signal leads to removal
of a response. The time between the
two is dependent on the rate of
destruction of the signalling molecule
and the rate at which the changes it
has caused can be reversed
How does communication • A typical cell is exposed to hundreds of
vary between cells? different signals in its environment.
Different cells respond to a
combination of signals from their
environment according to their own
specific character to regulate their
behaviour. Different combinations of
signals lead to different responses
• Different cells can respond differently
to the same signalling molecules, for
example by having different receptors
on their plasma membrane
What are the types of • Contact dependent – membrane bound
signalling? signal molecules interact with
receptors on the membranes of
adjacent cells
• Paracrine – signalling molecules are
released into extracellular space and
act locally on neighbouring cells – local
mediators
• Synaptic signalling – neurons transmit
signals electrically along their axons
and release neurotransmitters across
synapses
• Endocrine signalling – endocrine cells
secrete hormones into the
bloodstream which are distributed
throughout the body and bind with
target cells
• Autocrine signalling – cells responding
to a signalling molecule which they
have secreted themselves
What are the 4 types of • Type 1 – ligand-gated ion
receptors? channels/ionotropic receptors,
transmembrane, direct coupling to
intracellular effector systems, typically
the receptors on which
neurotransmitters act, e.g nicotinic
ACh receptor, GABAA receptor
(inhibitory effect on
neurotransmission)
• Type 2 – G-protein coupled receptors,
transmembrane, most abundant,
coupled to intracellular effector
systems by G-proteins, largest group
of receptors, e.g. muscarinic ACh R,
GNRH R (causes release of FSH and LH,
phosopholipase C second messenger),
MC2 R (stimulates cortisol production,
cAMP second messenger)
• Type 3 – kinase-linked receptors,
transmembrane, direct coupling to
intracellular effector systems via
intracellular enzymatic domain, e.g.
insulin receptor
• Type 4 – nuclear receptors, located in
cytosol/nucleus, regulate gene
transcription, coupled to intracellular
effector systems by DNA, e.g. steroid
receptors

What is the structure and • Structurally similar to other ion


function of type 1/ionotropic channels
receptors? • Involves the fastest synaptic
transmission – ligand binding causes
shape changes and channel opening
occurs over milliseconds
• Extracellular ligand binding domain
• 4 or 5 transmembrane alpha helices
• Intracellular domain may have a role
in cell regulating the channel
What is the structure and • Seven transmembrane alpha helices
function of type 2/G-protein • Extracellular ligand binding domain,
coupled/metabotropic large intracellular binding domain for
receptors? G-proteins (GNRH-R has no
intracellular C terminal tail domain –
slower internalisation. Though to be
the product of a mutation causing a
premature stop codon – C terminus is
still present in non-mammalian
species)
• When activated, receptor undergoes
shape change which allows larger
intracellular loop to bind to and
activate G proteins, which go on to
initiate a biological response
• Some receptors couple to multiple G
proteins, e.g. PAC1 R
What is the structure and • Trimeric GTP-binding proteins
function of G proteins? consisting of alpha beta and gamma
subunits – at least 15 different alpha,
12 different gamma and 6 different
beta subunits (many different G
proteins allows for diversity in
biological responses)
• In resting state, alpha subunit is
occupied by GDP. When associated
with the receptor, the bound GDP is
replaced with GTP and the alpha unit
is dissociated from the beta-gamma
dimer
• Alpha/ beta-gamma subunits goes on
to activate target proteins –
membrane bound enzymes or ion
channels
• G alpha s subunit stimulates adenylyl
cyclase
• G alpha i subunit inhibits adenylyl
cyclase
• G alpha q/11 subunit stimulates
phosphoinositide system
• G beta-gamma dimer interacts with
GIRK (G-protein activated inwardly-
rectifying potassium channels) to
increased K permeability
• G protein subunits may also activate
MAPK pathway (mitogen-activated
protein kinase)
• When the alpha subunit binds to its
target, its GTPase activity increases.
Hydrolysis of the bound GTP to GDP
causes the alpha subunit to reunite
with the beta-gamma dimer and
ceasing stimulation of target proteins
What is the adenylyl cyclase • Activation of receptor causes a G
system? alpha s subunit of a G-protein
complex to become dissociated from
the beta-gamma dimer
• G alpha s binds to and activates
adenylyl cyclase, which can then
catalyse the conversion of ATP to
cAMP. G alpha i subunit has an
inhibitory effect on adenylyl cyclase
activity
• Increased levels of cAMP within the
cells may lead to the activation of
protein kinase A and cyclic nucleotide
gated ion channels being opened
• The G alpha s subunit eventually
catalyses the hydrolysis of GTP to
GDP, inactivating it and shutting off
the cAMP dependent pathway, and
cAMP phosphodiesterase
dephosphorylates cAMP to AMP
• Guanylyl cyclase is similar – except
not G-protein coupled, activates
cGMP which in turn activates protein
kinase G
What is the phosphoinositide • Activation of receptor (e.g. by GnRH)
system? causes dissociation of G alpha q/11
subunit, causing activation of the
membrane-bound enzyme
phosopholipase C, which hydrolyses
PIP2 to diacyl glycerol and inositol
triphosphate
• IP3 moves into the cytosol and
activates calcium channels in the
endoplasmic reticulum, increasing
intracellular calcium which has a role
in further signal transduction
pathways
• Increase in calcium levels is biphasic
– spike phase occurs when calcium is
mobilised from the endoplasmic
reticulum, plateau phase occurs due
to calcium influx from outside the cell
via VG and capacitative calcium
channels
• Activation of capacitative channels is
thought to be due to STIM1 sensing
the depleted calcium levels in the
endoplasmic reticulum and relocating
to the plasma membrane to activate
the ORAI1 calcium channel
• Calmodulin can bind to intracellular
calcium, and then bind to other
targets such as CaM kinase to
generate a response
• DAG remains within plasma
membrane due to hydrophobic
properties, and activates protein
kinase C in combination with the
increased calcium by causing it to
migrate to the plasma membrane
(RACK proteins)
• Protein kinase C phosphorylates
target proteins causing further
downstream effects which differ
dependent on the tissue involved
What is the MAP kinase • May be activated by activation of
cascade? receptor tyrosine kinases, by G
proteins or protein kinase C
• Phosphorylation cascade which
targets serine and threonine residues
in target proteins
• A stimulus causes activation of
receptor tyrosine kinase, which
activates the SH2 domain protein
Grb, which activates Ras, which
activates MAP kinase kinase kinase
(Raf) which activates MAP kinase
kinase (Mek), which activates MAP
kinase, which can phosphorylate
target proteins such as transcription
factors and other kinases to cause a
biological response
• MAP kinases are deactivated by
dephosphorylation (protein
phosphatases)
What is the structure and • Extracellular ligand binding domain
function of type 3/kinase- which varies in structure between
linked receptors? different receptors connected by a
single alpha-helix to intracellular
catalytic domain
• Tyrosine kinase is incorporated into
the catalytic domain for the receptors
for various hormones/growth factors
• Cytokine receptors have intracellular
domains which bind to and activate
cytosolic tyrosine kinases (e.g.
Jak/Stat pathway)
• Agonist binding causes dimerisation
of receptors and autophosphorylation
(each receptor phosphorylates
tyrosine residues on the other). SH2
domain proteins bind to the
phosphorylated tyrosine residues and
are activated
• Different SH2 domain proteins bind to
specific receptors and cause different
biological responses, e.g. activation of
Ras leading to activation of MAPK
cascade
What is the Jak/Stat pathway? • JAKs become bound to activated
cytokine receptors and are
themselves activated
• They phosphorylate the tyrosine
residues on the receptors
• STATs are transcription factors which
possess SH2 domains allowing them
to bind to the phosphorylated JAK-
receptor complex, where they are
phosphorylated by JAK
• The STATs dimerise and are
translocated to the nucleus where
they regulate expression of STAT
responsive genes
• The pathway can be deactivated by
the action of protein phosphatases,
inhibitors of JAKs, factors which
compete with STATs for
phosphotyrosine binding sites and
proteins which inhibit transcriptional
activation, such as PIAS1 and PIAS3
which bind to and block STAT1 and
STAT3 target genes respectively
What is the structure and • Similarly to receptor tyrosine kinases,
function of receptor guanylyl receptor guanylyl cyclases are found
cyclases? on the plasma membrane have an
extracellular ligand binding domain
and an intracellular catalytic domain
linked by a single transmembrane
domain
• Soluble guanylyl cyclase is also found
in the cytosol and is activated by
nitric oxide
• When activated, the guanylyl cyclase
domain catalyses the conversion of
GTP to cGMP
• cGMP activates protein kinase G,
which phosphorylates target proteins
to cause biological responses
• Breakdown of cGMP by
phosphodiesterases ceases activation
of PKG
What is the structure and • Ligand activated transcription factors
function of type 4/nuclear found in the cytoplasm/nucleus
receptors? • They respond to hydrophobic signal
molecules such as steroid/thyroid
hormones which are able to diffuse
across plasma membrane of target
cell
• Some steroid receptors can also be
expressed on the cell membrane and
are G protein linked, and steroids can
also interact with G-protein coupled
receptors such as GABA-R
• Ligand binding causes a
conformational change in the steroid
receptor, and the dissociation of
inhibitory proteins (e.g. Heat Shock
Proteins), dimerisation of the
receptor, and the translocation of the
receptor to the nucleus, where it
binds to hormone response elements
(HREs) on the DNA
• The DNA-receptor complex can then
recruit coactivator proteins and RNA
polymerase to cause the expression
of genes which cause a biological
response
• The steroid receptor itself consists of
distinct domains. The A/B domain is
variable between different receptors
and containing the transcription
activation function AF-1. The C
domain is the DNA binding domain
and contains two ‘zinc fingers’. The D
domain is a hinge region which allows
conformational changes to occur. The
E/F domains are involved in ligand
binding, dimerisation, binding of
coactivators/corepressors and
contains the transcription activation
function AF-2

How are receptors • Sustained exposure caused


desensitised? desensitisation in all majors classes of
receptors
• Homologous – resulting from
continued stimulus from ligand acting
on receptor, e.g. causes
phosphorylation of receptor which
can then bind to arrestin
• Heterologous – resulting from
continued stimulus from another
effector, e.g. activation of receptor
activates protein kinases which can
phosphorylate the receptor
• Receptors can be inactivated by
undergoing conformational change
(phosphorylation), undergoing
endocytosis and being stored in an
endosomes/broken down by
lysosomes or by proteins which inhibit
the receptor itself or the signalling
proteins it activates.
What is cholesterol? • Precursor molecule for steroid
biosynthetic pathway
• Cholesterol may be obtained from
dietary sources or synthesised within
the body from acetyl CoA
(mevalonate pathway, DMAPP 
lanosterol  cholesterol). This
synthesis occurs mainly in the liver
• Cholesterol is converted into steroid
hormones by steroidogenic endocrine
organs
What is steroidogenesis? • Biological process by which steroids
are generated from cholesterol via
biosynthetic pathways
• Cholesterol passes readily across
plasma membrane due to lipid
nature, and is stored in vesicles in the
cytoplasm
• Rate of steroidogenesis is limited by
the rate at which cholesterol can be
transported from the cytoplasm into
mitochondria by StAR (steroidogenic
acute regulatory protein)
• Synthesis of a specific steroid is
achieved by the appropriate
sequence of enzymes being
expressed in the cells of a tissue
• Steroidogenic factor 1 is expressed in
steroidogenic tissues and regulates
the transcription of steroidogenic
enzymes by binding to the Ad4 site
found in the promoter region of
steroidogenic P450 (CYP) genes.

How do steroid hormones • Steroids are bound to serum proteins


have their effects? such as serum albumin when
circulating in the blood. This prevents
them from moving across the plasma
membrane of cells where their action
is not required
• As they are lipid soluble, when
dissociated from serum proteins they
are able to pass through cell
membranes to bind with steroid
receptors found in the cytoplasm or
nucleus
• These receptors then act as
transcription factors in the nucleus
What is the role of chaperone • Some steroid receptors, for example
proteins in steroid receptor glucocorticoid receptors are associated
regulation? with chaperone proteins (e.g. Heat
Shock Proteins)
• These proteins maintain the receptors
in a transcriptionally inactive state in
the absence of their steroid ligands
• This may be due to the chaperone-
receptor complex being too large to
cross the nuclear membrane, or the
chaperone moving the receptor away
from the nucleus
• These proteins become dissociated
from the receptor when it is bound to a
steroid

How is oestrogen synthesised? • Oestrogen is an example of a


steroid hormone
• The precursors for oestrogens are
the androgens androstenedione
and testosterone. In the ovary
these are synthesised in the thecal
cells before being transported into
granulosa cells
• The gene CYP 19 encodes for the
enzyme aromatase P450. It is
expressed in gonads, adipose
tissue and by osteoblasts and
found in the endoplasmic
reticulum
• Aromatase catalyses the formation
of the oestrogens oestrone and
oestrodiol from androstenedione
and testosterone respectively – an
aromatisation reaction
What is the role of oestrogen • Oestrogen precursors are
during the menstrual cycle? produced in the thecal cells,
before being transported into
granulosa cells and being
aromatised to form oestrogen
• Oestrogen levels peak just before
ovulation, stimulating production
of GnRH in the hypothalamus,
which in turn triggers the release
of LH from the anterior pituitary
leading to ovulation – an example
of positive feedback

What is the structure of • There are 2 forms – ER alpha and


oestrogen receptors? ER beta
• They are expressed in many
tissues across the body and bind
oestrogen with high affinity
• Both are nuclear receptors and
ligand-activated transcription
factors, but they have different
genetic origins
• ER alpha consists of 595 amino
acids, ER beta consists of 530
amino acids, and there is little
sequence similarity between the
two in certain regions
• When the receptors dimerise they
may for homodimers or
heterodimers
• Some oestrogen receptors may be
bound to plasma membrane –
associated with G proteins
How do oestrogen receptors • When oestrogen has bound to ER
respond when activated? in the cytoplasm/nucleus, the
oestrogen-ER complex is
translocated into the nucleus
• The ER then dimerises and binds
to oestrogen response elements
(ERE sites) found in target gene
promoter regions and acts as a
transcription factor. Gene
expression leads to a biological
response
• ERs may also interact with other
transcription factors to regulate
gene expression
• Oestrogen has different effects in
different tissues dependant on the
type of receptors presence and
whether they have additional
interactions – biologically
specificity

How is oestrogen receptor • Regulation of expression of ER


function regulated? genes – transcription and
translation
• Translocation of receptors to areas
where they are inactive
• Phosphorylation of receptor to
cause shape changes
• SUMOylation - Small Ubiquitin-like
Modifier proteins binding to
receptor may inhibit its function

What are the biological roles of • Sexual differentiation


oestrogen? • Folliculogenesis, spermatogenesis
and gonadotrophin production
• Lordosis behaviour
• Maintenance of brain function
• Effects in bone
• Cardiovascular effects
• Counteracts symptoms of
menopause – hormone
replacement therapy
• Reduces incidence of some
tumours (colon cancer) but
thought to be involved in incidence
of breast cancer
What are the roles of oestrogen • Inhibits remodelling and promotes
in bone? formation
• Blocks osteoblast synthesis of
interleukin 6 which stimulates
production of osteoclasts
• Stimulates TGF beta production
leading to osteoclast apoptosis
What are the effects of • May regulate nitric oxide levels,
oestrogen on the cardiovascular which in turn regulates cGMP
system? levels, which causes relaxation of
muscle surrounding blood vessels
– vasodilation and increased blood
flow
• Has been shown to increase
coronary flow and cardiac output
in sheep
• Reduces low density lipoprotein,
apolipoprotein, platelet and
chylomicron remnant levels in the
blood – decreased risk of
atherosclerosis
• IGF and VEGF levels increased –
involved in formation of blood
vessels
How is oestrogen implicated in • Aromatase is expressed in breast
breast cancer? tissue - adipocytes
• This expression is usually low
level, but tumours express it at
higher levels (use of different
promoter)
• This is an example of positive
feedback – aromatase causes
production of oestrogen, which can
in turn enhance aromatase
expression
• Inhibition of aromatase
expression/estrogen receptors
could aid treatment, but if not
specifically targeted would also
have detrimental effects on other
areas of the body
What is the TGF-beta • A group of around 40 structurally
superfamily? related cell regulatory proteins
which is named after its first
member – TGF beta (transforming
growth factor beta) and includes
bone morphogenetic proteins,
growth differentiation factors (such
as myostatin), activin and inhibin
• These proteins are active only as
dimers, and interact with type I/II
receptor serine/threonine kinases
What are some examples of • TGF beta – controls proliferation
members of this family and their (causes synthesis of p15 and p21
functions? which prevent phosphorylation of
Rb and therefore progression past
G1 in cell cycle), differentiation (in
pre-adipocytes, myoblasts and
osteogenic precursors), can induce
apoptosis, regulates adhesion
molecules/immune
system/inflammatory responses
(regulation of B and T
lymphocytes, macrophages,
natural killer cells and cytokine
production), mutations can lead to
development of cancer, elevated
levels are involved in pathogenesis
of Marfan syndrome
• Activin – role in development,
ovarian and testicular function,
wound repair and immune
responses
• Inhibin – inhibition of activin
action, binds to activin receptors
• Myostatin – inhibits muscle
differentiation and growth,
mutations to gene cause excessive
muscle growth (e.g. mighty mice)
• Bone morphogenetic proteins –
involved in
development/differentiation etc.
Mutant genotypes are uncommon
as lethal in utero in many cases
How are these ligands • They are expressed as inactive
synthesised? dimeric pre-pro proteins with a
disulphide bond between each side
of the protein, and required post
translational modification to
become active
• The pre or signal sequence is
required for the protein to be
secreted from the cell, it is cleaved
from the protein once it binds to
the endoplasmic reticulum and
enters the secretory pathway
• Subtilisin like pro-protein
convertase (SPC) cleaves the pro
sequence from the protein, leaving
the mature ligand dimer
• The protein may be secreted with
the pro sequence still bound to it
How does TGF beta signalling • Type I and type II receptor
occur? serine/threonine kinases - each
member of superfamily binds to a
characteristic combination of type
I and II receptors
• Typically, the ligand binds to and
activates a type II receptor dimer
which then phosphorylates a type I
receptor dimer causing
dissociation of an inhibitory protein
forming an active tetrameric
receptor complex
• The type I receptor binds to and
phosphorylates a SMAD protein –
TGFb and activin receptors
phosphorylate Smad 2/3, BMP
receptors phosphorylate
Smad1/5/8
• Phosphorylated SMAD dissociates
and binds to Smad 4, and moves
into the nucleus to act as a
transcription factor
What factors regulate TGF beta • Inhibitory SMADS such as Smad
signalling at the receptor level? 6/7 bind to activated type I
receptors to prevent other SMADs
binding. Smad 7 can be produced
by activation of the Jak-Stat
pathway
• BAMBI (BMP and activin membrane
bound inhibitor) - transmembrane
protein similar in structure to type
I receptors, lacking a kinase
domain. It also interacts directly
with BMP receptors to inhibit their
activity
• Betaglycan (TGFb receptor type III)
– membrane-associated protein,
co-receptor for TGFb and inhibin
which presents ligand directly to
kinase domain of type II receptor.
It enhances binding of inhibin to
activin receptors and therefore
inhibit the action of activin, and
has been shown to confer inhibin
sensitivity to normally poorly
responsive cell lines
• DRAGON -
glycophosphatidylinosital-
anchored membrane protein which
acts as a co-receptor by binding to
BMP 2/4 and associating with type
I and II BMP receptors to
potentiate BMP signalling
• Cripto – gpi anchored membrane
protein which facilitates Nodal (a
ligand) signalling, inhibits activin B
and overexpressed in many
human solid tumours (e.g. breast
cancer)
• Endoglin – accessory receptor for
TGF beta, aids binding to ALK1
receptor and is essential for
angiogenesis (endoglin deficient
mice die during gestation due to
cardiovascular defects)
What factors regulate TGF beta • Noggin/chordin/follistatin inhibit
ligands? BMPs
• Follistatin and follistatin-like
hormone 3 inhibit activins
• Cerberus related cytokines such as
Cerberus, DAN and Gremlin are
thought to bind to BMPs and
prevent them from binding to their
receptors
• Cerberus also antagonises Nodal-
like and Activin-like ligands
• These ligand inhibitors may be
released by cells on which the
factors they inhibit act, this is an
example of negative feedback

What is angiogenesis? • Formation of new capillaries from


existing blood vessels which is
regulated by various growth
factors and cytokines, there are
generally more inhibitory factors
• Important in both normal and
pathological physiology, occurs
during development, healing, the
monthly reproductive cycle in
females (rebuilding lining of
uterus) and during pregnancy
(formation of placenta)
• Distinct from vasculogenesis, the
formation of blood vessels when
there are no pre-existing ones
What is the mechanism of • Diseased/injured tissues produce
angiogenesis? angiogenic growth factors which
diffuse into nearby tissues and
bind to specific receptors on
endothelial cells of pre-existing
blood vessels
• Causes expression of proteases
from the cells (causing
degradation of the basement
membrane) and their proliferation
• The cells migrate through the
degraded basement membrane
towards the source of the agf
using adhesion molecules
expressed on the plasma
membrane called integrins.
Integrins are receptors which
mediate the cells attachments to
surrounding tissue
• Matrix metalloproteinases degrade
the extracellular matrix to
accommodate the migrating cells,
which ‘roll up’ forming a tube as
they continue to proliferate
• These tubes link up to form loops
which are stabilised by pericytes
(vascular smooth muscle cells),
and circulation can begin
What is vascular endothelial • A glycoprotein which forms
growth factor (VEGF) and its homodimers made active by
roles? proteolytic cleavage
• Several isoforms exist – different
number of amino acids and
isoelectric properties (more
acidic/basic – most active at
differing pHs) but all product of
same gene
• Stimulates angiogenesis and
vasculogenesis (mice
heterozygous for VEGF die in
utero)
• Regulates bone formation and
growth – vascularisation required
during development
• Prevents hepatic cells undergoing
apoptosis and stimulates their
proliferation
• Regulates glomerular function
(lack of VEGF causes renal disease
in mice)
• Regulates follicular/corpus luteum
development
• Expressed in pancreatic islets and
aids blood glucose regulation
• Secreted by many solid tumours,
increasing the blood supply
needed for growth
• Stimulates growth of
leukaemic/lymphoma cells –
VEGFR1 present on leukaemic cells
• Low VEGF in spinal cord neurons
leads to sclerosis – implicated in
Alzheimers etc.
• Role in intraocular neovascular
syndrome, inflammatory disorders
(e.g. rheumatoid arthritis),
endometriosis and polycystic OS
How is VEGF regulated? • Hypoxic cells/ischaemic tissues
produce the transcription factor
hypoxia-inducible factor (HIF1)
which causes expression of VEGF
• Cytokines, growth factors and cell
differentiation may stimulate VEGF
• Tumours produce proteases, which
cleave inactive VEGF to make it
active – leading to more
angiogenesis and tumour growth
• Oncogenes such as ras and rat
upregulate VEGF
How does VEGF exert its effect? • Interacts with receptor tyrosine
kinases (VEGFR1/2), co-receptors
and neuropilins (a receptor on
neuronal cells)
• Different SH2 proteins can bind to
the autophosphorylated tyrosine
kinases to activate different
pathways and therefore cause
different biological outcomes
• Different factors can also regulate
which responses occur – outcome
of VEGF signalling is dependent on
the tissue in which it occurs
• Pathways activated include MAPK
and phosopholipase C
How can anti-VEGF/VEGF • Anti-VEGF monoclonal antibodies
stimulating factors be used in bind to it and prevent it interacting
medicine? with receptors, helping to reduce
tumour spreading and
endometriosis
• Anti VEGF receptor antibodies can
have a similar effect by preventing
ligand binding
• Anti-VEGF treatments can cause
thrombosis, increased blood
pressure and polyuria
• VEGF treatment can help treat
neurological disorders such as
Huntingtons and Parkinsons by
aiding recovery of neurons – but
may lead to tumour growth

What makes up a tissue? • Cells and cell-cell cell-matrix


junctions
• Basal lamina – extracellular matrix
between connective tissue and
epithelia
• Extracellular matrix – network of
secreted macromolecules to
maintain integrity
How can connective tissues and • Majority of tissue volume in
epithelial cell sheets be connective tissue is extracellular
compared and contrasted? matrix, majority of tissue volume
in epithelia is cells
• Matrix in connective tissue
consists of many fibrous proteins,
matrix in epithelia is a thin lamina
• Matrix bears most of mechanical
stress in connective tissue, cell
cytoskeleton has this role in
epithelia
• Few cell-cell attachments in
connective tissue, cells are tightly
bound in sheets in epithelia
What is the structure and • Network of locally secreted
function of extracellular matrix? macromolecules which provides a
‘scaffold’ for cells and influences
their activity
• Cells determine the ECM produced,
its turnover and its organisation,
whilst the ECM can influence cell
shape, migration, proliferation and
function as well as creating
structure (e.g. stem cells
differentiate differently depending
on the mechanical properties of
their local ECM)
• Different constituents give the
different connective tissues their
physical properties
• The two main components of ECM
are glycosaminoglycans and
fibrous proteins
What is the structure and • Composed of repeated
function of glycosaminoglycans? disaccharides, which are generally
unbranched
• Negatively charged and
hydrophilic, making them able to
draw water into the ECM giving it a
huge volume relative to mass to
help resist compressive forces
• Consist of a uronic acid linked to
Chondroitin sulphate an amino sugar which may be
sulphated – different combinations
produce different
glycosaminoglycans and therefore
different proteoglycans
• The most prevalent GAG is
chondroitin sulphate
• Hyaluronan is a large atypical GAG
Hyaluronan
which is the most ‘simple’,
consisting of identical unsulphated
disaccharide units. It links ionically
to proteins and is synthesised by a
cell surface enzyme. It functions as
a ‘space-filler’ to create spaces for
cells to migrate into and as a
lubricant
What is the structure and • Protein cores are synthesised in
function of proteoglycans? the rough endoplasmic reticulum,
before being post-translationally
modified by glycosyltransferases
in the Golgi apparatus. GAGs are
covalently linked to the core
proteins to produce proteoglycans
• They can produce highly organised
structures by associating with
other proteoglycans, with fibrous
proteins such as collagen, or with
hyaluronan, which can bind
ionically to the globular domain of
the protein core
• Aggrecan is a proteoglycan which
forms a major structural
component of cartilage
• They are involved in regulation of
the movement of substances in
the ECM
• They bind and regulate the
activities of some signalling
molecules and enzymes, for
example TGFb can bind to decorin
inhibiting TGFb from stimulating
cells to synthesise ECM, FGF can
only increase fibroblast
proliferation when bound to
syndecan and proteoglycans can
modulate the activity of proteolytic
enzymes (e.g. matrix
metalloproteases) or their
inhibitors (e.g. tissue inhibitor of
metalloproteinases) by binding to
them
• Cell surface proteoglycans act as
co-receptors to facilitate effective
signalling
What fibrous proteins are • Collagen is the most abundant
present in ECM and what is their protein in the ECM. It has high
function? tensile strength and consists of
many amino acid chains arranged
in alpha-helices. The amino acids
are arranged as triplets, the most
common arrangement consists of
glycine, proline and
hydroxyproline.
• There are 3 groups of collagen
types: fibrillar (types 1-3, 5 and
11), network-forming (4 and 7) and
fibril-associated (9 and 12).
Fibrillar is rope like and is found in
connective tissue. Network
forming forms the meshwork
sheets of basal lamina and links
epithelia to it. Fibril-associated are
attached to collagen fibrils and
influence their arrangement by
forming linkages
• Laminins are prevalent in the basal
lamina. They form networks with
type 4 collagen, perlecan and
entactin
• Elastin fibres give elasticity to the
tissue
• Fibronectins connects cells to
collagen allowing cell adhesion
and movement
What is the structure and • Specific form of ECM found at
function of the basal lamina? junctions between connective
tissues and other tissues
• Synthesised by the epithelial cells
for which it forms a continuous
flexible base
• Made up of type 4 collagen, the
proteoglycan perlecan, the
glycoprotein entactin and protein
laminins, which associate together
and (all but entactin) self associate
giving the basal lamina its tensile
strength
• It forms a barrier between cell
layers, preventing migration of
cells
• It acts as a scaffold for
regeneration following injury along
which cells can migrate
• Due to its GAG component, it can
act as a selective molecular filter
(such as in glomerulus of kidney)
• It determines cell polarity – cells
each side of the basal lamina have
a specific character
What are the 3 classes of cell • Tight (occluding) junctions seal
junction? cells into a sheet, they encircle
epithelial cells and regulate their
permeability
• Anchoring junctions attach cells to
other cells or to ECM. They may be
actin-dependent (cell-cell
adherens junctions or cell-matrix
adherens junctions) or
intermediate filament dependent
(desmosomes between cells, hemi-
desmosomes between cells and
matrix)
• In cell-cell adherens junctions,
intracellular actin is bound to
intracellular attachment proteins
such as catenins, which are linked
to attachment proteins of adjacent
cells by transmembrane linker
proteins such as cadherins
• In cell-matrix adherens junctions,
intracellular actin filaments
terminate at focal adhesions,
intracellular attachment proteins
such as talin link actin to
transmembrane linker proteins
such as integrins which interact
with ECM
• Gap junctions allow chemical or
electrical signals in the form of
water soluble molecules and
inorganic ions to pass between
communicating cells via channels
known as connexons. They consist
of 6 connexin sub-units, the
rotation of which dictates the size
of the channel pore

Das könnte Ihnen auch gefallen