Description
BSC 300-001 Week 12 Notes
I. Chapter 20 – Integrating Cells into Tissues
a. Cell-Cell and Cell-Extracellular Matrix Adhesion: An Overview
i. General Concepts about Cell-Cell and Cell-Extracellular Matrix Adhesion 1. Cell-cell and cell-extracellular matrix interactions are critical for
assembling cells into tissues, controlling cell shape and function,
and determining developmental fate of cells and tissues
a. Dynamic – nothing is really permanent
2. Cell-adhesion molecules mediate direct cell-cell adhesions, and
adhesion receptor proteins mediate cell-matrix adhesions
3. The extracellular matrix is a dynamic, complex meshwork of
proteins and polysaccharides that contributes to the structure and
If you want to learn more check out What does the mulvery describes?
function of tissues
a. As cells interact with it, they change and modify it and vice
versa
ii. Major cell-cell and cell-matrix adhesive interactions
1. In multicellular animals cell-cell and cell-matrix adhesions
aggregate cells into distinct tissues in order to cooperatively
perform common functions
2. Humans have hundreds of unique cell types while simpler
metazoans, like the round worm C. elegans have only 12
3. Despite these differences all animal cells can be classified into 5 If you want to learn more check out What is the capital lease?
major tissue types:
a. Muscle
b. Neural
c. Blood
d. Epithelial – sheets of tightly connected cells composed of
one of more layers that cover or line the major body and
organ surfaces
e. Connective – found in between other tissues throughout
the body. They support and protect other tissues and
collagenous fibers, autonomous, non-connected cells and
ground substance: the gel-like material secreted by cells in
which they and the fibers are embedded. Collectively, the
ground substance and fibers are referred to as the
extracellular matrix
4. Cells adhere to one another and associate with the extracellular
environment through dynamic spatial and temporal expression and
functional regulation of diverse adhesion molecules
a. Cell-adhesion molecules (CAMs) generate cell to cell
interactions. Cytosolic domains bind diverse intracellular
adapter proteins that link them to the cytoskeleton. Don't forget about the age old question of What type of diabetes is more common?
Don't forget about the age old question of Where did psychology originate from?
Extracellular domains associate with other CAM proteins
b. Adhesion receptors link cells to the extracellular matrix.
Similarly are cytoplasmically linked to the cytoskeleton
i. Can transduce info to the interior of the cell
BSC 300-001 Week 12 Notes
c. Both types of adhesion link to the cytoskeleton and
participate in intracellular signaling pathways
i. In doing so, through these associations cells can
transduce signals and respond to environmental
cues and be influenced to change their shape and
behavior
ii. Likewise, cells can communicate to the
extracellular environment and alter activity and If you want to learn more check out What does the concept of empiricism promote?
function of molecules there
iii. These processes are referred to, generally as
outside-in and inside-out signaling
iii. Cell-adhesion molecules (CAMs)
1. We don’t necessarily have to define these terms on an exam, but they are referenced a lot in this lecture:
a. Some bind homophilic – only members of the same family can interact
i. CAM molecules themselves
b. Others bind heterophilic – associate with members of
another CAM family
c. May mediate homotypic interactions – association of cells of the same type or
d. Heterotypically – association of cells of different types Don't forget about the age old question of How does air circulation affect climate?
2. Four major families of CAMs – each family with functionally diverse members
a. Cadherins
b. Immunoglobin (Ig) superfamily
i. Antibodies are a member of this family
c. Integrins – can function as both CAMs, as well as adhesion receptors
d. Selectins
e. Members of each family share conserved domains (often found as repeated units within the cell) that are present in
multiple copies and therefore define the distance between
membranes of joined cells
i. Each family represents families as a result of
evolution – many members of each family
3. Two types of CAM interactions promote cell-cell adhesion: a. Trans interactions occur between CAMs on adjacent cells, while
b. Lateral interactions occur between CAMs on the same cell i. Cluster CAMs and promote more efficient trans
interactions between cells
c. Trans and lateral interactions are mutually reinforcing
4. General features of cell-cell adhesions
BSC 300-001 Week 12 Notes
a. Cell-cell interactions are dynamic – changing according to
the needs of the cell, the type of cell or extracellular matrix,
signals received and forces applied to the cells
b. Some CAMs require calcium for interactions, so regulated
release of Ca2+ plays important roles in their regulation
c. Association of intracellular molecules can alter CAM
binding, as well as promote clustering that enhances cell
cell associations
d. Other variables that affect CAM dynamics include:
i. Binding affinity of CAMs (thermodynamic
properties)
ii. Overall rate of association vs dissociation (kinetic
properties)
iii. Spatial distribution and abundance of CAMs
1. How many are produced
iv. Regulated active vs inactive state of CAMs
v. External forces like mechanical stress
(stretching/pulling)
vi. Turbulence of surrounding fluids
b. The Extracellular Matrix I: The Basal Lamina
i. The Extracellular Matrix (ECM) (Table 20-2 might be a good reference point)
1. A complex combo of proteins and polysaccharides secreted and assembled by cells into a cross-linked network
a. 4 major types of polysaccharides and proteins that re
secreted by the cells with which they are associated
2. Holds cells in place within tissues (example – autonomous cells of connective tissue)
3. Anchors epithelial cells to their substrate
4. Also provides positional and signaling info to cells, serving as a reservoir and conduit for the diffusion of signaling molecules
5. Serves as a lattice that can either prevent or promote (through guidance cues) the migration of cells – especially important during embryogenesis and a process hijacked by cancer during metastasis
6. Because individual cells of a tissue all associate with the ECM, it also serves as an indirect physical link between cells in tissue
7. ECM is connected to cell through transmembrane adhesion
receptors
a. Which through both mechano-sensation (AKA pulling on
our skin) and altered confirmation transduce info into the
cell
8. The ECM is not a static structure, but in a constant state of
dynamic remodeling through diverse cell regulated enzymatic
modification of its constituent components
9. Such remodeling results from enzymatically regulated:
a. Phosphorylation state
BSC 300-001 Week 12 Notes
b. Sulfation state
c. Cross-linking
d. Cleavage by proteases and glycosidases
e. Oxidation
f. Addition of glucose (glycation)
10. Principle ECM components:
a. Proteoglycans – core protein attached to sugar chains (GAGs – glycosaminoglycans)
b. Collagens – structural protein fibers that provide tensile strength
i. Absorb water and generate a cushion around the cell – important in receiving signals (Wnt and
Hedgehog)
c. Multi-adhesive matrix proteins – principally Fibronectin and laminin – important organizers of the ECM
i. Long, flexible molecules that contain multiple
domains
ii. Bind various collagens, other matrix proteins,
polysaccharides, and extracellular signaling
molecules and link the ECM to adhesion receptors
iii. Often serve as guidance cues for migrating cells
during embryogenesis
11. Relative density of cells and ECM varies
a. Relative volumes occupied by cells and ECM vary greatly among different animal tissues
b. Dense connective tissue contains mostly tightly packed ECM fibers interspersed with rows of relatively sparse fibroblasts (cells that synthesized ECM of connective tissue)
c. Sparse ECM is typical of epithelial cells – here squamous epithelial cells are tightly packed into a quilt-like pattern with little ECM between the cells
12. Molecular and Species Specificity
a. While CAMs and adhesion receptors can be divided into clear homolog families, their binding is so specific that even between closely related species cells with the same identity will preferentially associate with same-species cells following mechanical disruption (a classic developmental biology experiment by HV Wilson)
i. These specific interactions are driven by CAMs,
adhesion receptors and ECM molecules – as shown
in C & D. Beads coated with a collagen ECM
protein from two species of sponge preferentially
associate only with beads coated with the collagen
from the same species
BSC 300-001 Week 12 Notes
ii. Separated two sponges and the mixed them together – assumed orange would only interact with orange
and yellow only with yellow – not the case
iii. Evidence that cell interactions are driven by
different interactions with CAM proteins
13. Cell adhesion and morphogenesis
a. Cell-cell and cell-ECM associations play critical roles during embryo morphogenesis – when cell movements and rearrangements create the 3D structure of tissues and organs
i. Branching is a common theme in many tissues
(blood vessels, lung alveoli, etc)
ii. Some antibodies that bind and inhibit function of
fibronectin impede branching
b. Adhesion molecule diversity
i. The evolution of adhesion molecules was a critical step in the evolution of multicellular animals
(Metazoa)
ii. Many of these molecules are shared (have
homologs) in organisms as distantly related as
humans and sponges – others evolved as
components of unique types of tissue
iii. A common feature of adhesive proteins is repeating, nearly identical domains – referred to as repeats –
that generate the larger protein. Each domain, or
repeat, often encoded by a single exon
iv. The number and combination of these repeats
determines the overall size and binding specificity
as well and number and type of interactions
v. The evolution of gene families arose through gene duplication and divergence as well as accidental
“exon shuffling” between genes with similar
sequence
vi. Protein isoform diversity (the number of similar
proteins expressed in a species) is largely the result
of:
1. Families of multiple genes that arose by
duplication and divergence
2. Alternative splicing of exons that generates
multiple distinct isoforms from the same
gene (primary transcript)
c. Cell-adhesion molecules mediate signal transduction i. CAMs and adhesion receptors do not simply hold
cells in place, but also respond to changes in the
ECM, an interacting cell’s CAM expression or
mechanical force to transduce signals
BSC 300-001 Week 12 Notes
ii. Binding of ECM ligands to an integrin adhesion
receptor can stimulate many of the same
transduction pathways we have previously
described, and in doing so regulate:
1. Migration
2. Proliferation
3. Apoptosis
4. Enzymatic activities
d. Cell-adhesion molecules mediate mechanotransduction
i. Mechanotransduction – the reciprocal
interconversion of mechanical force and
biochemical processing (intracellular signaling) that
can result in altered gene. Expression, enzymatic
activity and/or mechanical action
1. In other words, CAMs and adhesion
receptors can serve as activators of signal
transduction into the cell (outside n
signaling), while signaling pathways that
converge on these molecules can alter CAM
and adhesion receptor
function/conformation and lead to inside out
signaling (communication with the ECM or
another cell)
2. Such mechanotransduction results from
physical stress on tissue that stretches
components of the ECM – like fibronectin
and talin
a. This alters the proteins’
conformation and exposes
interaction domains
b. This can, in the case of fibronectin,
allow the proteins to polymerize and
form fibers that strengthen the ECM,
or
c. In the case of talin (an intracellular
adapter for integrins) mechanical
stress exposes binding sites for an
actin binding protein called vinculin
which links integrins to the actin
cytoskeleton forming focal adhesions
in migrating cells or cells undergo
physical stress
c. Cell-Cell and Cell-Extracellular Matrix Junctions and Their Adhesion Molecules i. Principal types of epithelia
1. Epithelia – sheets of tightly connected sessile (non-motile) cells polarized into apical, lateral, and basal surfaces. Critical roles in
BSC 300-001 Week 12 Notes
animal development as migrating sheets that contribute the reorganization of germ layers during gastrulation and organ sculpting
a. All surfaces have distinct characteristics and function
b. General epithelial arrangements:
i. Simple columnar for absorbing/secreting (like
digestive tract)
ii. Simple squamous – line blood vessels (rapid
diffusion through the cells)
iii. Transitional line cavities subject to expansion
iv. Stratified protective layers (like our skin or
esophagus)
c. All epithelial cells are:
i. Anchored to a specialized ECM at their basal
surfaces via cell junctions called hemidesmosomes
and focal, fibrillary and 3D adhesions. The
specialized ECM is a dense framework that
contributes to organ/tissue morphology called the
basal lamina
ii. Tightly associated to one another through a number
of types of cell junctions called:
1. Tight junctions
2. Adherens junctions
3. Desmosomes
4. Gap junctions
iii. All of these structures link the extracellular
environment to the cytoskeleton and diverse signal
transduction pathways
ii. Principal types of cell junctions connecting the columnar epithelial cells lining the small intestine (we need to be familiar with the definition and function of each type of cell adhesion and the intracellular adapters that function with them) (Table 20-3 is a good reference for the subsequent
slides – EVERYTHING IN BLUE IS STUFF THAT I
INCORPORATED FROM THAT TABLE, NOT NECESSARILY NOTES THAT WERE IN THE SLIDESHOW)
1. Anchoring Junctions
a. Adherens junctions
i. Adhesion type – cell-cell
ii. Principal CAMs or Adhesion Receptors – Cadherins
1. Cadherins mediate cell-cell adhesions in
adherens junctions and desmosomes
a. Transmembrane glycoproteins that
mediate Ca2+ dependent cell-cell
adhesion
b. Mediate cell-cell adhesions through
homophilic interactions
BSC 300-001 Week 12 Notes
i. Can only interact with
cadherins of the same
subtype
c. Homophilic interactions that are
Ca2+ dependent:
i. Intestinal endocrine cells do
not express cadherins and do
not aggregate into sheets
ii. Same cells expressing
cadherin transgene:
iii. Ca2+ treatment, cells adhere
into a sheet
iv. Without Ca2+ cells do not
adhere
2. More than 100 protein types – but four
general families
a. E-cadherin (epithelial)
i. E-cadherin mediates adhesive
connections in cultured
MDCK epithelial cells
ii. E-cadherin clusters mediate
initial attachment of cells into
sheets
iii. Experiment:
iv. Time course study of
transgenic fluorescent E
cadherin in cultured kidney
cells
v. Fluorescent E-cadherin
distribution changes over
time to localize only the cell
junctions
b. N-cadherin (neural)
c. P-cadherin (placental)
d. Desmosome cadherins
iii. Cytoskeletal Attachment – Actin filaments
iv. Intracellular adapters – catenins, vinculin
v. Function – shape, tension, signaling, force
transmission
vi. Protein constituents of typical adherens junctions 1. Adherens junctions – a cell junction basal
to tight junctions, whose cytoplasmic face is
linked to the actin cytoskeleton. They
contribute to epithelial cell shape, changes
in cell shape and establish tensile strength in
the epithelia
BSC 300-001 Week 12 Notes
a. E-cadherin: principle CAM of
adherens junctions
i. Exoplasmic domains
clustered by cis and trans
interactions at adherens
junctions on adjacent cells
b. Cytosolic domains
i. Bind directly/indirectly
through adapter proteins
(principally catenins) to actin
filaments in the cytoskeleton
ii. Such adapter proteins can
transduce signals to cell
interior – Beta-catenin, for
example can function as
transcriptional activator
b. Desmosomes
i. Adhesion type – cell-cell
ii. Principal CAMs or Adhesion Receptors –
Desmosomal Cadherins
iii. Cytoskeletal Attachment – Intermediate filaments iv. Intracellular adapters – Plakoglobin, plakophilins, desmoplakins
v. Function – Strength, durability, signaling
vi. Desmosomes utilize two specialized cadherins – desmoglein and desmocollin
1. Strong cell-to-cell adhesion types found in
tissue that experience lots of mechanical
stress
2. Randomly scattered throughout lateral
surfaces of epithelial cells
3. Cytosolic domains bind intracellular adapter proteins (plakoglobin and plakophilins)
which bind a third adapter, desmoplakin
a. Collectively these adapters create the
thick cytoplasmic plaques
characteristic of desmosomes – do
not need to memorize plakoglobin
and plakophilins
b. Desmoplakins directly bind
intermediate filaments which radiate
into the cytoplasm and provide
strength to these cellular rivets
c. Hemidesmosomes
i. Adhesion type – cell-matrix
BSC 300-001 Week 12 Notes
ii. Principal CAMs or Adhesion Receptors – Integrin (alpha 6 beta 4)
1. Integrins mediate cell-ECM adhesions
a. Epithelial cells are anchored to the
basal lamina via integrin proteins,
both within and outside of anchoring
complex called hemidesmosomes
b. Integrins function both as adhesion
receptors as well as CAMs
c. 24 known alpha/beta heterodimers
promote broad ligand binding
potential
d. 3 principle binding domains:
i. RDG domain – binds
proteins containing Arg-Gly
Asp sequence
ii. Laminin binding domain
iii. The I-domain – collagen
interaction
e. Relatively high Kd (low binding
affinity), but the hundreds of
thousands of interactions in a single
cell promotes robust attachment
f. Diversity of integrin dimers and their
associations allow them to
participate in a variety of cellular
processes ranging from
inflammatory response to cell
migration
g. Like cadherins, integrin dimers of
focal, fibrillary and 3D adhesions are
linked via adapter to the actin
cytoskeleton – where they maintain
or modify cell shape, participate in
cell signaling and movement
h. Integrins of hemidesmosomes
interact with intermediate filaments –
where they maintain cell shape,
rigidity, and participate in signal
transduction
i. In addition to binding the ECM,
integrins are also bound by a diverse
ligand pool that can mediate signal
transduction via altered
conformation of the dimer and
signaling through various adapter
BSC 300-001 Week 12 Notes
proteins. Also, intracellular protein
interactions can alter integrin
conformations in such a way as to
promote inside-out signaling and the
interaction with other cells or the
ECM
iii. Cytoskeletal Attachment – intermediate filaments iv. Intracellular adapters – plectin, dystonin/BPAG1
v. Function – shape, rigidity, signaling
d. Focal, fibrillar, and 3D adhesions
i. Adhesion type – cell-matrix
ii. Principal CAMs or Adhesion Receptors – Integrins iii. Cytoskeletal Attachment – Actin filaments
iv. Intracellular adapters – talin, kindlin, paxillin,
vinculin kinase
v. Function – shape, signaling, force transmission, cell movement
2. Tight Junctions
a. Adhesion type – cell-cell
b. Principal CAMs or Adhesion Receptors – Occulin, claudins, JAMs
c. Cytoskeletal Attachment – Actin filaments
d. Intracellular adapters – ZO – 1,2,3, PAR3, cingulin e. Function – Controlling solute flow, signaling
f. Tight junctions seal off body cavities and restrict diffusion on membrane components
i. Tight junctions – specialized contacts at the apical most regions of the lateral surface of epithelial cells
1. Seals off body cavities
2. Located at apical end of the junctional
complex between adjacent cells
3. Serve as barrier to free diffusion of water
and solutes from the extracellular
compartment between cells
4. Some are permeable to specific ions or
solutes
g. Protein components of tight junctions
i. Primarily composed of the two TM proteins
occludins and claudins
ii. Link tight junctions to the cytoskeleton via various adapter proteins possessing PDZ domains
iii. Also contain Junction Associated Molecules
(JAMs) that promote homophilic adhesion via Ig
domains
iv. Transport across epithelia requires either
transcellular pathway requiring membrane
BSC 300-001 Week 12 Notes
transporters (example glucose absorption in the
intestine) or paracellular transport – small
molecules and ions that are selectively permeable to
cell-type specific tight junctions
3. Gap Junctions
a. Adhesion type – cell-cell
b. Principal CAMs or Adhesion Receptors – Connexins, innexins, pannexins
c. Cytoskeletal Attachment – Via adapters to other junctions d. Intracellular adapters – ZO- 1,2,3
e. Function – Communication, small-molecule transport between cells
f. Gap Junctions: Mediating Intracellular Communication i. Gap Junctions – regulated channels between animal cells for intracellular communication
1. Composed entirely of the membrane protein
connexin – 21 different human connexin
genes – different sets of connexins are
expressed in different cell types
a. Two connexons join together to
create a gap junction
2. Connexins are organized into a barrel
shaped complex called connexon – 6
individual connexin proteins
3. Allows small molecules to pass directly
between cytosols of adjacent cells
a. Remain open most of the time but
some can be gated
ii. Compatibility differences between connexins either promote or prevent communication between
different cells
iii. Gap-junction intercellular communication (GJIC) allows the passage of low-weight molecules like
ions, hormones, and second messengers
iv. Many types of neurons are connected by GAP
junctions to aid in increasing the speed of neural
impulses
v. Gap junctions can allow integration of activities of individual cells of a tissue into a functional unit
vi. Also promote metabolic coupling, in which a cell transfers nutrients or intermediate metabolites to a
neighboring cell that is incapable of producing them
itself
1. Sharing
vii. Open-closed state can be regulated by a number of factors
BSC 300-001 Week 12 Notes
1. Eg post-translational modification
(phosphorylation)
2. As well as pH, Ca2+ concentration,
membrane potential, and intercellular
potential between adjacent interconnected
cells (“voltage gating”)
4. Plasmodesmata
a. Adhesion type – cell-cell
b. Principal CAMs or Adhesion Receptors – Undefined
c. Cytoskeletal Attachment – Actin filaments
d. Intracellular adapters – NET1A
e. Function – Communication, molecule transport between
cells
f. Plasmodesmata are cytoplasmic channels passing through
cell walls of adjacent plant cells
i. Lined by plasma membrane
ii. Contain a central structure, the desmotubule, a
membranous extension of the ER
iii. Serve as sites of cell-cell communication
iv. Many types of molecules pass cell to cell through
plasmodesmata – including some transcription
factors, nucleic acid/protein complexes, metabolic
products, and plant viruses
g. Only plant-specific structure we need to know
II. Lecture 21 – Continuation of Chapter 20
a. The extracellular Matrix I: The Basal Lamina
i. The Extracellular Space
1. Extracellular matrix (ECM) – an organized network beyond the
plasma membrane that plays key regulatory roles in determining
cell shape and activities. Extensively involved in cell signaling as
it regulates diffusion of ligands and their interactions with
receptors
a. Unique composition of fibrous proteins and proteoglycans
are secreted by and surround all cell types and this ECM is
involved in regulating diverse cellular physiology including
cell-cell contact, proliferation, migration, cell signaling,
gene expression, and differentiation
b. Organizes cells into tissues and coordinates cellular
functions by activating transduction pathways that control
cell growth, division, and gene expression
c. Controls rates of signaling molecule diffusion and serves as
a repository of inactive or inaccessible signals that are
released when the ECM is disassembled or digested by
proteases
BSC 300-001 Week 12 Notes
d. The collective proteins that compose and modify the ECM is known as the matrisome – over 1000 human genes
encode the matrisome
e. Adhesion receptors bind to three types of molecules
abundant in the ECM of all tissues:
i. Proteoglycans – a group of glycoproteins that
cushion cells and bind diverse extracellular
molecules
ii. Collagen – fibers which provide structural integrity
and mechanical strength and resilience
iii. Multi-adhesive matrix proteins – soluble proteins
that bind and cross-link adhesion receptors and
other ECM components
ii. Basal lamina – specialized ECM under the basal surface of epithelia 1. Maintains cell attachment
2. Provide signals for cell survival (survival of most epithelial cells is anchorage dependent)
3. Separate distinct tissues within an organ
4. Serves as substratum for cell migration (critical during
embryogenesis)
5. Forms a barrier to macromolecules (for example the blood-brain barrier)
6. Major protein components of the basal lamina
a. Four ubiquitous protein types, synthesized by the cells
anchored to the basal lamina. Basal lamina of each tissue
composed of different combinations of these four principle building blocks
i. Each protein is composed of multiple distinct
repeats
1. Proteoglycans are interspersed between all 4
layers
ii. Type IV collagen – trimeric molecules with rod
like and globular domains that form a 2D network
onto which the other proteins are attached
1. All collagens, regardless of their type, are
homo- or heterotrimers, composed of a
combination of the collagen alpha chains
encoded b 43 human genes
2. These three subunits coil together into a
collagenous triple helix
3. 28 types of human collagen participate in
the formation of distinct ECMs in various
tissues
a. Do not memorize the 28 types –
recognize the 5 different divisions
(fibrillar collagens, fibril-associated
BSC 300-001 Week 12 Notes
collagens, sheet-forming and
anchoring collagens, transmembrane
collagens, and host defense
collagens)
4. The collagen triple helix
a. Fibrillar collagens
i. Trimeric protein composed of
three alpha chain
polypeptides – can be
homotrimer or heterotrimer
b. Collagen structure – longitudinal
view:
i. Repeating Gly-X-Y amino
acids of collagen alpha chains
(X-Y can be any protein, but
most often proline and
hydroxyproline)
ii. Each chain is twisted into a
left-handed helix – three
chains wrap around one
another in a right-handed
triple helix held together by
H-bonds
iii. Triple-helical structure with
left-handed twist of the
individual collagen alpha
chains
c. Collagen structure – cross-sectional
view:
i. Glycine proteon side chains
project into center of the
triple helix
d. The triple helix forms because of the
abundance of three amino acids:
glycine, proline and the modified
proline hydroxyproline
i. Proline is unique because it
folds onto itself
ii. Glycine is unique because it
only has H as its side chain
e. T-alpha chains are composed of
many repeated triple residues with
the sequence Gly-X-Y: where:
i. G = glycine
ii. X and Y can be any amino
acid, but predominantly
BSC 300-001 Week 12 Notes
X=proline and
Y=hydroxyproline (OH
attached to it) whose unique
cyclical structures promote
the curvature of the strands
iii. The small glycine (remember
it does not have a side chain,
but instead an H) allows the
crowded arrangement of the
triple helix
iv. The three strands are held
together by H-bonds
f. These triple helical structures are
considered a collagen monomer, but
assemble into the larger, more
complex 28 types of collagen
i. Diverse adhesion receptors
and signal transduction
receptors bind these distinct
types of collagen
g. Unique properties and affinities of
each type of collagen are due mainly
to differences in:
i. The number and length of the
triple-helical monomers
ii. Segments that flank or
interrupt the helical segments
and fold into other 3D
structures
iii. Covalent modifications to the
alpha chains, principally:
glycosylation, hydroxylation,
oxidation, and cross-linking
5. Structure and assembly of type IV collagen
a. For example, type IV collagen forms
the structural sheets of the basal
lamina
b. The principle units of mammalian
type IV collagen are called IV alpha
chains – there are 6 homologous
proteins that assemble into 3
different heterotrimers – each with
distinct properties
c. All type IV collagen forms a 400-
nm-long triple helix that is
interrupted by about 24 non-helical
BSC 300-001 Week 12 Notes
segments, that provide flexibility to
the helix
d. Globular domains at the termini
interact to form the branching,
irregular 2D lattice of the basal
lamina
iii. Laminin – multi-adhesive cross-shaped proteins that underlies the collagen network and binds
integrins and other adhesion receptors
1. Heterotrimeric multi-adhesive matrix
protein that cross-links basal lamina proteins
2. 16 vertebrate laminin isoforms from 5 alpha, 3 beta, 3 y chains – each with tissue and
developmental stage expression specificity
3. Coiled-coil region – these peptides
covalently linked by disulfide bonds
4. Globular domains at N-termini of each
subunit bind to one another mediating self
assembly
5. Five C-terminal globular LG domains of the alpha subunit mediate Ca2+ dependent
binding to cell0surface laminin receptors,
including integrins, sulfated glycolipids,
syndecan, and dystroglycan (do not worry
about these names that they bind to – just
know that it is the most diverse)
iv. Nidogen and perlecan molecules crosslink basal lamina networks
1. The proteoglycan perlecan cross-links
components of the basal lamina and cell
surface receptors
a. Perlecan is the major secreted
proteoglycan in basal lamina
b. Like all proteoglycans, it contains a
large filament-like core protein to
which many oligosaccharides are
attached
c. The protein core has multiple copies
of three protein interaction domains
(LG, EGF, and IG domains)
d. And is covalently linked to three
types of oligosaccharides: N-linked,
O-linked, and the
glycosaminoglycan (GAGs) called
heparan sulfate
BSC 300-001 Week 12 Notes
e. It is the presence of GAGs that
define proteoglycans as a specific
category of glycoproteins
f. The multiple protein interaction
domains and three types of
oligosaccharides give perlecan a
wide-range of potential interacting
partners including cell surface
receptors, ECM components and
growth factors
i. It therefore serves as the
principle cross-linking agent
of basal laminae as well as a
repository and conduit for
cell-cell signaling
b. The Extracellular Matrix II: Connective Tissue
i. Diversity of collagens
1. Most collagen of the body is fibrillary and located primarily in connective tissue (90%, Types I, II, and III)
2. These proteins are produced by fibroblasts of connective tissue and secreted into the ECM
3. Less abundant collagen includes:
a. Fibril-associated collagens that cross-link other collagens
b. Sheet forming and anchoring collagens that generate a 2D
network in basal laminae
c. Transmembrane collagen which functions as adhesion
receptors
d. Host defense collagens which help the immune system
identify and eliminate pathogens
ii. Synthesis and secretion of collagen
1. Because of their large size when mature, ECM collagens cannot assemble as fibrils intracellularly. Rather, after translation into the ER they contain N- and C- termini propeptides that inhibit
polymerization
2. Such immature collagen proteins are called pro-alpha chains 3. In the ER:
a. The pro-alpha chains receive N-linked glycosylation on
their C-terminal propeptide
b. Propeptides associate to form trimers and are covalently
linked by di-sulfide bonds
c. Prolines and hydroxyprolines of G-X-Y are hydroxylated
and some prolines undergo cis to trans isomerization
d. Some hydroxylysines are covalently attached to galactose
e. These modifications facilitate formation of the triple helix,
which is stabilized by a chaperone protein to prevent
premature aggregation
BSC 300-001 Week 12 Notes
4. In the Golgi
a. Lateral association via H-bonding occurs and the chains are secreted from the cell
b. Procollagen peptidases remove N and C termini
i. Exposes interaction domains to allow different fiber
aggregates to assemble with each other
c. Trimers assemble into staggered arrays forming fibrils and are covalently cross-linked via their exposed termini
d. Within these termini lysine and hydroxylysine residues are oxidized to aldehydes that form the covalent cross-links
that hold the trimers together
i. Forms very long and stable structures
5. Human health relevance – the post-translational modifications to the alpha chains are essential for forming the mature collagen fibers
6. For example, ascorbic acid (vitamin C) is a necessary cofactor for the hydroxylase that add hydroxyl groups to prolines and lysines a. In the absence of vitamin C hydroxylation of the monomers does not take place and collagen fibers disassemble
b. The ancient mariner disease Scurvy resulted from lack of vitamin C in sailor’s diets causing degeneration of
connective tissue
i. No vitamin C = no collagen formation = lots of
nasty stuff (loss of teeth, etc)
ii. This is why British soldiers are called Limeys
iii. Type I and II collagen
1. Type I collagen form long fibers with great tensile strength a. For example, tendons are primarily composed of type I
aligned in long parallel fibers capable of stretching and not
breaking
b. Other collagens (type VI) and proteoglycans associate with these fibers and non-covalently link them together into
strong collagen fibers
c. Type I is also used in as reinforcing rods in bones
2. Type II does not form fibers, but instead generates more mesh-like networks, like that found in cartilage
a. Here, other accessory collagens (type IX) cross-link type II collagen to matrix proteoglycans
3. Diverse human health implications:
a. Osteogenesis imperfecta: aka brittle bone disease. Can result from a single point mutation in the alpha 1 genes that encode osteogenic collagen. A single mutation to almost
any of the encoded glycines promotes poorly formed and
unstable triple helices. Autosomal dominant recessive –
meaning only copy (allele) of the gene must be mutated for
the disease to manifest
BSC 300-001 Week 12 Notes
b. Dominant or recessive muscular dystrophy: muscle
weakness, respiratory insufficiency, muscle wasting and
joint abnormalities
c. A variety of skin and vascular diseases
iv. Proteoglycans and their constituent GAGs play diverse roles in the ECM 1. As we saw with perlecan, proteoglycans are secreted glycoproteins with a core protein covalently cross-linked to specialized
polysaccharide chains called glycosaminoglycans (GAGs)
a. GAGs are long, linear polymers of specific repeated
disaccharides: one sugar is usually a uronic acid or
galactose, while the other is N-acetylglucosamine or N
acetylgalactosamine
i. One or both sugars have at least one anionic group
(carboxylate or sulfate), so each GAG bears many
negative charges
v. Gels of polysaccharides resist compression
1. Four major GAGs:
a. Hyaluronan
b. Chondroitin sulfate
c. Heparin/Heparan sulfate
d. Keratan sulfate
2. All but hyaluronan are sulfated, and all but hyaluronan are the major GAGs that occur naturally as a part of proteoglycans
3. The arrangement of sugars in GAG chains and their various covalent modifications determine their function and that of the proteoglycan in which they are found
a. For example, certain GAGs of heparin sulfate
proteoglycans control the binding of specific growth factors or morphogens to their receptor or the activities of protein
involved in blood clotting
4. Typically, many GAG chains are attached to a core protein, which may then be linked end to end to form a huge molecule
5. GAGs can assemble as complex chains
6. The negatively charged molecules attract cations like Na+ which draws water into the matrix forming a gel-like solution that
provides cushioning to the embedded cells
vi. The repeating disaccharides of glycosaminoglycans (GAGs) 1. Syndecans are cell-surface proteoglycans expressed in epithelial and non-epithelial cells that bind collagen and multi-adhesive matrix proteins
a. Link ECM to cytoskeleton
2. As with adhesion receptors, the cytosolic portion of syndecan interacts with the cytoskeleton and intracellular regulatory proteins 3. For example, in the hypothalamic region of the brain that controls eating behavior in response to food depravation and regulates binding of anti-satiety peptides to their receptors
BSC 300-001 Week 12 Notes
a. Anti-satiety peptides tell our brain that we are no longer
hungry
b. People who cannot seem to control their appetite may have an issue with anti-satiety peptides
c. Mice engineered to over-express syndecan1 never
experience fullness after eating and continue to overeat,
becoming obese
vii. Hyaluronan, aka hyaluronic acid (found in many lotions because it is very large sized)
1. The only non-sulfated GAG is a major component of the ECM surrounding migrating and proliferating cells, especially during embryogenesis
2. Also forms the backbone on large complex proteoglycan aggregates found in many ECMs, particularly cartilage
3. It imparts stiffness and resilience as well as lubricating properties to many types of connective tissues and joints
4. Typical hyaluronan molecule has 10,000 repeats that bend and twist into many configurations
5. Because of the large number of anionic residues on hyaluronan it attracts a large amount of water and swells into a hydrated sphere that cushions cells and maintains osmotic pressure – preventing cell swelling in hypotonic solutions
6. The predominant proteoglycan in cartilage, called aggrecan, assembles. With hyaluronan into a large complex aggregate that gives cartilage its gel-like properties and resistance to deformation
7. A core hyaluronan molecule is bound by hundreds of aggrecan proteoglycans to generate one of the largest macromolecules known – about the size of a bacterial cell
viii. Organization of fibronectin and its binding to integrin
1. Fibronectin (Fn) is an abundant multi-adhesive domain matrix protein found in all vertebrates
2. About 20 isoforms derived from alternative splicing of a single transcript
3. Essential for cell migration and differentiation during
embryogenesis, as well as wound healing through promotion of blood clotting
4. Dimers of two polypeptides linked by disulfide bridges at their C termini
5. Each protein composed of several functional regions each with distinct binding affinities
6. Each region contains multiple copies of three types of domains: Fn I, II, and III repeats
a. Only be familiar with Type III
7. Type III repeats contain the tri-peptide RGD motif (Arg-Gly-Asp) which is recognized by integrins – other integrin binding proteins likewise possess RGD motifs
BSC 300-001 Week 12 Notes
ix. Integrins mediate linkage between fibronectin in the ECM and the cytoskeleton
1. Inside out signaling: to promote robust adhesion to the cell’s substrate, binding of cell-surface integrins to fibronectin in the
ECM induces cytoskeletal dependent movement of integrin in the plasma membrane
a. This generates force on fibronectin proteins, causing them
to unfold
b. Activated fibronectin molecules aggregate into fibrils that
mature into stable matrix components via covalent cross
linking
2. Matrix metalloproteases remodel and degrade the ECM
a. Processes like cell migration, wound healing, immune
response and proliferation require not only an ECM but
also its remodeling or degradation
b. This is driven by cell synthesized zinc-dependent proteases
– the principle class being matrix metalloproteases – 23
different ones in humans
i. Do not memorize the names of any of them
ii. Cancer hijacks the expression of MMPs, which
promotes metastasis
c. Adhesive Interactions in Motile and Nonmotile Cells
i. Interactions of Cells with Extracellular Materials
1. Focal Adhesions (all we really need to know about integrins at this point is that their conformation can promote inside-out and
outside-in signaling)
a. Focal adhesions – docking sites where cells adhere to their
substratum and send signals to the cell interior
i. Cytoplasmic domains of integrins contain binding
sites for a variety of cytoplasmic proteins
ii. These proteins relay both mechanical and chemical
information to the cell interior altering cytoskeleton
dynamic and gene expression
iii. Can be rapidly assembled/disassembled in order to
promote cellular migration
ii. Regulation of integrin-mediated adhesion and signaling controls cell movement
1. Integrin binding: integrins exist in low affinity, inactive
conformations and active high affinity states
2. In the inactive state the dimer subunits are closely positioned and bent, while in the active state, the dimer is extended and its ligand binding domains are exposed with higher affinity and its
cytoplasmic tails are separated allowing binding to adapter proteins 3. This dual conformational change explains the ability of integrins to participate in both outside-in and inside-out signaling
BSC 300-001 Week 12 Notes
4. Binding of extracellular ligands or ECM components alters cytoplasmic conformation and accessibility to adapter proteins like talin
5. Similarly, binding by adapter proteins like talin, causes the extracellular domains to extend and allow interaction with the ECM
BSC 300-001 Week 13 Notes
I. Lecture 22 – Stem Cells, Cell Asymmetry, and Cell Death (Ch 21) a. Early Mammalian Development
i. Overview of the birth, lineage, and death of cells
1. Not all cell divisions are symmetric – meaning both daughter cells receive the same amount of cytoplasm and the various proteins in
the parental cytoplasm
2. Examples of symmetric division include yeast, fungi, and most
protists. Similarly, mature liver hepatocytes divide symmetrically,
producing both genetically as well as functionally identical
daughter cells
a. Don’t have a lot of differentiation
3. But, while every cell in a multicellular organism has the exact
same genotype (with a couple of exceptions like immune cells as
we’ll see in Ch 23) there are many unique cell types, each
expressing only a portion of the genes within their genome
a. Cells in our body have all the same genes but don’t express
the same ones
4. How does cell diversity arise in multicellular organisms?
a. One mechanism that contributes to cell diversity is
asymmetric cell division, in which daughter cells receive
different amounts or components of the cytoplasmic
material
i. Such daughter cells may differ in size, shape,
protein composition – all of which may cause them
to function/behave differently and importantly
transcribe different sets of genes, which lead them,
and their descendants, among very different fates
ii. Asymmetric division during animal development is
one of the fundamental mechanisms used to
establish cell lineages – a line of descent from a
founder cell that acquired a difference in gene
expression compared to a sibling cell
iii. Separate cell lineages become more and more
restricted in developmental potential as they
proliferate and acquire unique combinations of gene
expression
1. Ultimately such cells exit the cell cycle as
fully differentiated cells
5. Metazoan development begins with the union of two gametes: ova and sperm (haploid) which combine their haploid genomes to
generate the first cell of a metazoan organism – the zygote
(diploid)
a. For some organisms, the first cell divisio of the zygote is
asymmetric – producing daughter cells with unique protein
complements that direct differential gene exression, and
those whose developmental fate is therefore restructured
very early
BSC 300-001 Week 13 Notes
b. In other organisms, like humans, early cell divisions
produce daughter cells that are totipotent – capable of
giving rise to any cell lineage
i. In humans, however, at the 16 cell-stage – known as
blastula state, some cells become committed to
generating the extraembryonic tissues population
(trophoblasts), while a smaller cell population –
the inner cell mass – in the interior of the embryo
remains pluripotent – capable of generating all of
the lineages of the embryo body
6. Major Stages During Mammalian Embryo Development
a. Blastula have a fluid-filled interior called the blastocoel and
the ICM
i. Both ICM and TE cells are stem cells -they can
proliferate indefinitely in cell culture and can be
induced to adopt any of the cell fates that these cell
types would generate during development, provided
they are cultured with the appropriate growth and
differential factors to push them down that ppath
b. Embryonic Stem Cells and Induced Pluripotent Stem Cells
i. Genomic equivalence and animal cloning
1. In 1996 Ian Wilmut at the University of Edinburgh proved that mammalian adult cells possess genomic equivalence – all somatic cells of an adult possess the same genetic material
a. They are difference because each cell type expresses a
different subset of gees
b. Wilmut showed that the nucleus of a differentiated adult ell
could be transplanted into an enucleated ovum and be
induced to begin development and give rise to a genetic
clone of the animal that donated the nucleus. This
technique is called somatic cell nuclear transfer
i. Showed that all cells in the adult body have the
exact same genetic material in them
c. The first mammal that was successfully cloned was a sheep
named Dolly. During SCNT the donor genome undergoes
reprogramming and most of the epigenetic code from the
donor genome is erased – wiping the slate clean of the
epigenetic programming of the differentiated donor cell
i. Does not erase all programming, so they get
different diseases that manifested in their life much
faster than usual
d. Dolly and subsequent clones generated by SCNT
experienced diverse medical problem that stemmed from
the fact that not all epigenetic codes were cleared
i. Epigenetic codes are the various histone and DNA
covalent modifications like acetylation and
methylation that accompany genomic differentiation
ii. Genomes display genetic totipotency
BSC 300-001 Week 13 Notes
1. Somatic cell nuclear transfer for a while held the promise of incredible scientific investigation and therapeutic advancement a. Ex: this technique can be used to generate an early embryo clone of any individual, and from the blastula stage embryo harvest embryonic stem cells for research or therapy
b. However, this technique still had great ethical challenges. Embryos must be destroyed to harvest the embryonic stem
cells
iii. Transcriptional network regulating pluripotency of ES cells 1. Shinya Yamanaka shared the 2012 Nobel Prize I nMedicine of Physiology for his discovery that a small number of transcription factors are exressed in ICM cells and maintain them as stem cells 2. Three master transcription factors – Oct4, Sox2, and Nanog a. Oct4 and Nanog – expressed only in ES cells
i. Sox2 is also expressed in multipotent neural stem
cells that give arise exclusively to neuronal and glial
cell types
b. These transcription factors bind one anothers’ promoters, establishing positive autoregulatory loops
c. They also regulate expression of many other genes, which: i. Encode proteins important for the proliferation and
self-renewal of ES cells
ii. Repress genes and micro-RNAs that are essential
for cell differentiation
3. Amazingly, Yamanaka found that forced expression of these transcription factors in differentiated cells reprograms them into cells virtually identical to embryonic stem cells. Such
reprogrammed cells are now known as induced pluripotent stem cells (iPSCS), and obviate the need to create an embryo
a. Cultured differentiated cells from any individual can be
reprogrammed as iPSCs
b. Work subsequently to Yamanaka showed that one of the critical jobs of Oct4 is to remove epigenetic silencing of
genes to establish stem-cell fate
i. Doing this, they worked out mechanisms for erasing
the genetic code and created embryonic stem cell
4. Two potential medical applications of iPSCs
a. iPSCs can be induced to develop and differentiate as a
mature cell type
b. Mature cells from a patient can be harvested and
transformed into patient-specific iPSCs
c. Ways that medicine is advancing new techniques to cure diseases using embryonic cells
i. New technology (like CRISPR-Cas9) could be used
to repair genetic mutations
1. Repaired iPSCs would then undergo in vitro
differentiation before transplant back into
the patient. Importantly, because this
BSC 300-001 Week 13 Notes
technique uses patient-specific cells there is
no fear of host-rejection
ii. Alternatively, differentiated patient-specific cell
types affected by a specific genetic disorder could
be used to screen for drugs that reverse or treat the
specific disease
d. Such thereapeutic approaches are close for diseases like
ALS, Cystic fibrosis, diabetes, etc
5. Production of normal insulin-secreting Beta islet cells from human iPS or ES cells
a. What is required for such approaches, however, is knowing
the appropriate combination of growth factors to induce
iPSCs to specific developmental fates
i. Doug Melton, at Harvard, for example has
researched the discovered the correct sequence and
timing of growth factors required to transform
iPSCs into pancreatic insulin secreting Beta cells
ii. Mice pancreas transplanted with human iPSC
derived B cells repond to increase blood sugar
levels and appropriately promote its uptake and
respiration
6. Multipotent stem cells generate a continuous supply of terminally differentiated cells
a. Terminally differentiated cells do not divide, so where do
replacement cells come from?
i. Most tissue types contain populations of
underdifferentiated self-renewing cells known as
multipotent stem cells (aka somatic or adult stem
cells)
1. These are distinct from embryonic
pluripotent ste cells in that they are not
pluripotent. Their developmental potential is
restricted by expression of tissue-specific
transcriptional regulators
c. Stem Cells and Niches in Multicellular Organisms
i. The pathway from stem cells to lineage-restricted progenitors to differentiated cells
1. Multipotent somatic stem cells have three key properties:
a. They can give rise to multiple types of differentiated cells
b. They are undifferentiated
c. They increase in number during embryogenesis and then
remain relatively constant over an individual’s life. In this
sense they are immortal
i. Give rise to a set number that will reside for the rest
of our lives
2. MP stem cells often divide asymmetrically – producing daughters that remain as undifferentiated stem cells and non-stem cell
BSC 300-001 Week 13 Notes
daughters that undergo multiple rounds of division as transit amplifying cells before differentiating as mature cells
ii. Patterns of stem-cell differentiation
1. However, some stem cell populations responod to hormonal signals that can alter this pattern of division and produce identical stem cell daughters in order to increase stem cell number, or produce identical transit amplifying daughter cells to increase the pool of proliferating cells that will differentiate
iii. The stem cell niche
1. Stem cells require intrinsic regulatory signals, like the expression of specific transcription factors to maintain their fate
2. Additionally, stem cells require a specialized microenvironment known as the stem cell niche, that possesses extrinsic regulatory signals like hormones and growth factors
a. Stem cell niche provides the appropriate source of signals to maintain stem cells in their proliferative and self
renewing state
b. As well as providing the physical context for signals that will be received by proliferating precursor cells to receive
the signals that will promote their differentiation
3. Intestinal stem cells and their niche
a. For example, intestinal epithelial cells turn over every 3-5 days
b. The intestinal epithelium generates microvilli – long
protrusions into the lumen of the gut in order to increase
surface area
i. At the base of each microvillus lies a specialized
compartment known as the intestinal crypt: the
stem cell niche for intestinal epithelial cells
ii. Within the crypt lie the intestinal stem cells which
express the GPCR LGR5
iii. And a second population of cells called Paneth
cells
1. Paneth cells secrete many anti-microbial
peptides, and therefore play an important
role in immune function
2. They also express a Wnt ligand which
stimulates neighboring stem cells to
maintain their stem cell fate
3. Paneth cells secrete the ligand R-spondin
which binds the LGR receptor and
transduces a signal the potentiates Wnt
signaling
4. Therefore, Paneth cells – which are progeny
of intestinal stem cells themselves –
constitute much – if not all – of the niche for
intestinal stem cell maintenance
BSC 300-001 Week 13 Notes
c. As transit amplifying cells move up and away from the
stem cell population they are no longer exposed to the Wnt
and R-spondin signals
i. They proliferate as transit amplifying cells and
terminally differentiate as secretory and absorptive
cells within the villus
ii. Other precursor cells migrate downward and adopt
the Paneth cell fate
d. Mechanisms of Cell Polarity and Asymmetric Cell Division
i. General features of cell polarity and asymmetric cell division
1. Three core principles of cell polarity:
a. Cells have intrinsic cell polarity program that allows them
to polarize in the absence of external cues
b. The intrinsic program can be directed by internal and
external cues
c. Polarity of individual cells is often maintained by
intracellular mutually antagonistic complexes
2. Although the mechanism of location varies across the Eukarya, the small G protein Cdc42 plays a critical role in establishing cell
polarity
a. Once localized to discrete cell locations Cdc42 polarizes
the actin cytoskeleton and provides tracks for the
asymmetric distribution of secretory granules and
cytoplasmic proteins that promote asymmetric localization
and distribution of cytoplasm and protein within cells as
well as between daughter cells during division
3. Cell polarity determinants – mRNAs, proteins, and lipids:
a. Asymmetrically localized in mother cell
b. Mitotic spindle – positioned so that polarity determinants
are segregated differentially into daughter cells during cell
division
4. Cdc42 – master regulator of cell polarity
a. Intrinsic polarity program – establishes asymmetry in
cells before asymmetric cell division that yields cells with
different fates
b. Cdc42-GTP is required to establish cell polarity
i. Cdc42-GTP locally activates formins
ii. Recall, formins nucleate and elongate actin
filaments with (+) ends oriented toward Cdc42-GTP
at membrane
iii. Myosin V motor transports secretory vesicles
toward actin filament (+) ends
5. Establishing polarity in vertebrate epithelial cells
a. Vertebrate polarized epithelial cells – cues from adjacent
cells and the extracellular matrix orient polarization axis by
regulating location of three protein complexes
b. Interactions between adherens and tight junctions signal
epithelial cells to recruit the Par complex to these sites
BSC 300-001 Week 13 Notes
(Par stands for partitioning defective). The Par complex
includes regulatory proteins and the Cdc42 G protein
c. Apical to the Par complex another complex called Crumbs
is assembled, while basally the Scribble complex is
assembled
d. Though not completely understood, this arrangement of
protein complexes reorganizes the cytoskeleton:
i. Actin filaments – distinct arrangements associated
with the apical and basolateral membranes
ii. Microtubues – not all associated with a centrosomes
iii. Intermediate filaments – associated with basolateral
cell-cell adhesion junctions (desmosomes
hemidesmosomes) and ends of microvilli actin
bundles
e. This arrangement also polarizes membrane traffic to direct
transport vesicles to their appropriate membranes
e. Cell Death and Its Regulation
i. Apoptosis (Programmed Cell Death)
1. Apoptosis is an ordered and molecularly activated process
involving killing: molecules that activate the process, Destruction: proteins that destroy the cell from the inside, promoting cell
shrinkage, loss of adhesion to other cells, degradation of
chromatin, and Engulfement: phagocytosis by scavenger white
blood cells
a. Plays a key role during animal development as well as to
remove damaged cells from tissue
b. Apoptotic changes are activated by proteolytic enzymes
called caspases, which target:
i. Protein kinases, some of which cause detachment of
cells
ii. Lamins, which line the nuclear envelope
iii. Proteins of the cytoskeleton
iv. Caspace activated DNase (CAD) which degrade
DNA
c. Very deeply conserved process with genetic pathways
shared by all Metazoa
2. Death by apoptosis
a. Cells undergoing necrosis (uncontrolled cell death) exhibit
very different morphological changes:
i. Swell and burst
ii. Release intracellular contents that can damage
surrounding cells and cause inflammation
3. Apoptosis during development
a. Apoptosis may be activated due to a lack of an external
signal (trophic factor) that is necessary for survival. In its
absence, cells essentially commit suicide
i. Alternatively, other signals may signal the cell to
initiate apoptosis – essentially cellular murder
BSC 300-001 Week 13 Notes
b. During embryogenesis apoptosis sculpts tissue and organs by the controlled removal of specific cells
i. For example, vertebrate limb buds begin
development a paddle shaped appendages.
Apoptosis removes interdigital tissue to sculpt
hands, toes, paws, etc
c. Apoptosis is also active in the adult, where about 1010 – 1011 cells die everyday
d. Reduced or elevated apoptosis is linked to several human diseases:
i. Cancer – murder pathway can’t be activated
ii. Parkinson’s, Alzheimer’s, and Huntington’s
diseases – increased death
iii. Diabetes type I – increased death
4. Evolutionarily conserved proteins participate in apoptosis a. Comparison between major components of the apoptotic pathway in C. elegans and humans – color coded to
identify. Conserved homologous proteins
b. Pathways in both organisms converge on the activation of caspases – proteases bearing a conserved cysteine in their active site
c. In vertebrates and initiator caspase (Caspase 9) is activated by dimerization induced by upstream activatory proteins (principally Apaf-1 following inernal or external stimuli)
d. Activated initiator caspases cleave and activate effector caspases (principally Caspase 3 and 7, though there are many in vertebrates)
e. Prior to cleavage and activation, caspases and referred to as procaspases – inactivated form
5. Mitochondrial Mediated Apoptosis
a. Key regulatory proteins lie upstream of caspaces in the apoptotic pathway and fall into one of two categories: pro apoptosis or anti-apoptosis (pro-survival)
b. All of these proteins are always expressed and the pro survival proteins bind and inhibit the activities of the pro apoptotic proteins
c. All are members of a large family of proteins called the Bcl-2 family, which share up to 4 characteristic domains called the BH 1-4 domaisn (Bcl2 homology domains)
i. Through these domains the proteins oligomerize
and regulate one another’s activities and ability to
bind other proteins
6. Vertebrate Apoptosis and Mitochondria: Intrinsic Apoptosis a. In healthy vertebrate cells the anti-apoptotic protein Bcl-1 binds to Bak and Bax, which span the outer mitochondrial membrane – preventing them from oligomerizing
BSC 300-001 Week 13 Notes
b. In response to intrinsic signals like excessive DNA damage or loss of trophic factors or pro-apoptotic signals, other members of the Bcl-2 family are released
i. Puma is released from the nucleus, following
excessive DNA damage, and Bad being released
upon loss of trophic factors
ii. These pro-apoptotic factors bind either Bcl-2 or
Bak/BAx disrupting the complex
iii. Bak and Bax are able to oligomerize
1. Bak and Bax oligomerize to form a pore in
the outer mitochondrial membrane
2. This pore is specific to the Cytochrome C
peripheral membrane protein (of the ETC),
which is released into the cytoplasm
3. In the cytoplasm, Cyt C binds Apaf-1,
causing it to dissociate from Caspase-9
a. Caspase-9 cleaves the effector
procaspase-3 to an active form and
caspase-9 promotes proteolytic
cleavage of its many targets
7. Apoptosis: The Extrinsic pathway – Death receptor-regulated a. Initiated by external stimuli
i. Tumor necrosis factor (TNF) is detected by a TNF cell surface receptor
ii. The activated receptor recruits Receptor
Associated Death Domain proteins (TRADD and
FADD)
iii. These proteins bind and dimerize a different
initiator caspase (Caspase 8)
iv. Which cleaves other effector procaspases to active forms
v. Caspase 8 also cleaves a Bcl-2 family member
called Bid into an active form, which similar to the
intrinsic pathway promotes Bax and Bak activity
and the release of Cytochrome C peripheral
membrane protein – leading to activation of the
effector caspase Caspase-9
8. Apoptosis Clearance
a. Apoptotic cell death occurs without spilling cellular contents to prevent inflammation, compared to necrosis b. During apoptosis, a phospholipid “scramblase” moves phosphatidylserine molecules to the outer leaflet of the plasma membrane where they are recognized as an “eat me” signal by specialized macrophages
c. Apoptotic cells are cleared by phagocytosis
BSC 300-001 Week 14 Notes
I. Lecture 23 – Immunology Part I (Ch 21 Part I)
a. Overview of Host Defenses
i. Overview of Immune Defenses
1. Pathogenic attack has spawned an evolutionary arms race. Metazoa have evolved diverse mechanisms to destroy infectious agents, and
pathogens likewise continue to evolve means of evading or
disarming these systems
2. Many organisms possess an innate immune system that serves as
a rapid, somewhat specific, system for recognizing and destroying
invading pathogens
3. Vertebrates have evolved a complex adaptive immune system.
This system is highly specific and changes in response to new or
increased numbers of pathogens
ii. The three layers of Vertebrate Immune Defenses
1. Mechanical and chemical defenses provide protection against most pathogens – such protection is immediate and continuous, but has
little specificity
2. Mechanical defenses consist of epithelia and skin that serve as
barriers to infection, with associated cell derived molecules that
weaken potential pathogens
3. The innate immune system includes phagocytic cells, complement
proteins and glycoproteins called interleukins secreted by certain
white blood cells. Innate defenses are active within minutes to
hours of infection and have a small degree of specificity
4. Pathogens not cleared by the innate immune system are dealt with
by the cells and highly specific molecules of the adaptive immune
response
5. The adaptive immune response of vertebrates is activated within
days of the primary infection and includes four key features
lacking from the innate response shared by many Metazoa:
a. Specificity – it can distinguish between closely related
molecules
b. Diversity. -it can recognize millions, if not billions, of
unique molecules, or molecular structures
c. Memory – the host’s immune system can remember
previous infection and mount a faster and more specific
response upon re-exposure
d. Tolerance – it can avoid mounting an immune response
against the host’s own cells
6. These features arise from the production of diverse proteins
including antibodies and cell-surface receptors, each with unique
affinity for target molecules
iii. A few key terms:
1. Antigen – any material that can evoke an immune response
2. Antibody – specialized members of the Immunoglobin
superfamily that bind to specific molecular features of antigens
3. B-cells – White blood cells (lymphocytes) that mature in the bond marrow and express antigen-specific receptors. Once encountering
BSC 300-001 Week 14 Notes
an antigen, B-cells mature to become antibody-expressing plasma cells
4. T-cells – a lymphocyte that matures in the thymus and expresses antigen-specific receptors
a. Two major classes:
i. Cytotoxic T-cells that kill virus infected and tumor
cells
ii. Helper T-cells that are required for activation of B
cells
iv. Overview of Host Defenses
1. Our skin, GI and genital tracts collectively possess a surface area of approximately 4000 square feet, constantly being exposed to bacteria and viruses
2. Many bacteria are commensal – they live on and in us without causing harm, and in many cases are beneficial (they represent our microbiota)
3. Rupture of epithelia is the most effective and common route for viral and bacterial infection
4. Viral replication is restricted to host cell cytoplasm or nuclei, and can then spread to other cells following release as free viral particles or direct transfer from adjacent cells
5. Most bacteria replicate extracellularly, though some are intracellular obligates – either in intracellular vesicles or free in the cytoplasm
6. Therefore an effective host immune system must be capable of eliminating both extracellular pathogens as well as host cells harboring pathogens
v. Vertebrate immune defense: leukocytes
1. Leukocytes (white blood cells) include a variety of cell types including B and T cells, monocytes (precursors to scavenger cells called macrophages), dendritic cells, neutrophils and Natural Killer cells – each with distinct functions in the immune response
2. Lymphocytes (B and T cells) are born in the bone marrow and are mobilized in the circulatory system to sites where they mature and become activated (thymus gland, lymph nodes, tonsils and spleen)
3. The circulatory system also moves mature lymphocytes throughout the body where they exit to sites of infection
vi. Vertebrate Immune Defense: Lymph nodes
1. The pressure of the circulatory system forces cell-free fluid across the blood vessels (interstitial fluid) that bathes all cells in the body 2. A portion of this fluid is absorbed by lymphatic vessels and is conducted to enlarged capsules of the lymph system called lymph nodes (vis afferent vessels)
3. The arriving lymph carries cells that have encountered antigens, as well as soluble antigens themselves
4. Within lymph nodes the cells and molecules of the adaptive response encounter these antigens and count an appropriate
BSC 300-001 Week 14 Notes
response. They are activated, leave the lymph node (via efferent vessels) in lymph that is returned to the circulatory system
vii. The innate immune response following infection
1. Innate immune response is activated once the mechanical/chemical barrier is breached. It includes:
a. Phagocytes – a diverse group of cells (including
leukocytes: the phagocytic white blood cells macrophages,
neutrophils and dendritic cells) capable of ingesting foreign
cells and destroying them by phagocytosis. They are
widespread through our circulatory system, tissues and
epithelia
b. Several soluble proteins that are constitutively present –
known as the complement system
c. The phagocytic white blood cells are known as
professional phagocytes because they are much more
effective than non-professional cells at identifying and
removing foreign cells
d. Their professional status is a result of expressing specific cell surface receptors known as pattern recognition
receptors (PRRs) that recognize and bind broad patterns of
pathogen-specific markers like bacterial cell wall
molecules, nucleic acids lacking methylated guanines, and
double-stranded RNA
e. The most important of these PRRs is the Toll-like family of receptors (TLRs). Cells activated by their TLRs secrete
numerous effector molecules, including antimicrobial
peptides that destabilize biological membranes, causing
targeted bacteria to rupture
f. Macrophages and dendritic cells, once activated by their TLRs, become antigen presenting cells (APCs), by
processing and displaying foreign material on their cell
surfaces. These presented antigens are bound by and
activate antigen-specific T cells. Therefore the TLR system
of macrophages and dendritic cells serves as a bridge
between the innate and adaptive immune system
2. The innate response: Inflammasome and non-TLR nucleic acid sensors
a. A mammalian family of leucine rich repeat receptors
(LRRs) recognizes a wide array of foreign molecules
(bacteria cell wall components, uric acid crystals, bacterial
DNA etc)
b. In response to these cytosolic insults these proteins
stimulate assembly of a large protein complex called the
inflammasome, which activates a caspase called caspase-1
that stimulates secretion of interleukin cytokines
i. Cytokines – small secreted proteins that promote
inflammation and attract white blood cells of the
immune response
BSC 300-001 Week 14 Notes
3. The innate response: The Complement System
a. The complement system is a system of serum proteins that can bind directly to bacterial cell walls and fungal surfaces
b. Distinct pathways in response to different insults lead to the assembly of a bacterial membrane attach complex. For
example, in the classic pathway, pathogens bound by
antibodies recruit the complement system to destroy the
bacteria
c. All three pathways converge on the function of the protein C3 which promotes assembly of the membrane attack
complex
i. This promotes cell lysis as well as attracts
macrophages which ingest and destroy the
pathogens
ii. Additionally, this labeling of pathogenic bacteria
attracts and activates T-Cells of the adaptive
immune system- hence its name the complement
system
4. The innate response: Natural killer Cells
a. The innate immune system also functions to eliminate cells that have been infected by viruses
b. Cells infected by viruses secrete cytokines called type 1
interferons, which activate Natural Killer Cells
i. NK Cells release perforin, which oligomerizes to
form pores in the target cell membrane
1. The pores allow for the passive diffusion of
a family of pro-apoptotic proteases, known
as the granzymes, into the target cell
c. NK Cells not only kill viral infected cells. They also secrete type II interferon y, which promotes diverse anti-viral
responses, and can target stressed or cancerous cells that
have been bound by antibodies
viii. The Inflammatory Response
1. Inflammation is characterized by redness, swelling, heat and pain caused by:
a. Increased leakiness of blood vessels due to increased blood flow
b. Attraction of immune system cells to the site of infection due to localized release of cytokines
c. And production of additional soluble mediator of
inflammation
2. The response is initiated by tissue resident dendritic cells that identify pathogens via their Toll-like Receptors, and respond by releasing cytokines and chemokines – which attract other immune cells – critically neutrophils
a. Neutrophils are phagocytic, and directly engulf invading bacteria and fungi
BSC 300-001 Week 14 Notes
b. Neutrophils also release lysozymes and proteases that
digest bacterial cell walls and small peptides with
microbicidal activity called defensins that puncture
bacterial cell walls
3. Additional inflammation responses are mediated by local mast cells which secrete histamine, a small peptide that binds specific GPCRs promoting increased vascular permeability and therefore the migration of immune cells into the infection
4. If the bacterial burden in the infected tissue is high dendritic cells also migrate to lymph nodes where they present bacterial antigens to activate T-Cells that will mount an acquired immune response by activating B-Cells that secrete antibodies against the pathogens ix. Adaptive immunity – a vertebrate specific process
1. Acquired immunity imparted by lymphocytes bearing antigen specific receptors in their membranes
2. In 1905 von Behring (won the first Novel in physiology) and Kitasato showed that an antibiotic factor appeared in the serum of infected rodents and that this factor conferred resistance to other naïve rodents upon inoculation
3. This work also showed that serum contains a heat inactivated substance that “complements” the serum from exposed rodents.
Today, we know this as the complement system described earlier b. Immunoglobins: Structure and Function
i. Immunoglobins have a conserved structure consisting of heavy and light chains
1. Several classes (isotypes) of immunoglobins involved in adaptive response (IgM, IgA, IgE, IgD, and IgG)
2. Most abundant class (IgG) possesses two identical heavy chains covalently attached by disulfide bridges to two identical light
chains and one another: the H2L2 configuration
3. Each isotype is distinguished by possessing distinct heavy chains resulting from genetic recombination events in the gene encoding the heavy chain, a process called class switching
4. Each monomer can be divided into two functionally distinct regions, Fab – the antigen binding portion and Fc, the constant
portion that is identical for all antibodies of the same isotype
5. Each B-cell lineage produces a slightly different version of heavy and light chains in the Fab portion through another recombination process called V(D)J recombination
6. As a result of this recombination an almost limitless amount of antigen specificity is generated by the adaptive immune system
7. While there is a single heavy chain locus (gene) there are two distinct light chain loci called κ and λ
8. Each B-Cell transcribes only one of the two light chain genes, therefore the light chains are always identical in any given Ig
ii. Immunoglobin diversity
1. IgM is the principle B cell receptor, expressed in the membrane of naïve B cells
BSC 300-001 Week 14 Notes
a. IgM is also secreted as a pentamer of H2L2 chains joined by another protein, the J fragment
b. This arrangement greatly increases the affinity of interaction, known specifically as avidity: the combination of affinity and the number of binding interactions
c. The IgM conformation is highly conductive to activating the complement cascade
2. IgA is also bound by J fragment, forming an H2L2 dimer a. IgA binds an IgA receptor on the basolateral side of epithelial cells, where binding triggers receptor mediated endocytosis
b. Following this, the receptor is cleaved and the dimer is secreted to the apical side of the epithelium: a process called transcytosis
c. This plays an important part of the immune response to bacterial infection following tears and abrasions in epithelia 3. IgG is the major secreted antibody of B-Cells
a. It is important for neutralizing virus particles and for preparing antigens from viruses or bacteria for cells
equipped with receptors for the Fc portion of the antibody. Such receptors are called FcRs
b. Phagocytic cells (dendritic cells and macrophages) can recognize bacteria and virus coated with IgGs and
endocytose them. Such coating of bacteria by antibodies is called opsinization
c. Natural Killer cells can recognize and attack virally infected cells that express viral proteins in their plasma membraned bound to antibodies
4. Each naïve B cells produces a unique immunoglobin a. The clonal selection theory stipulates each naïve lymphocyte (having not bound its cognate antigen)
expresses an antigen binding receptor with unique binding specificity
b. Once it binds an antigen there is rapid cell division to generate a genetically identical lineage – a clone
c. Most antigens are complex 3D structures with a number of potential binding sites for antibodies.
i. Such binding sites are referred to as epitopes
d. Thus, when exposed to an antigen many naïve lymphocytes may be activated, each expressing a distinct antibody specific for a different region (epitope) of the antigen
e. Therefore, antibody production against an antigen is usually polyclonal – several, rather than just one, lineages are established
5. Specificity determined by the complementarity-determining region of each Ig
a. Over 1 million unique antibodies can be generated by naïve B-cells in vertebrates
BSC 300-001 Week 14 Notes
b. Each heavy chain is composed of four globular domains,
each light chain 2
c. The N-terminal domains of each chain is referred to as
variable regions VL and VH
i. Within each variable region reside three
hypervariable regions – HV1, HV2, and HV3 –
loops sandwiched between two B-sheets
d. Within the globular conformation of the variable domain,
these loops are in close proximity to one another, and
collectively the HV regions of the light and heavy chains
constitute the complementarity determining region
(CRD) of each antibody
c. Generation of Antibody Diversity and B-Cell Development
i. Generating antibody diversity
1. Naïve B and T cells both express cell surface receptors with high specificity for an epitope they have never encountered
2. If there are only 3 genes that encode the chains of antibodies, where does all of this diversity come from? Somatic gene
arrangement. The principles of which are similar for T-Cell
receptors as well as B Cell receptors and their soluble isotypes
3. Because of the almost infinity number of epitopes that can be recognized, there exists the potential for auto-immunity, so
organisms must have a way of distinguishing self from non-self
ii. The Cellular and Molecular Basis of Immunity:
1. DNA Rearrangement of Light Chain loci: V(D)J Recombination a. The single heavy chain and two light chain loci possess a
number of clustered gene segments that will encode the
distinct domains of the peptide produced
b. These segments are akin to exons, in that they will be
transcribed and spliced together after recombination occurs
c. The domains are the:
i. Variable gene segments
ii. Joiner gene segments
iii. Constant gene segments
iv. Diversity gene segments (Heavy chain only)
2. Focus on the kappa light chain
a. The C gene (constant) encodes the constant portion of the
protein and is located at the 3’ end of the locus. A single C
segment in the light chain loci, multiple segments in heavy
chain
b. The V locus (variable) contains multiple similar V genes –
40 in humans
c. The J locus (joining) contains a smaller number of
segments – 5 in humans
d. As B Cells are generated in the marrow the heavy and light
chain loci undergo recombination to produce one antibody
species with unique epitope specificity
BSC 300-001 Week 14 Notes
e. A protein complex composed of the Rag1 and Rag2 recombinases is activated and excises a random number of V and J segments. This is mediated by the complex
recognizing Recombination Signal Sequences at the 3’ end of each V segment and 5’ end of each J segment
f. The complex then joins a single V gene and J segment into one continuous exon
g. The RAG enzymes make single-stranded cuts, and the free -OH groups attack the complimentary strands, generating hairpin loops
i. RAGs open the hairpins, either symmetrically or
asymmetrically, leading to single-stranded
overhangs from asymmetric cleavage. A DNA
polymerase added random nucleotides to
symmetrically cleaved strands
h. As a result, random nucleotides are added to each junction – increasing the amino acid diversity in the encoded CDRs i. DNA ligase IV then joins the two strands generating a new light or heavy chain locus
j. Transcription initiates at the 3’ most V segment and terminates at the end of the single κ C gene
k. Non-joined J fragments are removed by splicing (as if they were an intron) to generate the mature mRNA
l. Such mature light chain mRNAs therefore contain coding sequence for one C and one VJ domain
3. The lambda light chain and heavy chain
a. The same V(D)J recombinase complex similarly performs recombination at these loci, with the added complexity of Diversity genes in the heavy chains – hence V(D)J
recombination
b. A single B Cell expresses only kappa or lambda light chains, not both
c. V(D)J recombination occurs only at one of the two alleles for these three loci. The second allele is transcriptionally silenced
4. Focus on the kappa light chain: V-J recombination a. Collectively these arrangements can produce approximately 2000 kappa chains (5 x 40) and an equivalent number of lambda chains
b. Heavy chain recombination is estimated to produce approximately 100,000 unique peptides
c. Therefore, over 200,000,000 unique antibody combinations are possible from heavy and kappa light chains alone
d. Junctional imprecision:
i. Of the three hypervariable loops in the peptides two are encoded within the V gene. The third falls at the
joint between the V gene segment and the J gene
BSC 300-001 Week 14 Notes
ii. Recombination at this site is imprecise, such that
the same V and J genes joined together in different
cells will have different amino acid sequences in
this loop due to random addition or loss of
nucleotides
5. Somatic Hypermutation
a. When a B cell recognizes an antigen, it is stimulated to
divide
b. During proliferation the V region undergoes somatic
mutation at a rate that is at least 105 to 106 greater than the
rest of the genome. In other words, somatic
hypermutation – increased single base pair substitution
c. Mechanisms:
i. Activation-induced cytosine deaminase: converts
C to U, leading to U:G mismatches and removal of
the uracil
ii. Translesion DNA polymerases: error-prone DNA
polymerases that fill in the spot where uracils were,
creating mutations that are propagated when the cell
divides
d. As a result of somatic hypermutation, there is increased
diversity in the hypervariable region of B cell antibodies
e. This can produce antibodies with even greater specificity
for the offending antigen. And these cells will be
preferentially selected following antigen re-introduction
6. Class Switching
a. Class switching – once a B cell is committed to producing
a specific antibody it can be induced to change the class of
antibody it produces
b. In response to T cell cytokines, during B cell maturation
other enzymes rearrange the C genes encoding the different
heavy chains
c. The VDJ region remains the same, but genes for class
specific heavy chains are moved proximal to the VDJ gene
II. Lecture 24 – Immunology Part II (Ch 21 continued)
a. The MHC and Antigen Presentation
i. The MHC (Major Histocompatibility Complex) encodes many proteins involved in immune response
ii. Class I and II MHC proteins are highly polymorphic and responsible for graft acceptance/rejection
iii. Class I and II MHC proteins display antigens on the cell surface and present them to antigen specific receptors on T cells – activating the T cell 1. Cytoxic T cells then produce cytokines and/or kill viral infected
cells
2. Helper T cells activate cognate B cells, stimulating them to mature
iv. Helper T Cells aide B Cell maturation
v. Both naïve B cells and T cells have receptors that allow them to interact with specific epitopes – a similar mechanism to V(D)J recombination
BSC 300-001 Week 14 Notes
generates epitope specific receptors on the surface o cytoxic and helper T cells
vi. While naïve B cells can directly interact with antigens via their IgM receptors, the antigen-specific receptors of T cells can only recognize epitopes that are associated with specific glycoprotein complexes on the surface of antigen presenting cells
vii. The various antigen presenting cells that digest foreign pathogens present portions of proteins from the pathogens associated with transmembrane MHC proteins (class I and II)
viii. The genes encoding these glycoprotein complex proteins are contained within a large genomic region called the Major Histocompatibility Complex (MHC)
ix. The MHC locus determines the ability of members of the same species to accept tissue grafts
1. Success of graft transplants is determined by genes encoded in the MHC
2. The human MHC is called the HLA complex (human leukocyte antigen) – which contains dozens of genes, most encoding proteins with immunological relevance
3. Most cells in vertebrates express proteins from the MHC and thus have the potential for presenting antigen peptides to the immune system
4. Class I MHC proteins are single pass transmembrane
glycoproteins, non-covalently associated with a non-MHC encoded protein called B2-macroglobin
5. Class II MHC proteins are heterodimeric, each a single pass protein
x. The killing activity of Cytoxic T cells is Antigen-specific and MHC restricted
1. MHC proteins play a critical role in recognition of viral infected cells by cytoxic T cells – aka Cytolytic L lymphocytes (CTLs), or Killer T Cells, which following identification of infected cells promote lysis of the infected cell
2. The experimental assay depicted here simply shows that CTLs from influenza infected mice are able to recognize and promote lysis of target cells infected with the same strain of influenza (right tubes), but not genetically identical cells that are not virally
infected
3. Importantly, killing activity is both restricted to cell infected with the exact same strain of flu. Even a single amino acid change in flu proteins (virus X vs virus Y) prevents CTLs from the host rat from recognizing and destroying virally infected cells
a. Additionally, this killing activity is restricted to rate of the same genotype – virally infected cells from rats that
genotypically differ from the host rate cannot be recognized
and destroyed by its CTLs
b. This phenomenon is referred to as MHC restriction and the MHC molecule involved is called restriction elements
BSC 300-001 Week 14 Notes
xi. T cells with different functional properties are guided by two distinct classes of MHC molecules
1. The MHC encodes two types of glycoproteins essential for immune recognition: MHC class I and class II molecules – there are multiple genes for each class
2. In addition the MHC encodes proteins involved in antigen processing, presentation, and proteolysis
3. Also, the vertebrate MHC encodes components of the complement cascade
4. Class I and class II MHC molecules are both involved in presenting antigens to T cells, but serve two distinct functions: a. Class I: Present antigens to CTLs (Cytotoxic T Cells)
triggering them to destroy virally infected cells. CTLs
express a cell surface glycoprotein called CD8 which
determines the ability of a T cell to interact with MHC
Class I proteins. Most nucleated cells can support viral
replication, and therefore all express MHC class I proteins
b. Class II: Are found exclusively on profession antigen
presenting cells (B cells dendritic cells and
macrophages), which present antigens to Helper T cells
and initiate the cascade on interactions that will stimulate
naïve B cells to differentiate as active plasma cells and give
rise to Memory B cells. Helper T cells express the
glycoprotein CD4 which is required for interaction with
Class II MHC proteins
c. Like B Cell receptors and their isotypes, T Cell receptors, CD4 and CD8 belong to the immunoglobin superfamily
5. Important Concept – B cell activation involves rapid proliferation in the lymph system. A subset of B cells undergo class switching to soluble antibody secreting cells. Such activated B cells are referred to as plasma cells. In addition, a subset of these cells undergo somatic hypermutation and remain in the lymph nodes and spleen as memory B cells, which can be reactivated more rapidly if the body is exposed to the same antigen in the future
xii. MHC molecules bind peptide antigens and interact with the T-Cell receptor
1. The source of graft rejection is that both Class I and Class II MHC molecules are highly polymorphic: there are many alleles of all members of each class throughout the human population – more
than 2000 alleles combined for all of the Class I and II genes a. As a result, except for closest relatives, the chances that any two individuals share the same MHJC variants is very
small
b. Regardless of this variation, all members of both Class I and II MHC protein share a highly similar conformation as
well as interaction mechanism with the T Cell receptors
BSC 300-001 Week 14 Notes
i. This is why transplant surgeons must MHC match potential donors with patients in need of a
tissue/organ transplant
2. Class I MHC Molecules
a. There are 3 MHC Class I genes (HLA-A, HLA-B, and HLA-C), and the encoded single pass Ig glycoproteins non covalently interact with the non-MHC encoded B2-
macroglobulin, which has a conformation highly similar to an Ig domain
b. Two Ig domains form the peptide binding cleft
c. Antigen peptides bound by Class I molecules are 8-10 residues long, and there are binding pockets in the MHC protein for the charged N and C termini of the peptide
d. Affinity for the peptides is enhanced by a small number of binding pockets that accommodate amino acid side chains of each peptide
e. In this manner, a given MHC protein can accommodate a large number of peptides of diverse sequence
f. Most allelic diversity among MHC protein lies in amino acids in and around the peptide binding cleft, which
therefore alters specificity of peptide binding between variants
i. As well as generates distinct conformations of the MCH protein itself. A T Cell which can interact
with one Class I MHC allele will therefore not be
able to physically interact with another – this is the
basis of MHC restriction
g. The CD8 protein on cytoxic T Cells (CTLs) serves as a co receptor binding to conserved portions of Class I MHC proteins
i. The CD8 protein, thus sets the Class I preference of all mature T cells
3. Class II MHC Molecules
a. Six human genes
b. Encoded protein similar structure to Class I: two Ig domains near the membrane. Both proteins of the
heterodimer (alpha and beta) contribute to the peptide binding cleft, which is therefore larger and can
accommodate larger peptides than Class I
c. Similar to Class I polymorphisms affect architecture of binding cleft and therefore peptide affinity and ability to interact with T-Cell receptors (MHC restriction) and the co receptor CD4
i. CD4 is the protein through which HIV virus gains entry into T cells to initiate immunodeficiency
associated with AIDs
d. Binding of peptides to MHC Class II takes place in endosomes and lysosomes
BSC 300-001 Week 14 Notes
i. MHC Class II proteins are targeted to these
organelles by a chaperon called the invariant chain
xiii. Antigen processing and presentation
1. Class I is involved in presenting antigens produced by the cell itself, including pathogen-associated proteins
2. Class II is involved in presenting antigens from endocytosed pathogens
3. General steps to processing and presentation:
a. Acquisition and targeting of antigens
i. Class I: relies on error-prone translation that results
in peptides destined for ubiquitin-mediated
proteasomal degradation to produce peptide
fragments for presentation
1. Such peptides are delivered to the ER by the
TAP peptide transporter (an ABC
transporter)
a. The TAP complex has affinity for
peptides terminating in Leu, Val, Ile
and Met), which matches the binding
preference of the MHC proteins
ii. Class II: relies on phagocytosis, pinocytosis,
receptor mediate endocytosis followed by
lysosomal degradation, which mature into late
endosomes
1. In naïve B cells the IgM receptors initiate
receptor mediated endocytosis
2. Professional phagocytes (antigen presenting
cells) possess Fc receptors (FcRs) that
recognize opsonized bacteria initiating
phagocytosis, degradation, and presentation
at the cell surface
3. It is not only peptides that can be presented
at the cell surface – a Class I like MHC
protein called CD2 presents lipid antigens
b. Delivery and binding of peptides to MHC proteins:
i. Class I: In the ER newly synthesized MHC proteins
are part of a complex called the peptide-loading
complex, that includes the chaperones calnexin and
calreticulin, which promote peptide association.
Conformational change after loading releases the
MHC and bound peptide from the loading complex
ii. Class II: MHC proteins are produced in the ER and
assemble with the invariant chain, which provides
the targeting signal for delivery to late endosomes.
In the lysosomes peptides are trimmed in length by
proteases which also remove the invariant chain
from the MHC proteins. A distinct chaperone called
DM helps load the peptides onto Class II proteins
BSC 300-001 Week 14 Notes
c. Presentation of MHC/peptide complexes
i. Class I: complexes are transported from ER via
vesicles through Golgi and to the plasma membrane
ii. Class II: Complexes are mainly found within late
endosomal multivesicular bodies. These are
recruited to the endosomal membrane and bud off
as tubular endosomes that are transported to the
plasma membrane on microtubules
4. Naïve B Cells bind soluble antigen by their IgM B Cell receptors 5. Complex is endocytosed, degraded, and peptide fragments are presented on cell surface in association with MHC Class II proteins a. This step, as we will see, is necessary for the ultimate
activation of B cells
b. T-Cells, T-Cell Receptors, and T-Cell Development
i. T Cell receptors are structurally related to the F(ab) portion of B Cell Ig isotypes
ii. Genes encoding these receptors undergo almost identical genomic rearrangements to V(D)J somatic recombination of the Ig Heavy and light chains – the details of which we will not review
iii. Unlike B Cell Ig’s T Cell receptors do not undergo somatic hypermutation during differentiation
iv. In the course of their development T Cells must learn to recognize self derived from non-self-antigens to avoid an autoimmune response
v. T Cell Receptor Structure
1. Like B Cells, T Cells that have been activated through antigen binding on their R Cell Receptors also undergo clonal selection
and proliferation
2. They then acquire the ability to kill antigen-bearing target cells (as cytoxic T cells, CTLs) or they secrete cytokines that will assist B Cells in their differentiation (Helper T Cells)
3. A T Cell Receptor is a heterodimer compose of two glycoproteins, each of which have undergone somatic rearrangement. Each
heterodimer is composed of either an alpha or beta or y subunit
with the N termini of the two subunits forming the antigen binding site – as with B cell Ig’s, this is the region where somatic
recombination generates diversity
4. Most T cell receptors contain one subunit that was the result of VDJ recombination and another the result of VJ recombination – in this respect they are highly similar to B Cell Ig’s
vi. T Cell and B Cell Proliferation and Differentiation
1. Antigen binding to TCR’s and BCR’s to naïve T and B Cells
triggers their immune response
2. The antigens for TCRs are the peptide bound MHC proteins on the surface of APCs
3. Antigens for BCRs do not need to be associated with antigen presenting cells
4. Several auxiliary proteins function in collaboration with the TCR’s and BCR’s to initiate a signal transduction event
BSC 300-001 Week 14 Notes
5. The cytoplasmic portion of the receptors themselves are too short to interact with cytosolic proteins and initiate transduction
6. Instead auxiliary proteins with cytosolic ITAM domains (immunoreceptor tyrosine-based activation motif) associate with TCR’s and BCR’s
7. Antigen binding by the receptors alters their conformation and recruits members of the Src family of tyrosine kinases, which phosphorylate the ITAMs on specific tyrosines
8. Phosphorylated ITAMs recruit additional non-Src kinases via SH2 domains, which in turn recruit and phosphorylate adapter proteins that serve as scaffolds to recruit proteins and initiate signal
transduction
9. Signal transduction includes activation of phospholipase C-y and PI3-Kinase – which generate IP3 and Protein Kinase B, the major effectors of signal transduction in B and T cell activation
10. T Cells depend on production of the cytokine Interleukin 2 (IL-2) for clonal expansion. It is one of the first genes to be transcribed following antigen binding to TCRs. I:-2 stimulates the T Cell to produce more IL-2 (autocrine signaling) in order to sustain
proliferation
11. The transcription factor that transcribes IL-2 is named NF-AT (nuclear factor of activated T Cells), and is always expressed but sequestered in the cytoplasm in a phosphorylated state
12. The phosphatase that removes this inhibitory phosphorylation is calcineurin, a Ca2+-dependent enzyme
13. IP3 release by PLCy stimulates Ca2+ efflux from the ER, thus activating calcineurin, and as a result promoting NF-AT entry into the nucleus
14. Human Health relevance – how can organs from unrelated donors be successfully transplanted? Immunosuppressants, like cyclosporin, bind and complex with calcineurin – inhibiting its ability to dephosphorylate NF-AT. As a result T cells cannot undergo clonal expansion, and therefore the tissue is not seen as being foreign
vii. Comparison of T and B Cell Development
1. Both are born in the bone marrow. Pre-T cells migrate to the thymus, while pre-B cells remain in the marrow
2. Before becoming mature both experience life as a pre-lymphocyte 3. The membrane bound receptors are not yet capable of binding ligands, but instead the B cell IgM heavy chain and T cell Beta or y subunit associate with different proteins and form pre BCRs and TCRs – the function of which is to transduce signals via distinct ITAM proteins that shut down RAG mediated recombination, promote proliferation and promote transcription of the missing subunit (K: κ or λ light chain or T:α or δ).
4. The receptor and lymphocyte are now considered to be mature, but still naïve
viii. T Cell Differentiation
BSC 300-001 Week 14 Notes
1. In the thymus naïve T Cells are presented with antigens both foreign and derived from self
2. T Cells that cannot interact with antigens, and T cells that react with self-derived antigens and killed by extrinsic apoptosis
3. A protein called AIRE (autoimmune regulator) allows the expression of tissue specific proteins (like those of the pancrease) in the thymus and the screening of T cells with auto-affinity
4. Defects in AIRE lead to a wide array of autoimmune disorders 5. Pre-T Cells express both CD4 and CD8, while differentiated T Cells express only one of the cofactors
6. The mechanism appears to be stochastic and determined by a battle between two transcription factors, both of which are expressed in pre-T Cells, but only one remains active in mature T cells
7. ThPOK promotes the CD4 lineage
8. Runx3 promotes the CD8 lineage
9. Both are regulated by pre-TCRs and appear to fight for supremacy, suppressing one another’s expression until there is only one
ix. T Cells Require Two Signals for Activation
1. All T Cells require antigen binding
2. The second co-stimulatory signal is mediated via the receptor CD28, expressed on all T cells, which is bound by other CD protein on the surface of professional APCs
3. Upon activation, T Cells promote attenuation of activation by expressing an additional protein, CLTA 24 that competes with CD28 for these APC originating signals
4. In this manner, the T cell is able to fully commit to proliferation and activation
x. Cytoxic T Cells
1. Once bound to virally infected cells through their TCRs and co receptors CD8 cytoxic T Cells (like Natural Killer Cells of the Innate Immune System) release into the narrow clefts between the cells the combination of perforin and granzymes
a. Perforin punctures the membrane of the infected cell while granzymes activate caspases that promote apoptosis
xi. Interleukins
1. Lymphocytes and other cells in the lymph produce an array of cytokines that control lymphocyte activity – primarily to
proliferate or differentiate
2. Cytokines that act on leukocytes are called interleukins – of which there are approximately 35 encoded by the human genome. Each bound by distinct receptors in the array of leukocyte cell types. Interleukins may act to alter the fate of cells, or as chemokines – attracting leukocytes to a site of infection
3. These signals act through cytokine receptor activating, principally the JAK/STAT pathway
a. Ex: most B Cells bound by an antigen require direct contact with a helper T cell bound by the same antigen in order to
be activated and to initiate proliferation and class
BSC 300-001 Week 14 Notes
switching. This signal is mediate via Interleukin 4 (IL-4)
secreted by the helper T cells
4. Additionally, when tissue damage occurs resident fibroblasts secrete IL-8, which attracts neutrophils to the site of infection
c. Collaboration of Immune-System Cells in the Adaptive Response i. A functional immune system requires constant interplay between the various cells of the immune system. Resident APCs must be able to activate T Cells as either cytoxic T Cell or helper T cells, and helper T cells must be able to activate B Cell in order for antibodies to be produces ii. Langerhans cells represent one of the first line of defenses of the innate immune system – they constitute a network of dendritic cells within the skin that makes it virtually impossible for invading bacteria to not trip the alarm
iii. These and other professional APCs primarily conduct their screening of the environment via the action of Toll-like Receptors, discussed briefly in the previous lecture
iv. Production of High Affinity Antibodies Requires Collaboration Between B and T cells
1. Activation of naïve B cells requires both a local source of antigen as well as a helper T Cell already activated by the same antigen
2. Soluble antigen reaches the lymph nodes where B and T Cells reside, either as a result of bacterial lysis through the various
mechanisms of the innate immune system, or through presentation by professional APCs that have migrated into the lymph system
after engulfing and digesting a pathogen
3. Dendritic cells present the antigen to CD4+ T Cells which mature into helper T cells
4. Soluble antigens are bound by the BCRs of naïve B Cells
a. These B Cells then internalize the receptor and antigen,
process the antigen via proteolytic degradation and present
it in their own surface coupled to Class II MHC proteins
b. Activated helper T Cells interact with antigen presenting B
Cells via interaction between the TCR and peptide bound
MHC proteins on the B cell
c. The B cell also displays receptors for a number of cytokine
and membrane bound signals from the helper T cell
d. And a second required interaction for maturation mediated
by the B cell receptor CD40 and the T Cell membrane
bound ligand CD40L
e. Both IL-4 and CD40 binding are required for B Cell
activation
f. Only then will B Cells proliferate
5. A subpopulation of these cells undergo class switching and
become soluble antibody secreting plasma cells – these cells are short lived – a few days – and are responsible for the immediate immune response of the adaptive system
6. A second subpopulation of B-cells do not undergo class, but instead experience somatic hypermutation and exit the cell cycle.
BSC 300-001 Week 14 Notes
These quiescent cells remain in the lymph nodes and spleen as memory B cells that can mound a faster and, often more time efficient, attack against future invasion by the same pathogen
7. Similarly, helper T cells proliferate and differentiate as either activated T cells that aide the antigen exposed B cells, and
additionally a subpop remains quiescent in the lymph system as Memory T Cells (no somatic hypermutation
v. Vaccines elicit protective immunity against a variety of pathogens 1. Inoculation with compromised virus activates the immune system and raises antibodies against viral protein
2. Viral inactivation can result from heating and denaturing the virus, but this is not as effective as attenuation, in which the virus is passaged through successive rounds of cell culture. Along the way it becomes less potent (reason is unsure, but likely related to mutation)
3. Inoculation with live attenuated strains results in activation of the immune system and mild reaction caused by inflammation and weak proliferation of the virus
4. Additional methods of vaccination include subunit vaccination, in which only a single protein, rather than the entire virus, is
delivered
vi. The immune system also defends against cancer
1. The elevated mutation rate of cancer – next lecture – not only produces driver mutations that cause cancer, but also passenger mutations that do not contribute to cancer itself
2. Rather passenger mutations may slightly alter the amino acid sequence of membrane proteins – generating neo-antigens – and allow the immune system to recognize the wayward cells and eliminate them
3. Additionally, genetic mutation associated with cancer may lead to the expression of fetal-specific genes whose protein products were expressed prior to the development of the immune system. These, likewise, can be recognized as abnormal, and their carrier cells eliminated by the immune system
BSC 300-001 Week 15 Notes
I. Lecture 25 – Cancer
a. Intro
i. Cancer cells proliferate, invade, and metastasize
1. One in 5 people will die from cancer: an aggressive,
hyperproliferative, malignancy of rogue cells that, through
uncontrolled growth and hypermutability, wage an evolutionary
arms race against one another and the healthy tissue they infest
a. Metastasis and secondary tumors are what are killers, not
primary tumors
2. Inherited and acquired mutations allow cancerous cells to:
a. Sustain proliferative signaling
i. Either through uncontrolled production of mitogens
or uncontrolled activation of their pathways
b. Evade growth suppressors
i. Often by disabling mutations in the suppressor’s
receptors or the pathways they regulate
c. Activate invasiveness and metastasis
i. Allows cancer cells to leave a primary tumor and
establish secondary tumors
d. Enable replicative immortality
i. Continue to proliferate past the number of
generations when normal cells senescence and die
e. Induce angiogenesis
i. Stimulate growth of blood vessels to supply
nutrients and oxygen to the tumor
f. Resist cell death
i. Inactivate the apoptotic pathway and evade the
immune system
3. Cancer, like many diseases, results from an interplay between an
individual’s genotype and environmental influences, often referred
to as gene-by-environment interactions; i.e., some people are more
susceptible to cancer promoting influences due to differences in
our genetic makeup
a. Most cancer arises from combined genetic mutations that
result from exposure to carcinogens, chemicals, and/or
other environmental factors (UV light and radiation) that
perturb the DNA replication process, as well as the inherent
random error of DNA replication
b. Some people also inherit genetic mutations that increase
their chances of developing cancer
c. Importantly, cancer causing mutations work through the
combinatorial action of mutated genes. No single mutation
is cancer-causing
d. This usually takes many years, therefore most cancer is a
disease associated with advanced age. However, some
mutations are more oncogenic than others and can lead to
development much earlier in life
BSC 300-001 Week 15 Notes
i. 90% of all cancer is from epithelial cells because
they are in a constant state of division
e. Because most cancer requires multiple mutations, and mutation only takes place in proliferating populations of cells, those cells with higher rates of proliferation (epithelia like skin, lung, esophagus, colon) are the most common sites of cancer formation
f. Cancer is usually classified based on the embryonic tissue of origin:
i. Carcinomas – derived from epithelia (endodermal and ectodermal) and are the most common type of
malignant tumors (greater than 90%)
ii. Sarcomas – derived from mesoderm (muscle, bone, connective tissue, and blood)
iii. Both carcinomas and most sarcomas are solid mass tumors
iv. Leukemias – cancers of individual types of white blood cells
4. Mutations in three broad classes of genes are implicated in the onset of cancer:
a. Protooncogenes
i. Their encoded proteins normally control the rate of cell proliferation. Gain of function mutations
convert protooncogenes into oncogenes
1. Because of this, mutation in only one copy
of a protooncogene may convert into an
oncogene (just a gamma mutation in mom or
dad to convert)
b. Tumor Suppressor Genes
i. Their encoded proteins normally restrain growth
and function as regulators of the cell cycle. Loss of
function mutations release these regulatory controls
and allow cells to proliferate even following
without appropriate checks on cell cycle
progression or DNA damage
1. Mutations in both copies of a tumor
suppressor gene are necessary to exert an
oncogenic effect (both mom and dad)
c. Genome maintenance genes
i. Maintain stability and integrity of the genome and prevent excessive mutation from occurring
1. As with tumor suppressor genes, loss of
function mutation in both copies of such
genes are required for full oncogenic effect
2. Hypermutation – increased mutation rate
when affected
5. Cancer is a clonal process. A single cell acquires mutations that allow it to begin proliferating outside of the controls of normal
BSC 300-001 Week 15 Notes
cells. If it evades the immune system, it can establish a tumor, a benign pool of precancerous cells. Such cells acquire additional mutations and establish subclonal populations – some better, some worse – and growth and evasion
a. Eventually, a subpop will acquire mutations that allow
metastasis: cells exiting the primary tumor and invading
other tissues and organs
b. Most cancer deaths are due to metastasis and the formation of secondary tumors that spread throughout the body
ii. Genetic makeup of most cancer cells is dramatically altered 1. Point mutations are not the only, and usually not the most, important drivers of cancer progression – tumors harbor all types of genetic alterations, including:
a. Point mutations
b. Small and large deletions
c. Amplifications
d. Insertions
e. Translocations and inversions – movement of portion of one chromo into another chromo or flipping a chromo left
or right
f. Aberrant number of chromosomes, usually too many
(aneuploidy)
g. Mistakes in DNA replication, through loss of tumor
suppressors activity, increases replication error and
mutation number
2. Other mechanisms of increased mutation rate:
a. Kataegis (thunderstorm) – the broad-scale activation and deployment of the enzymes that promote somatic
hypermutation in B cell Ig domains, like activation induced deaminase
b. Chromothripis (chromosome shattering) – in some
aberrant cells following mitosis, some chromo are not re
integrated into nucleus, but form their own micronuclei,
where replication and segregation is much less efficient.
Rampant chromo breakage and random stitching together
or the pieces produces dozens to hundreds of chromo
fragments in odd combos
iii. Cancers have highly abnormal karyotypes
1. Chromo of most cancer cells are dramatically altered. In addition to point mutations in individual genes:
a. Individual chromo number is altered (aneuploidy)
b. Chromo rearrangement (Composite chromo)
2. Normal karyotype vs SW403 colorectal adenocarcinoma cell line chromo: analysis of the number, type and shape of the entire complement of mitotic chromo of a eukaryotic cell
iv. Housekeeping functions are fundamentally flawed
1. Cancer cells are usually less differentiated than and visually distinct from normal cells – high nucleus to cytoplasm ratio,
BSC 300-001 Week 15 Notes
prominent nucleoli, increased percentage of mitotic cells and little differentiated structures
2. The increase in gene copy number, due to chromo and regional duplications, leads to higher protein production that alters cell physiology. This activates the stress response pathway – proteins of which are principle targets of many cancer drugs: i.e., if you block the stress response pathway of cells with high protein
production it will induce apoptosis
v. Energy production in cancer cells by aerobic glycolysis
1. Cancer cells, even in the presence of oxygen preferentially generate ATP via glycolysis: The Warburg effect: a 200-fold increase in ATP production from glycolysis compared to normal cells
a. This is how PET scans can identify tumors following
consumption of radioactively labeled glucose
2. The glycolytic metabolites can be used by cancer cells for macromolecules biosynthesis
3. Many cancers, like proliferating embryonic cells, abnormally express the embryonic isoform of pyruvate kinase (PKM2). It has a high Km, therefore does not turn over pyruvate quickly. Cell is locked in glycolysis
4. Additionally, PKM2 promotes expression of a key transcription factor that maintains the pluripotency and high proliferation rate of embryonic stem cells, and thus converts adult cells into an
embryonic stem cell-like state
vi. Hydroxyglutarate is a cancer-specific metabolite
1. Some cancer types produce novel metabolites
a. In normal cells the TCA cycle enzyme isocitrate
dehydrogenase converts isocitrate into alpha-ketoglutarate
b. Mutant IDH forms (found in many brain cancers):
i. Convert isocitrate into 2-hydroxyglutarate (SHG)
ii. High 2HG inhibits proteins that require alpha
ketoglutarate for function, including enzymes that
control the methylation state of histone proteins:
1. TET family of DNA hydroxylases
2. Histone methyl transferases
3. Histone demethylases
iii. Histone methylation is part of the epigenetic code
that generates heterochromatin and shuts down gene
expression. So, the absence of this activity open
regions of chromosomes up for gene expression
vii. Uncontrolled proliferation is a hallmark of cancer
1. In addition to the uncoupling of controls of the cell cycle and lack of dependence on growth signals, cancer cells must also evade the exit to senescence
2. Chromosomes ends are protected by regions called telomeres, repeated non-coding sequences of DNA that are shortened each time DNA is replicated
BSC 300-001 Week 15 Notes
3. Normal cells can divide only about 50 times before the telomeres are consumed and replication begins to erase coding portions of the genome – such cells become senescent
4. During gamete formation telomere are lengthened by the action of a germline-specific enzyme called telomerase, a reverse
transcriptase that contains an RNA template that adds the repeated TTAGGG sequences
5. Most cancer cells are immortalized by mutations that activate telomerase expression, thus preventing telomere shortening and allowing such cell lines to live forever
viii. Cancer Cells escape the confines of tissues
1. Contact inhibition – normal somatic cells grown in culture arrest as a monolayer when they make contact with other cells (anti mitogens are expressed upon crowding). This is true of cells in vivo as well
2. Anchorage dependency – normal somatic cells require contact with other cells or basement membrane (via cadherins and
integrins). Lack of such contact promotes apoptosis
ix. Basic Properties of a Cancer Cell
1. Malignant cells are not responsive to the influences that cause normal cells to stop growth and division
2. Malignant cells do not display:
a. Contact inhibition
b. Anchorage dependency
c. Have lost the requirement for stimulatory growth
factors
3. And are said to have undergone oncogenic transformation x. Tumors are heterogeneous organs that are sculpted by their environment 1. Mutation of the initial cells within a tumor leads to a genetically heterogeneous population
2. Not all cells of a metastatic tumor are capable of metastasis; such cells are called cancer stem cells and have acquired stem cell-like properties. This source of cells may be:
a. A mutated normal tissue multipotent stem cell
b. Dedifferentiation of terminally differentiated cells
3. Like stem cell niches, the tumor microenvironment also contributes to the heterogeneity of tumor cells, where neighboring cells may be more, or less, conducing to tumor growth than others
4. CD8+ Cytoxic T Cells and Natural Killer Cells surround tumors and migrate into them. Part of the evolutionary arms race is cancer acquiring the ability to evade such detection
5. Additionally, our own immune system may help promote the onset of cancer. Chronic inflammation promotes the immune system to produce cytokines, including growth factors, that may help
increase rates of tumor cell proliferation
xi. Tumor growth requires blood vessel formation
1. Without a blood supply tumors cannot grow larger than 2mm
BSC 300-001 Week 15 Notes
2. Most tumors induce angiogenesis by acquiring expression of key signaling molecules, specifically:
a. Basis fibroblast growth factor (Beta-FGF)
b. Transforming growth factor (TGF)
c. Vascular endothelial growth factor (VEGF)
xii. Invasion and metastasis are late stages of tumorigenesis
1. Following the mutations that allow release from anchorage
dependency, epithelial tumor cells must loose association with one another to allow metastasis to occur
2. During normal development, a phenomenon called the epithelial to mesenchymal transition (EMT) plays an important role in
allowing cells to move and migrate in the embryo
3. Some tumor cells mimic this process, turning off the cadherins that promote their epithelial associations, activating expression of genes involved in migration as well as expression of basement
membrane digesting enzymes, like matrix metalloproteinases…
4. Two developmentally critical transcription factors appear to be essential for developmental and tumor EMT, named snail and
twist, which regulate the various genes promoting these processes b. The Origins and Development of Cancer
i. Carcinogens induce cancer by damaging DNA
1. Chemical carcinogens, aka mutagens, can be classified into two broad categories:
a. Direct-acting carcinogens: Seek out and react with
electron rich centers in other molecules. By chemically
interacting with N and O in DNA these compounds can
modify bases resulting in incorrect incorporation of
nucleotides during subsequent replication. Relatively few
of these compounds – including ethylmathane sulfonate
(EMS) and nitrogen mustards
b. Indirect-acting carcinogens: Most chemical carcinogens
have little effect until they have been modified by cellular
enzymes. Such mutagens are water insoluble and to be
cleared from our cells must be made soluble through the
addition of an electrophilic center – usually an -OH group
added by the P-450 family of enzymes. While this process
solubilizes the molecules for elimination, it can also
convert harmless compounds into carcinogens
i. Basically makes them active by making them
soluble
2. Chemical carcinogenesis by tobacco smoke
a. Benzo(a)pyrene was shown in 1933 to be a principle
carcinogen found in coal tar
b. Later this chemical was shown to be a principle agent of
carcinogen in cigarette smoke – it’s an indirect acting
carcinogen modified by P450 to produce a byproduct that
reacts with DNA – primarily at the N2 atom of guanines
BSC 300-001 Week 15 Notes
c. The conversion results in the replacement of G with T in the next round of DNA replication – a transversion
mutation in which a primidine is replaced with a purine
d. A principle gene randomly hit by this process is p53 – the major tumor suppressor involved in DNA repair – such
mutations are found in about 1/3 of lung cancers
e. Other known carcinogens include:
i. Asbestos – mesothelioma
ii. Aflotixin – a fungal metabolite associated with liver
cancer
iii. Heterocyclic amines – colon and breast cancer
(found in the char or well-done meat)
ii. A multi-hit model explains cancer progression
1. Cancer is principally a disease associated with age because it takes time and random change for a correct combination of mutations to drive tumor formation and metastasis
2. This model is supported by lab experiments in which mice are transgenically engineered to carry combinations of specific
mutations found in many cancer types
3. For example gain of function mutations in the Ras G-protein prevent it from hydrolyzing bound GTP, and promote increased cell proliferation
4. Similarly, gain of function mutations in the MYC transcription factor, required for transition from G1 to S phase, increase cell proliferation rates
5. In transgenic mice with either mutation expressed in mammary tissue, less than 50% develop aging associated cancer
6. However, when these mutations are combined, by crossing the mice, the offspring have a much higher incidence of cancer and it is more aggressive and malignant
iii. Successive oncogenic mutations can be traced in colon cancer 1. CRC is most often a cancer found in elderly patients and highlights the multi-hit hypothesis of cancer progression, with easily
observable morphopological progression of precancerous to cancerous growths
2. Polyps, benign adenomas and malignant carcinomas can be surgically removed and genotyped to identify mutations associated with each stage of progression
3. Excessive polyp generation can produce many benign growths each with the potential to develop into cancer and is found in families with familial adenomatous polyposis (FAP), a
predisposition for colon polyp formation
4. Families with FAP share loss of function mutations in Wnt signaling pathways, usually in the regulatory protein
Adenomatous Polyposis Coli – a key inhibitor of Wnt signaling 5. Random mutation in the second copy of APC promotes formation of benign polyps
BSC 300-001 Week 15 Notes
6. Subsequent mutations in other driver genes promotes progression from polyp to adenoma to malignant tumor, including:
a. Activating mutations in one of the three human Ras genes,
followed by loss of p53
b. DNA from human colon carcinomas generally show
mutations in all three: APC, Ras and p53 that are the
principle drivers of tumor formation
c. The loss of p53 function then promotes increased mutation
rates that hit other drivers of metastasis and invasion
c. The Genetic Basis of Cancer
i. Virtually all human tumors have inactivating mutations in cell cycle check point genes – these are the principle tumor suppressor genes
ii. Likewise, virtually all cancers have gain of function mutations in genes that promote cell proliferation
iii. Activating mutations in proto-oncogenes
1. Proto-oncogenes are usually involved in one of the many signal transduction pathways that promote cell proliferation. They may encode signaling molecules, receptors or components of the
pathways that lead to activating proliferation
2. The gain of function mutations overrides the build-in control mechanisms of regulated expression or activation of some
component of the pathway
3. Gain of function mutations are genetically dominant, meaning it takes only one mutation at one locus, rather than the homologous loci of both chromosomes, to exert an effect
4. Three general types of gain of function mutations convert proto oncogenes to oncogenes:
a. Point mutations: that alter the activity of the encoded
protein
i. Ex: mutation that inhibits the GTPase activity of
Ras prevent it from turning itself off. Always in
glycine 12
b. Gene amplification: multiple copies of a gene lead to
excessive production of a component of the pathway –
results from inaccurate DNA replication
c. Chromosome translocation:
i. Places the proto-oncogene under control of different
regulatory DNA sequences leading to constitutive
expression
1. Ex: a signaling molecule is no longer
regulated, but always expressed. Results
from chromosomal breaking and improper
repair
ii. Or fuses two genes together to produce a chimeric
protein with constitutive activity
iv. Cancer Causing viruses contain oncogenes or activate proto-oncogenes
BSC 300-001 Week 15 Notes
1. In 1911 Peyton Rous showed that certain viruses could cause cancer when injected into suitable vertebrate hosts (birds and some rodents, but fortunately not humans)
2. The virus called Rous Sarcoma Virus is a retrovirus: ssRNA genome that is reverse transcribed and integrated into the genome. It possesses one gene critical for tumorigenicity, an oncogene that was named v-src (viral sarcoma)
3. In the 1970’s it was discovered that vertebrates possess a proto oncogene with a high degree of similarity to v-src, and it was named c-src (cellular)
4. Current hypothesis is that an ancient virus took a copy of the c-src gene with it after infection and new virus production
5. RSV is an acute retrovirus in that it can induce tumor formation within days of infection
6. Other slow-acting retroviruses lack oncogenes, but rather integrate into host genomes near proto-oncogenes and a portion of the viral genome acts as an enhancer to increase expression of specific proto-oncogenes
v. Loss-of-function mutations in tumor suppressor genes
1. Tumor suppressor genes normally function to regulate the cell cycle and halt its progression if cell cycle checkpoints identify defects (DNA damage, failure to produce chromosome
biorientation, etc)
2. Intracellular proteins that regulate or inhibit entry into the cell cycle – for example retinoblastoma (pRb)
3. Receptors or signal transducers for secreted hormones/signals that inhibit cell proliferation
a. Ex: TGTF-Beta
4. Checkpoint pathway proteins that arrest the cell cycle following DNA damage
a. Ex: p53
5. Proteins that promote apoptosis
6. Enzymes that participate in DNA repair
7. In general, tumor suppressor mutations are loss of function and require inactivating mutations in both copies of the affected gene a. The mutations are said to behave recessively
8. In some cases, however, reduction to half the normal amount of protein is sufficient to produce an effect
a. Such genes are said to be haplo-insufficient
9. Some people inherit a loss of function copy of specific tumor suppressors, which predisposes then to develop cancer, since it would only take one random somatic mutation in any dividing cell to completely shut off that gene
a. Ex: children with hereditary retinoblastoma inherit one non-functional copy of the RB gene. Such children develop retinal tumors early in life, generally in both eyes
10. Mutations to the RB gene to not only lead to retinal cancer
BSC 300-001 Week 15 Notes
a. pRB blocks the function of the S phase promoting transcription facto E2F, until a go-ahead signal inactivates pRB. Mutations in RB are therefore associated with many cancers
11. Other examples include familial adenomatous popyposis and the APC tumor suppressor
a. And breast cancer associated with mutation in the tumor suppressor BRCA1, in which an inherited loss of function mutation increases a woman’s risk of cancer up to 50%
12. Three mechanisms for loss of heterozygosiy of a tumor suppressor gene:
a. Point mutation
b. Aneuploidy
c. Mitotic recombination between nonsister chromatids. -most common mechanism
13. Other mechanisms of tumorigenesis
a. Epigenetic changes
i. Altered covalent modifications to DNA and
histones (methylation and acetylation) that either
activate or inactivate chromosomal regions for
transcription. This results from aberrant
misexpression of DNA and histone modifying
enzymes, as we saw earlier for the absence of alpha
ketoglutarate
b. Micro-RNAs
i. MiRNAs are small (20-22 nucleotide) RNAs
encoded by nonprotein encoding genes. They bind
the 3’ regions of transcripts with which they have
sequence complementarity, and in doing so recruit a
protein complex that prevents translation (either by
degrading the transcript or inhibiting ribosomes
from translating). Depending on the function of
their target genes miRNAs may function in manners
similar to either oncogenes or tumor suppressors
c. LOF mutations in specific miRNA genes are involved in formation of some cancers, like chronic lymphocytic
leukemia (CLL) in which two miRNAs miR-5-a and miR 16 prevent cell proliferation by binding and targeting
transcripts for destruction. In their absence there is
increased proliferation of B cells
14. Identifying drivers of tumorigenesis
a. Rapid genomic sequencing ability has only recently been developed (past 10 years) which now make identifying mutations associated with cancer easier to identify
b. Before that, few mutations were identified. One notable exception is the BCR-ABL mutation associated with
Chronic Myelogenous Leukemia (CML)
BSC 300-001 Week 15 Notes
c. Many cases of CML result from chromosomal breakage
and rearrangement between two regions of chromosomes 9
and 22 – resulting in an easily identifiable hybrid
chromosome called the Philadelphia Chromosome
d. The translocation occurs between two genes BCR and
ABL, generating a hybrid gene and protein called BCR
ABL
i. BCR-ABL is a constitutively active kinase that
phosphorylates numerous cell proliferation proteins
like JAKs and STATs – promoting rapid
proliferation
1. It is also an example of a very successful
screen for molecules that can bind and
inhibit the hybrid protein only, and not
normal BCR or ABL
e. The drug named Gleevec (imatinib) bind and blocks only
BCR-ABL and effectively prevents its function
f. And has essentially made this type of CML as treatable as
diabetes, with a regular dosage of the drug
g. At what cost? Up until its patent expired in 2017 it cost
about $120,000 per year
i. Now the patent is expired and its annual cost is
about $8800
h. Sequencing cancer genomes has revealed there are few
driver mutations that are shared by diverse classes of
cancers
i. Most are shared among only 2-10% of tumors in any given
cancer type, so it is still not possible, for most types of
cancer, to pinpoint the most critical mutations that could be
the target of new drugs
j. However, as more and more cancers are sequenced patterns
may emerge that lead to this type of tumor-type specific
therapy
d. Misregulation of Cell Growth and Death Pathways
i. Effects of oncogenic mutations in proto-oncogenes that encode cell surface receptors
1. Receptors:
a. Oncogenic receptors do not require signaling molecules to
be activated
b. Mutations that alter HER2 and EGF receptor
transmembrane domains causes dimerization and constitute
activation of the receptor, even in the absence of thr normal
EGF-related ligand
ii. RTK/RAS/MAP kinase pathway components are frequently mutated in cancer
1. Signal-transducing proteins:
a. Ras is one of the most commonly mutated proto-oncogenes
in human cancers
BSC 300-001 Week 15 Notes
b. RTK/RAS/MAP kinase pathway components:
i. Gain of function mutations in many components of
this pathway generates an oncogene
ii. Recessive loss-of-function mutation in the NF1
GTPase-activating protein (GAP) leaves Ras in
GTP on state. The NF GAP was named after a type
of cancer associated with such LOF mutations
(neurofibromatosis)
iii. Chromosomal translocation in Burkitt’s lymphoma
1. Transcription factors:
a. In this example a translocation between chromosomes 8
and 14 positions the MYC proto-oncogene adjacent to an
antibody heavy chain (CH) gene, where MYC is regulated
by the antibody-gene promoter/enhancer
b. This causes overproduction of MYC transcription factor in
lymphocytes, which causes transformation into a
lymphoma
iv. Loss of TGF-Beta antiproliferation signaling
1. Transforming growth factor Beta (TGF-Beta) normally inhibits proliferation to control growth
2. Recall TGF-B binds to its receptor, leading to activation of Smad transcription factors
3. So cancer can be initiated by mutations that:
a. Prevent receptor/ligand binding
b. Prevent receptor dimerization
c. LOF in SMADs
d. Or the principle genes regulated by the SMADs
v. Two notable and often mutated tumor suppressor genes are the Retinoblastoma (Rb) and p53 genes
1. Rb blocks transcription of genes required for G1 to S transition. It is inactivated by G1 and G1/S Cyclin/Cdks. Loss of Rb allows
cells to progress into S phase without regulation
2. P53 – the guardian of the Genome – is activated in response to widespread DNA damage and transcriptionally activates the CDK inhibitor p21 in order to block entry into S phase, allowing time for cells to repair DNA damage. If repair is unsuccessful, p53 triggers apoptosis
3. With p53 absent cells proliferate with DNA damage leading to genetic instability and more DNA damage
4. A subcategory of p53’s target genes are the proapoptotic BAX and BAK encoding genes. Therefore in the absence of p53 cells cannot initiate apoptosis, neither intrinsic nor extrinsic
e. Deregulation of the Cell Cycle and Genome Maintenance Pathways in Cancer i. Restriction point control: pRB and unregulated passage from G1 to S 1. pRB guards against uncontrolled proliferation
2. mutations that promote unregulated passage from G1 to S phase are oncogenic in about 80% of human cancers
3. Not only due to mutations in RB
BSC 300-001 Week 15 Notes
4. Overproduction of Cyclin D
5. Expression of an inhibitory protein E7 – encoded by the Human Papilloma Virus
6. Loss of function mutations that prevent the inhibitory p16 from binding the Cyclin/CDK complex
ii. Loss of p53 abolishes the DNA damage checkpoint
1. P53 guards against excessive DNA damage
2. Recall DNA (UV irradiation) activates ATM kinase activity triggering three pathways leading to arrest in G1:
a. Phosphorylated Chk2 phosphorylates Cdc25A, marking it for degradation and blocking its role in S phase cdk
activation
b. Phosphorylated p53 is stable and promotes transcription of genes that arrest cells in G1, promote apoptosis, and repair DNA
c. Phosphorylation of MDM2 releases p53 from targeted
degradation
d. Another regulator p14ARF is activated and binds MDM2 preventing it from acting on p53
iii. The Most vulnerable locus in the human genome
1. A small 30kb region on human chromosome 9 is considered the most vulnerable locus in the human genome to oncogenic change 2. Tt encodes both the p16 protein and p14ARF as well as another cyclin D regulator p15
3. Therefore if this locus is deleted, both pRB and p53 functions are disrupted
12/6 Review Session
I. Lecture 23 – slide 13 pic
a. Mechanism for activating inflammazone
i. Priming signal – synthesis of NaLP3
1. Caspase 1 is the active component
II. V(D)J
a. Light chains have constant copy of locus
b. In order for functional gene, each B cell has to undergo VJ recombination in light and V(D)J in heavy chains
i. Loop out
1. Exposes OH group at end of gene itself
2. OH attack adjacent strand and cleaves intervening strand of DNA
a. Symmetric or asymmetric cleavage
i. Asymmetric – polymerase just adds correct nucleotides
b. Opening ends allows things like DNA polymerase to come in
and add nucleotides
c. DNA Ligase 4 joins them back together
d. Now we have a new locus – added together one joiner and one
variable segment
c. Junctional imprecision
d. C corresponds to constant region of light or heavy chain
i. End terminal region is where hydrovariable loops are
ii. C portion identical for every light chain
iii. Also identical for heavy chain but switching occurs (M and D to A or G) III. Lec 24 – slide 15
a. Big pic of processing and presentation?
i. Class I and II found in different cells
1. Class I in all nucleated cells
a. Present self to immune system so it will recognize cell so that
it won’t be killed
b. Introduces viral particles if that cell has been infected so it can
be recognized by t cells and be destroyed
2. Class II used by antigen presenting cells (macrophages, B cells,
dendritic cells)
a. Introduce antigens at cells following phagocytosis,
endocytosis, etc
b. Mechanism inside cell is very different
IV. How do helper T cells help B cells?
a. Bind to through cell surface B cell receptor – initiates endocytosis and digest it i. Present peptides on cell surface
ii. Bind to T cell that has binding affinity through t cell receptor
iii. Physical contact – release cytokines
iv. Leads to activation (proliferation, class switching, etc)
V. Cancer Slide 32
a. This is one way that viruses can cause cancer:
i. Src viral and Src cellular phosphorylate target proteins
1. Viral has acquired mutations in B start, so it is always on and mimics phosphorylation
a. So activation not necessary when it attaches bc its already on
12/6 Review Session
2. C src has to be phosphorylated
VI. Loss of function is losing that gene
VII. P53 becomes phosphorylated – MDM dissociates
a. P53 moves into nucleus and transcribe p21 – cell cycle checkpoint
i. Puts pause on cell cycle til damage can be removed
b. P53 since it has loss of function cannot tell the damaged cell to stop replicating – replication of damaged cell causes even more damage
VIII. Cancer has both gain and loss of function
IX. Benign – gain of function mutation
a. Grows to a certain point
b. If the cells are healthy they will be able to recognize that they are doing something wrong and put it in senescence (these are moles)
X. P16 and p15 turns on retinoblastoma
XI. P14 turns on p15
XII. Genes that encode these 3 are all within the same region of a chromo – tumor suppressors
a. Lose region – lose activators on tumor suppressor pathway
XIII. MHC restriction – incompatibility of MHC complex to present antigen that can be recognized by more than one type of T cell receptor and cell itself from being recognized by genotypic variant of T cell
XIV. MHC are a fancy type of cell surface receptor – have antigen or ligand bound to them – present that to receptor on another cell
a. MHC complex presents antigen to T cell receptor
b. Only if there is a perfect fit can that T cell be activated by it (lock and key) XV. CD4 and CD8 are located on respective T cells
a. Half of a binding site – other half is antigen
XVI. Helper T cell vs cytoplasmic T cell
a. Immature T cells can go either way – controlled by transcription factors XVII. Dendritic cells- branch of white blood that are scavengers
a. Role of getting into peripheral tissues and looking for non-self pathogens like bacteria/viruses
b. Migrate there in response to inflammation
c. Professional APCs
XVIII. Invariant chain
a. Involved with MHC class II – things that have been digested and ingested into endosome
b. Prevent class II from binding to random peptides that they have affinity for