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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/198

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

198 Cards in this Set

  • Front
  • Back
Steps involved in the formation of the bilaminar embryo
-zygote divides into the morula then the blastocyst
-cells of the inner cell mass rearrange into epiblast and hypoblast via differing adhesive properties
-the epiblast expands dorsally froming the amnion and amniotic cavity
-the hypoblast expands ventrally forming the primary yolk sac and releasing extraembryonic mesoderm cells
Development of the oocyte from ovulation to fertilization
-the prepature follicle in the ovary begins responding to FSH and LH
-oocyte matures and becomes polar
-oocyte is expelled into the peritoneal cavity.Ovum is surrounded by the zona pellucida, corona radiata, and associated cells
-ovum is collected by the fembriae and moved down the fallopian tube by smooth muscle contractions
-sperm moved up by swimming and smooth muscle contractions
-corpus luteum matures and secretes hormones
Twinning (types)
-fraternal: two (or more) eggs are released and fertilized
-monozygotic: cells splits when all are still totipotent
--early cleavage: 2 embryos, 2 placentas
--CM splitting: 2 embryos, 1 placenta (most common)
--ICM splitting with overlap: conjoined twins, may result in organ symmetry
Ectopic pregnancy (definition and common locations)
-When the embryo implants in non-uterine tissue. Constitutes an emergency because the fetus draws blood to it, risking hemorrhage on extraction.
- 54% ampullary, 25% isthmic (lower fallopian), 17% fimbrial, 4% interstitial, abdominal, ovarian or cervical
Embryo development: fertilization to implantation
- embryo divides as it travels down the fallopian tube
- day 4/5 blastocyst reaches the uterus and "hatches" from the zona pellucida
- day 6: trophoblast fuses with the endometrium on the ICM side. The synctiotrophoblasts surround the embryo and bury it in the endometrium and form the vascular network
Events of Fertilization
-spermatoza penetrate the corona radiata until reaching the zona pellucida
-sperm binds to zona pellucida via specific receptors on head
-sperm undergoes the acrosomal reaction releasing enzymes that bore a hole into the zona pellucida
-sperm membrane fuses with the ovum membrane releases DNA into the egg and triggering blocks to polyspermy
Reactions/blocks that prevent against polyspermy in the egg
-Fast: rapid depolarization of the egg membrane (-70 to +10). Prevents other sperm from adhering to the membrane
-Slow: calcium waves from the site of sperm entry induce cortical granules to fuse with the plasma membrane and release contents. This pushes the zona pellucida away from the zygote and hardens it
Gastrulation (definition and steps involved)
= movement of cells in the embryo to for the gastrula

- the primitive streak forms from the caudal end
-epiblast cells migrate to the primitive streak, pass through it and form the ventral endoderm layer, expanding caudally
-other epiblast cells make an epithelial to mesenchymal transition and from a mesodermal layer between the ecto and endoderms
-cells that pass through the primitive node (at the end of the primitive streak) become the notochord and precordal plate
Define induction
when antagonist cells signal to neighboring cells, triggering them to differentiate in a certain way
TFG-β signaling pathway
-TFG-β protein binds to a receptor in the plasma membrane
-receptor activates Smad protein, which complexes with a second Smad protein
-Smad complex moves into the nucleus and recruits other regulatory proteins
-complex activates transcription of specific genes
Growth Factor signaling pathway
- GF receptor phosphorylates itself upon binding GF
- Phosphorylated receptor activates Ras protein
-Ras protein activates MAP kinases
-MAP kinases phosphorylate and activate transcription factors which change gene expression
Fundamental characteristics of signal pathways
-Reception: receptor binds ligand
-Transmission: signal transduced through the cell, often through a G-protein coupled receptor
-amplification: signal is multiplied during transduction, allowing gene to be transcribed multiple times from one signal
-Outcome: often gene expression
-Termination: signal is stopped/degraded
Define cell signaling
when a cell releases a ligand that is received by another cell and usually induces change in the receiving cell by (de)activating transcription factors (signal transduction)
Types of cell signaling
-Inductive: cell triggers adjacent cells
-Gradient: response is based on ligand concentration
-Antagonistic: restricted gradient, response is based on the concentrations of ligand and antagonist recieved
-Cascade: cells successively signal adjacent cells
-Combinatorial: multiple ligands from multiple cells combine (Most realistic)
Hedgehog signaling pathway
-Hedgehog morphogens are processed within a cell and secreted.
-In the receptor cell, Patched recptors usually inhibit Smoothened receptors
-Hedgehog disrupts the binding of the two receptors so that Smoothened can send a
signal inside of the cell.
-Hedgehog functions based on a gradient of its concentration
Wingless signaling pathway
-Wingless functions on a gradient and is a morphogen.
-Its receptor protein is called Frizzled.
-Without a Wingless signal, the protein Disheveled is inactive, causeing Gsk3‐ß to phosphorylate ß‐catenin, so the latter degrades.
-With a Wingless signal, Disheveled is activated, so ß‐catenin cannot be phosphorylated and it goes into the nucleus to activate transcription.
Notch signaling pathway
-Notch signaling mediates lateral inhibition between adjacent cells
-A dominant cell differentiates then releases a signal to the cells around it, which trigger them to differentiate differently (ex: helper cells)
-The signaling protein Delta binds to the transmembrane protein Notch very tightly. The intracellular domain of notch is cleaved and travels into the nucleus to affect gene transcription.
Transcription factors in gastrulation leading to polarity
-in the bilaminar embryo the epiblast secretes Nodal and the hypoblast secretes an antagonist at the anterior end, setting up the anterior/posterior axis
-Noggin and Chordin from the notochord inhibit an inhibitor (BMP-4) allowing the neural plate to form
-cilia at the primitive node create a current so nodal is only expressed on the left side
Levels of gene regulation
Transcriptional
post-transcriptional modification
Translational
Post-translational modification
Achondroplasia
an inherited form of dwarfism resulting from defective signaling (growth factor FGFR3 is missing)
Holoprosencephaly
a disorder in which the embryonic forebrain fails to seperate fully, due to defective or absent Hedgehog signaling
Hox Genes
-encode homeodomain containing proteins
-involved in defining polarity
of the embryo
-Their arrangement on a strand of DNA correlates spatially to which body parts they affect during development.
-mutations can cause polydactyly and mis-arrangement of body parts
Pax genes
-homeobox containing genes
-control the expression of organs
-mutations will result in no development of organs or misplaced expression
-ex: Aniridia: no iris
stratified squamous epithelia (description, function and locations)
-multi-layered, surface cells are flattened and irregular
-protect against abrasion of moist tissues
-oral cavity, esophagus, vagina
columnar epithelia (description, function and locations)
-cells are taller than they are wide
-good for absorption (often have microvillae)
-intestine, gall bladder
simple cuboidal epithelia (description, function and locations)
-single layer of square cells, central nucleus
-excretory, secretory, or absorptive functions
-kindney tubules, salivary glands, pancreas
simple squamous epithelia (description, function and locations)
-single layer, flattened, irregular cells
-form a continuous lining, maintain selective diffusion barrier, good for moist surfaces
-endothelial lining of the circulatory system, mesothelial lining of the peritoneal cavity
pseudostratified columnar epithelia (description, function and locations)
-single layer though appearing multi-layered, elongated cells, nuclei in the basal 2/3 of the cell, often ciliated
-absorptive functions
-respiratory tract
stratified cuboidal epithelia (description, function and locations)
- 2-3 cell layers, square cells, central nuclei
- protection, secretion
- larger excretory ducts of the exocrine glands ex: sweat and salivary ducts
stratified squamous keratinized epithelia (description, function and locations)
- stratified squamous cells with a dead, denucleated layer on top (stratum cornea)
-resist abrasion, infection, desiccation
-epidermis
transitional epithelia (description, function and locations)
-Surface cells are dome shaped, large and pale stained, may be multinucleated; nuclei are large, round with prominent nucleoi; Luminal surface of the cells appears thick and more densely stained
-withstand stretch and toxicity
-urinary tract only
Morphological characteristics of epithelia
- number of cells: simple v. stratified
- shape of cells: squamous, cuboidal, columnar
-surface modifications:
--apical: cilia, microvilli, stereocilia
--lateral/basal: tight junctions, adherens junctions, desosomes, gap junction, hemidesosome
(- organization of cells: pseudostratified, karotinized, transitional)
tight junction (zonula occludens)
-barrier between the apical and basolateral membrane surfaces
-regulates solute movement in the paracellular pathway (in between cells)
-keeps membrane proteins in the appropriate domain (maintains cell polarity)
Adherens junction (zonula adherens)
-band below the tight junction
-attached to actin terminal web in adjacent cells, links the actin cytoskeletons
-uses cadherin molecules (Ca+ dependent) to span the intercellular space
-important for cell shape and motility
Desosome (macula adherens)
-spot adhesions between cells
-link the intermediate filament networks of adjacent cells (keratin) via cytoplasmic plaques
-highly organized, use cadherins to span the intercellular space
-important for cell integrity
-railroad track appearance on imaging
Gap Junction
-intercellular channels for small molecules ( <1000 MW)
-composed of connexin proteins, 6 group in a connexon which interacts with a connexon of an adjacent cell to form a channel. Connexons often form plaques.
Hemidesosome
-anchors intermediate filaments to the basement membrane
-prominent in epidermis
Cilia
-motile structures that move wave-like to propel fluid
-made of microtubules organized in "axoneme"
-common in the respiratory and female reproductive tracts
microvillae
-non-motile structures that increase surface area
-stabilized by actin filaments anchored in the terminal web
-found in the small intestine, and proximal kidney tube
stereocilia
-large, non-motile structures that facilitate absorption (are branched microvilli)
-supported by actin
-found in the epididymus
Tissues formed from the ectoderm
Neural tube: central nervous system (brain, spinal cord, retina, posterior pituitary), neural crest (peripheral nervous system)

Outer epithelia: anterior pituitary, lens + cornea, inner ear, epidermis and appendages, tooth enamel, anal and or epithelium
Melanocytes: identity and origin
-melanocytes are pigment cells in the epidermis
-derived from the neural crest, and migrate to the dermis then epidermis during development
-if mutated can result in albinism
Langerhan cells: identity and origin
- antigen presenting cells of the immune system
-mesodermally derived from precursors in bone marrow. During development they migrate to the epidermis
Neuralation (steps)
-the notochord induces the dorsal ectoderm to form the neural plate
- the neural plate folds laterally to form a groove and continues to fold up into itself.
-the two most lateral apical surfaces of the groove fuse and separate to form the neural crest.
-Closure of the tube begins midway and extends the closure so that there are only two open pores (anterior and posterior neuropores) at either end.
-Neural tube becomes CNS
-Neural crest cells undergo EMT and migrate away to form PNS
Development of mammary tissue (as demonstrated by experiments with testicular feminization, TFM)
the mesoderm, in the absence of testosterone, induces the ectoderm to develop an elaborate duct system in the breast tissue.
Development of hair follicles
Hair is generated by the downgrowth of epithelial cells induced by the mesodermal cells of the skin, the dermal papilla. The dermal papilla signal via FGF-5 to induce the epithelia to develop a hair follicle.
Development of teeth
-The dental papilla (mesencyhmal ectoderm from the neural crest) induces the oral epithelia to form down-growths into the papilla. These cells are converted to enamel. Dentin is formed from mesenchyme-dervide cells, filling in the cavity.
Inductive interactions in skin differentiation
-ectoderm induces mesoderm to differentiate into dermis
-mesoderm induces ectoderm to differentiate into epidermis
-the body location of the mesoderm determines the characteristics of the epidermis
Characteristics and cause of ectodermal dysplasia
- 2+ abnormal ectodermal features (generally in skin and skin appendages)
-due to mutations in ectodermal cell-cell communication, adhesion, transcription regulation, other functions
-EX: mutation in ectodysplasin pathway (signaling protein) results in anhidrosis (too little sweating)
Epithelia derived from each germ layer
Ectoderm: epidermis
Mesoderm: mesothelium, endothelium, epithelia of ducts and tubules
Endoderm: epithelia lining the gut and derivatives (respiratory tract)
Tissues derived from the endoderm
-gut (pancreas, liver, digestive tract, trachea, lungs)
-urinary bladder
-pharynx, thyroid, pharyngeal pouches
Major derivatives of the neural crest cells
-entire PNS (except axons of motor neurons)
-adrenal medulla
-melanocytes
-cranial neural crest cells contribute to the bones, dermis, fat, and muscles of the face and head
Morphogenic movements of the endoderm that creates the gut tube
- the endorm folds laterally while the entire embryo elongates and curves medially
- the outer endoderm and amnion fold around, creating tub of endoderm
- the yolk sac is constricted and reduced to a yolk stem
tracheoesphageal fistula: characteristics and cause
-during development the trachea buds off from the gut tube. A septum forms to divide it from the esophagus. If the septum does not form correctly a TEF results
-Most TEFs have an atetic or stenotic segment in the E or T
-4 types: TEF followed by a gap in the E, a gap in the E before the TEF, two TEFs with a gap in between in the E, TEF without gap
How do derivatives of the gut form?
The are induced to bud off from the main tube by signaling interaction with the surrounding mesenchyme cells
What features distinguish eukaryotes from prokaryotes
- membrane bound organelles
- cytoskeleton
- complex plasma membrane systems (important for communication with other cells) (and has cholesterol)
The importance of compartmentalization in eukaryotic cells
- localizes functioning and allows multiple different environments, optimized for different functions, to exist within the cell (increase efficiency)
- increase regulation (particularly though membranes)
- protection (isolate processes that could damage the rest of the cell)
Importance of membrane fluidity
- allows for proteins to diffuse and interact with each other
- important for signaling mechanism
-establishes cell polarity
-flexibility in adapting to the enviroment
Membrane Fluidity and Asymmetry
- different proteins and lipids are expressed in different areas of the membrane based on function creating cellular polarity
-fluidity is controlled by lipid tails (length and saturation), cholesterol
Types of membrane proteins
transmembrane: have domain that spans the membrane, usually made from alpha helixes with + charged amino acids on either end
membrane associated: anchored to the cytosolic surface by an amphipathic alpha helix.
lipid-linked: attached to either side of the bilayer by covalent attachment to a lipid molecule
Protein attached – attached by relatively weak, noncovalent interactions with other membrane proteins
Lipid raft
- patch of tightly packed lipids and cholesterols within the membrane creating a rigid segment
- have transmembrane proteins with longer transmembrane domains
-concentration of signal transduction receptors, acts as a “signaling platform.”
Glycocalyx
-a collection of sugars on the extracellular surface that are attached to glycolipids and glycoproteins of the plasma membrane.
-help protect the cell surface from mechanical and chemical damage, while aiding in cell communication and adhesion. -hydrophilic, conferring a negative charge and associated nucleophilicity to the cell.
-it's deterioration is a signal for cell death
microfilaments
The thinnest filaments of the cytoskeleton. They are polymers that allow cells to change shape by building itself up and breaking itself down (e.g. actin)
intermediate filaments
These filaments are static, providing mechanical support for stress (expansion or contraction). In the heart, intermediate filaments create a mechanical scaffold that allows the heart to beat without breaking down. They are often anchored to the plasma membrane at desmosomes and hemidesmosomes to provide connectivity. (E.g. keratins).
microtubules
These are long, hollow polymers that drive movement within a cell. Microtubules are the “railroad” on which vesicles are transported between organelles and the membrane. They have the ability to grow and shrink to give shape to the cell membrane. (E.g. tubulin).
Nuclear envelope
-double membrane contiguous with the ER
-segregates immature messengers and DNA from the rest of the cell
-nuclear pores: (8-point symmetry, basket and fibrils); selectively allows proteins and signals in/out of the nucleus
Endoplasmic reticulum
Rough ER: 1/3 of proteins synthesis, includes transmembrane proteins and secretory/excretory proteins
Smooth ER: modifies proteins, espeically hormones, produces cholesterols,
Trans-location of membrane-bound proteins to the ER during translation
-membrane bround proteins have signal sequence: 20AA hydrophobic sequence that binds to SRP and stops translation
-SRP complex binds to receptor on ER and nacent protein slides into a channel, translation continues
-signal protein is cleaved after transcription
-May also have stop-transfer sequences that make the protein transmembrane (rather than secreted protein)
Glycosylphosphatidylinositol (GPI)-modification
-After a protein has translated to the lumen of ER, cleavage of the C-terminus signal sequence creates a signal (set of AA) that allows the attachment of a complex lipid (GPI) anchoring the protein to the membrane
- GPI-bound proteins have the potential to end up in the extracellular leaflet of the plasma membrane, and these serve a purpose to concentrate proteins in the lipid raft and to direct proteins to different places in the cel
N-linked oligosaccharide initiation
-As the polypeptide chain is translocated into the ER lumen, it is glycosylated by oligosaccharide side chains at particular asparagines (ASN)
-Each oligosaccharide is transferred as an intact unit to the asparagine from a lipid called dolichol, catalyzed by a transferase
-Glycosylation plays an important role in helping the folding of proteins and also in the recognition of one cell by another on the cell surface
Disulfide bonds in protein folding
-Disulfide bonds are formed by an enzyme that resides in the ER lumen.
-Disulfide bonds help to stabilize the structure of the proteins that may encounter changes in pH and degradative enzymes outside the cell, either after they are secreted or after they are incorporated into the plasma membrane
Golgi structure and function
-cis face: faces ER, receives (and returns) proteins from ER via vesicles, other proteins will progress through the stack and be modified
-Trans face: facing plasma membrane, releases fully modified proteins for secretion/implantation
-medial: middle region
-Each region contains different set of modifying enzymes (biochemical asymmetry), especially for glycosylation
-Proteins sorting: proteins are tagged with signals that determine their ultimate destination (eg. lysosome)
Endocytotic pathway
-Molecules and receptors on the cell surface are engulfed by the plasma membrane, which invaginates and buds off.
-Binding of a ligand allows the receptor tail to bind GTP receptor proteins in the cytosol which recruit clatherin coat proteins and adapters. More coat molecules are recruited as invagination progresses
-Dynamin then forms a spring around the stalk and restricts it separating the vesicle
-A v-SNARE (transmembrane protein) and specific Rab proteins (GTP-ases) associate with the vesicle and are responsible for recognizing the target
-coat/cage is mostly released after budding is complete
Receptor fates after endocytosis
Recyling:
-acid in the endosome detatches the ligand from the receptor
-receptor (and sometimes carrier) bud off and return to the membrane
Degradation:
-receptor is moved to the lysosome and degraded
-shuts down the signal pathway
-acts a part of the signal transduction (endosome moves the receptor to different parts of the cell)
Constitutive vs. Regulated endocytosis
-Constitutive endocytosis occurs whether or not a ligand is bound to the receptor on the surface of the cell. The cell will continuously pull the receptor in and sort it. If no molecule is attached, the receptor is recycled and the process continues
-Regulated endocytosis is mediated by the binding of a ligand such as a growth factor to a cell surface receptor. The binding triggers internalization of the receptor, which initiates signal transduction
Lyosomes
- Responsible for degrading certain cell components and material internalized from
the extracellular environment
Key Features:
- single membrane,pH 5 (critical for enzyme function), acid hydrolases carry out
degradation reactions, can store heavy metals and calcium
Vesicle targeting with SNARE and Rab
- Snares on the membrane of the vesicles recognize snares in target organelles. They zipper together and bring the membranes in contact
-Membrane fusion is regulated by GTPases called Rabs -- multiple Ras create specificity. In order to bind, the vessicle Rab need to match the specific rab effector on the target membrane. Rabs/effectors bring the SNARES in contact and allow them to bind
Peroxisomes
small organelles that are responsible for breaking down materials brought into the cell. Peroxisomes contain enzymes that allow for oxidation of lipids and toxins. These are associated with Zellweger disease.
Diseases of the endocyotic pathway
There are many diseases associated with abnormalities in the endocytic pathway, in particular with abnormalities in the lysosomes. The main feature is that some enzyme in the lysosome fails to properly digest a key protein that leads to disease (Huntington’s, Parkinson’s, and Alzheimers)
-hypercholesterolemia: Disease can occur if endocytosis does not begin appropriately and the cell fails to engulf the appropriate molecules in the extracellular space
Pathology of Familial Hypercholesterolemia
-inherited disease (recessive or dominant depending) in which the cell is unable to recognize or use dietary LDL and so produces its own
-4 types:
Synthesis defect (receptor never made), transport defect (receptor degraded before reaching the golgi), defective receptor (can't bind LDL), clathrin defect (can't find clathrin to form coat and vesicle)
-Class 4: can have multiple types: nonsense, frame shift, false stop that determines the extent of the disease
-Statins work by turning off cell LDL production
Intermediate filament structure and function
Subunits are composed of an N­‐terminal globular, a C-­‐terminal globular tail, and a central elongated domain of an extended alpha-­‐helical region.The alpha-­‐helix allows for two subunits to wrap around each other forming a coiled-­‐coil dimer. Dimers associate to form a tetramer. The tetramer is the functional unit of IFs and come together to form long chains. Eight tetramer chains form a ropelike filament.
-There 4 different classes of subunits including keratins, vimetins, and lamins. (useful for determining tumor identity)
-IFs provide a scaffolding to withstand mechanical stress
Intermediate filaments in epithelia
- provide support and scaffolding in the cell against mechanical stress
- anchor cells to other cells and the basement membrane via desosomes and heidesosomes
Intermediate filaments in neurons
- stabilized and reinforce the axon and prevent against shearing forces
Intermediate filaments in nuclei
- provide structural for the nuclear envelope by forming the nuclear lamina
- also anchors cell membrane proteins and chromatin
- must be disassembled during mitosis
intermediate filaments and tumor origin
Because there are several types of IFs, antibodies to specific kinds can be used to identify the original tissue in metastatic tumors
Functions of actin filaments
- support microvilli (core has actin bundles, providing sturdy scaffolding)
- act as contractile bundles in the cytoplasm (esp. in muscle cells)
- form temporary structures such as dynamic protrusions, especially in cell migration
- form the contractile ring in cell division
- interact with adherens junctions to stabilize cell structure and interact with other cells
Polarization of actin filaments
Actin filaments have directionality:
- subunits are added more rapidly at the + end (though they can be added at the - end)
- addition/degradation at opposite ends allows for treadmilling of the filaments, and movement independent of myosin (listeria take advantage of this to move around)
- myosin motors can only move toward the + end
Functional classes of actin binding proteins
- nucleating proteins: bind and end of the filament to a membrane
- severing proteins: split a monomer into smaller subunits (usually during disassembly)
- cross linking proteins (in cell cortex): binds two crossing filaments into a mesh just under the PM
- capping proteins: blocks extension and degradation of the filament end
- side-binding proteins: binds to the side of the filament to regulate and stabilize contractility
- motor protein: forms contractile bundles (as in muscle)
- bundling protein (in filopodia): bundles proteins together in microvilli
- monomer-sequestering proteins: binds to subunits, preventing polymerization
Function of myosin motor proteins
- hydrolize ATP to move along actin/thin filaments.
- 30+ different types
- Myosin I: single contractile head, variable length tails, associates with membrane or vesicle to move things in around the cell
- myosin II: 2 contractile heads, exclusively in muscle cells
- myosin IV: important in hair cells and hearing
Conformational changes in the myosin head that couple ATP hydrolysis to movement
- Myosin is associated with a subunit on the actin filament
- Head binds ATP, releasing it from the filament
- Head hydrolizes ATP, causing conformational change that pivots it toward the + end and binds to a new subunit
- phosphate is released, causing the head to pivot back, pulling the filament (power stroke)
- ADP is released, allowing new ATP to bind
kinesin
- Motor protein family responsible for antograde movement in axons and contraction of the mitotic spindle
- Move toward the + end of mictotubules (though some can reverse). Can move continuously along the tubule.
- Motor proteins have two heads that interact with the microtubules
dynein
- motor proteins involved in movement of flagella & cilia (sliding movement along microtubules produces waving) and degradation of chromosomes
- move toward the - ends of microtubules
- Have a conserved large head domain
Organization of microtubules in the cell
- microtubules radiate outward from the microtubule organizing center (centrosome)
- The centrosome has two centrioles which are arranged perpendicularly to eachother, and may be used as templates for new MT synthesis
- MTs are nucleated at the centrosome at the - end
- in ciliated cell, centrioles duplicate, becomeing basal bodies, the organizing centers for cilia MTs (have 9+2 organization of MTs in cilia)
- during replication, centrosome is duplicated, and MT's expand from opposite poles of the cell
Microtubule structure and assembly
- MTs are made up of 13 parallel protofilaments. Each protofilament is made up of end to end heterodimers of α and β tubulin. α & β tubulin proteins each associate with GTP as a cofactor (important for assembly and dynamics of MTs). The β-tubulin subunit is toward the + end.
- can form doublet (flagella, cilia) or triplet (basal bodies, centrioles) MT's by splicing rings together
- MTs can self assemble in vitro, but in vivo it's very controlled: gamma tubulin forms a template at the minus end
Microtubule polarity
- + end grows more rapidly than the - end
- the - end is the nucleating end and is embedded in the centrosome
- MTs of the dendrite are of mixed polarity (unlike the axon)
dynamic instability of microtubules
- unless the end is capped or captured, MTs will always be continually growing and dis-assembling
- alternates between assembly at the + end and catastrophic disassembly via hydrolysis of the GTP associated β-tubulin
- this ends when the plus end is captured (and capped) and bound to stabilizing proteins
Anti-microtubule agents as anti-tumor drugs
Two mechanisms that upset the formation and function of the mitotic spindle, inhibiting mitosis and preferentially killing dividing cells
- cause MT depolymerization by destabilizing the MT structure or binding to and stabilizing free tubulin
- overly stabilize MT (ex taxol), causing a net increase in polymerization and preventing disassembly/shortening
Functions of cilia
3 classes:
- motile: move fluid and materials, protective barrier
- primary: immotile, involved in signal transduction
- sensory: involved in detecting sensory stimuli (sound, light)
Mictrotubule organization in the mitotic spindle
- MTs radiate from centrosome at opposite poles of the cell.
- 3 major MT types involved:
--astral: radiate in all directions, interact with dynein motors at the cortex, important in separating the poles in anaphase B (provide the force for separating chromosomes, anchor centromeres).
--kinetochore: attach to chromosomes and retract to separate them, undergo slow flux (treadmilling) despite being capped
--overlap: MT's form opposite poles overalap at the equator and push the poles apart with kinesin motors (anaphase B)
Segregation of chromosomes during anaphase A by microtubules
- once bound to the all the chromosomes, MT's retract to the poles by depolymerizing at both ends. Most shortening occurs at the kinetochore (+ end)
Two theories of movement:
- MT motor proteins at kinetochore use the ATP in the MT to move and consequently depolymerize the tubule
-kinetochore motor proteins have a higher affinity for polymerized subunits, so as the tubule depolymerizes they move toward the pole to maintain binding
Activity of muscle types
Skeletal: strong, quick, discontinuous, voluntary contraction
Cardiac: strong, quick, continuous, involuntary contraction
Smooth: weak, slow, involuntary contraction
muscle fiber
A muscle fiber is a single muscle cell. In skeletal muscle each fiber has several nuclei because it is formed by fusion of myoblasts
-The plasma membrane of the muscle fiber is the sarcolemma
Paraxial mesoderm
- develops adjacent to the neural tube
- develops into the somites, the dermis of the skin, axial and limb muscles, and axial skeleton
Intermediate mesoderm
- develops between the paraxial and lateral mesoderm
- gives rise to the genitourinary system (kidney, ureter, adrenal cortex, gonads, vagina, uterus, uterine tubes (not bladder or gametes)).
Splanchnic (visceral) mesoderm
- derived from the lateral mesoderm after it splits to form the coelom cavities
- gives rise to the mesenteries, the epicaridum, the blood cells/blood vessels, endothelium, the myocardium, the respiratory tract wall, and the gut wall (basically the tissues that’s intimately associated with the organs of the body).
Somatic (parietal) mesoderm
- derived from the lateral mesoderm after it splits to form the coelom cavities
- gives rise to the limb skeleton and the parietal tissue, or the tissue that covers the body wall.
Lineage tracing to determine muscle origin
- chick somites were replaced with quail somites, allowing researchers to see where those cells developed in the adult animal
- also can use fluorescent tracers, or engineering fluorescent proteins
Development of skeletal muscle fiber from myoblasts
- mononucleate myoblasts leave the cell cycle (fully differentiated) and fuse together
- assembly of the contractile apparatus pushes nuclei to the edge of the cell
- further growth of the fiber involves the fusion of satellite cells (muscle stem cells)
Fast vs slow twitch muscle
- slow twitch muscle have lots of mitochondria and blood supply (giving it a red color to due to the heme in the myoglobin)
- fast twitch has few mitochondria and less myoglobin because it relies on anaerobic glycolytic pathways to contract
Coelom
- cavity formed by the splitting of the later mesoderm into the splanchnic and somatic mesoderm. It forms the peritoneal, pleural, and pericardial cavities
Splanchnic
- of or relating to the viscera: visceral
- splanchnic mesoderm becomes most visceral tissue, or those that are intimately associated with the organs
Viscus or visceral
- an internal organ of the body, especially one located in the great cavity of the trunk proper (heart/liver).
Somatic
- of or relating to the wall of the body, parietal
- somatic mesoderm forms the parietal tissue, or the tissue that lines the body wall
Parietal
- of or relating to the walls of a part or cavity.
- The parietal tissue lines the body wall of an organism, as opposed to tissue that lines the organs themselves.
Axial
- relating to or situated in the central part of the body, in the head and trunk as distinguished from the limbs.
Somite
- one of the longitudinal series of segments into which the body of many animals is divided.
- somites are derived from the paraxial mesoderm, and organized into series of segmentally arranged blocks from the occipital region caudalward
Mechanisms of bone formation
- endochondrial ossification: cartilage model of the bone formed during embyrogenesis then calcified
- intramembranous ossification: direct ossification by mesenchymal cells (no cartilage intermediate)
Developmental origins of the circulatory system
- derived from the splanchnic mesoderm
- initially forms two parallel left and right systems. The parallel structures fuse and form the primordial heart in the 3rd week
-Initially the primordial heard is anterior to the oropharyngeal membrane but migrates as the embryo folds
Major subdivision of somites and their adult derivatives
- the ventromedial portion of each somite becomes the sclerotome which forms the axial bones.
- the dorsolateral and dorsomedial portions of the somite form the myotome and differentiate into muscle cells.
- The dorsolateral region forms the limb and body wall muscles while the more medial cells form the muscles of the back.
- The most dorsal part of the somite forms the dermatome that differentiates into the dermis.
Signal pathway that differentiates axial bone in somites
Sonic hedgehog (SHH), a secreted signal molecule expressed in the floor plate of the neural tube and the notochord, initiates PAX1 expression in presumptive sclerotome cells, and this transcription factor regulates differentiation of these cells into bone.
Signal pathway that differentiates back muscle in somites
Expression of the growth factor bone morphogenetic protein 4 (BMP4), by overlying ectoderm, causes secretion of WNT growth factors from the dorsal region of the neural tube. In turn, these growth factors cause expression of the muscle-specific gene MYF5 in the dorsomedial (closest) portion of the somite, and this gene regulates development of epimeric (back) muscle.
Signal pathway that differentiates limb and body wall muscle in somites
WNT proteins expressed by overlying ectoderm, together with BMP4 expressed by lateral plate mesoderm, turn on MYOD expression in the dorsolateral (closest to splanchnic mesoderm) part of the somite. MYOD is another muscle-specific gene that causes this region to differentiate into limb and body wall muscles (hypomeric musculature)
Signal pathway that differentiates dermis in somites
Dermis development is controlled by the transcription factor PAX3, the expression of which is initiated by the growth factor neurotrophin 3 (NT3), secreted by the (central) dorsal region of the neural tube.
Mesenchymal-epithelial transitions and epithelial-mesenchmyal transitions in somites
- The somitocoel cells that line the cavity are epithelial cells, having undergone an MET
- The ventromedial portion of the somite undergoes an EMT to become the sclerotome
Hox 10 mutants in axial skeletal development
- Hox 10 is normally expressed in the lumbar and sacral regions
- a mutation (deletion) in Hox10 genes allows for continued expression of earlier Hox genes, resulting in thoracic vertebral morphology (ribs) in lumbar and sacral regions and disrupts normal rostrocaudal differentiation.
Function of cyclin dependent kinases in the cell cycle
- control progress through the phases of the cell cycle with successive activation and deactivation (prevents regression)
- cdk concentration is stable, though activity oscillates throughout the cycle due to the presence of cyclins
- active cdk phosphorylates intracellular proteins that initiate, regulate, and terminate major events of the cell cycle.
4 main classes of cyclins in cell replication
- G1/S-cyclins – complex with Cdks (to form MPF) in late G1 and trigger progression through the restriction point and entry into S phase.
- S-cyclins – bind Cdks and help stimulate chromosome duplication; levels remain elevated until the beginning of mitosis.
- M-cyclins – activates Cdks which stimulate entry into the mitosis phase and cause cell division.
- G1-cyclin – binds to Cdks which then push the cell into the G1 stage after mitosis.
G1 in cell cycle
- This is a stage of growth directly after mitosis has created two new daughter cells.
- Provides time for the cell to monitor internal and external environment to ensure optimal conditions and preparation for proliferation.
- Cells may delay passage from G1 phase for even years if conditions are not correct.
- Includes a point of no return called the G1/S transition restriction point after which cells are committed to DNA replication and division.
S phase in cell cycle
- After G1, when chromosome duplication occurs.
- S-CDK responsible for initiating DNA replication once per cycle for cells:
Signaling cascade begins in G1 with the prereplication complex (pre-RC). S-CDK initiates nucleation of the pre-RC into preinitiation complex which unwinds the DNA helix and adds DNA polymerases/other replication enzymes. DNA is then replicated.
G2 phase in cell cycle
- phase after completion of DNA replication.
- Growth of cell and organelles to prepare for division of cell.
- Includes DNA damage checkpoint.
- Monitoring of internal and external environment to determine if ideal for cell division to commence.
Mitosis
- cell cycle phase in which division occurs
- 6 phases: Prophase, prometaphase, metaphase, anaphase, telophase, (and cytokinesis)
cdk activation
- cdk binds to specific cyclins as they become synthesized in the cell
- cyclin-cdk complex has 2 phosphorylation sites. Initially both are phosphorylated by activating and inhibitory kinases (primed complex)
- the inactivating phosphate is then removed by an activating phosphatase (active complex)
- the active cyclin-cdk complex inbibits the inhibitory kinase and activates the activating phosphatase, rapidly increasing overall cdk activity/signal
-
cdk inactivation
- cdk inactivation occurs when cyclin is degraded, which is mediated by ubiquitin (a protein tag for proteasome degradation; mechanism involves 3 protein complex that loads, transfers, and targets the signal)
- active cyclin-cdk activates a subunit which activates the E3 protein in the ubiquitin complex. Ubiquitin then rapidly labels cyclin-cdk for destruction in the proteasome, inactivating cdk
Major events of the G2/M transition
- cyclin, which has been slowly being synthesized during interphase, binds to cdk forming inactive MPF. which is then primed by phosphorylation by activating inhibitory kinases
- A small amount of MPF is activated by Cdc25 phosphatase, creating a positive feedback resulting in rapid activation of MPF and spike in MPF activity
- MPF activates numerous kinases (including nuclear lamins), triggering mitosis
G1 restriction point
- when the cell becomes independent of growth factor for triggering cell division
- The point at which the cell becomes committed to divide
Growth factor mediated signalling for division during M1
GF (mitogen) binds to PM receptor--activates Ras protein--activates MAP kinases--activates transcription factors--increases cyclin-D transcription--increased active G1 complex (cyclin-D/Cdk4)--activates G1/S complex (cyclin-E/Cdk2)---eventually accumulates enough that the cell no longer responds to this pathway and is committed to division (restriction point)
Mechanism of G1/S transition past the restriction point
- G1 and G1/S complexes, which have been triggered by GF signalling, enter a positive feedback loop of kinase activation.
- Both complexes phosphorylate Rb protein, inactivating it, releasing E2F (which then positively activates other E2F)
- E2F promotes transcription of S phase proteins further activating cyclin complexes (esp S cdk) and moving the cell toward synthesis
S Cdk complex activation of synthesis
- DNA replication is initiated in multiple places on the genome by origin recognition complexes (ORC) which are normally inactived by binding to Cdc6
S-phase Cdk phosphorylates Cdc6, releasing ORC and leading to Cdc6 degradation
- Unbound ORC is phosphorylated (active) and initiates replication
Cell cycle checkpoints
- end of S phase: ensure chromosomes fully replicated
- metaphase: ensure all chromosomes are lined up at the midline (this is accomplished by signals sent from unattatched kinetochores that suppress anaphase promoting complex (a cdk))
- end of S phase: repair any damage to DNA (damaged areas will recruit repair proteins while activating p53 which activates expression of p21, CKI, which inhibits S-phase cdk, preventing the cell from leaving S phase)
-
G0 withdrawal
- generally fully differentiated cells will no longer go through the cell cycle
- In G0, cells are kept from replicating by CKIs (cyclin-dependent kinase inhibitors) that bind cdk complexes.
- cells usually enter G0 if they are deprived of growth/divide signals during G1.
Ataxia telangiectasia
- A childhood neurodegenerative disease associated with an increased risk of cancers. This is caused by a defect in the ATM/ATR kinases, which are responsible for recruiting DNA repair enzymes and stalls the S-phase until the DNA has been repaired
Xeroderma pigmentosum
- A disease characterized by hypersensitivity to UV rays and an increased riskof melanoma. This is caused by a mutation in the DNA repair genes.
Li Fraumeni syndrome
- This is associated with an increased risk of a variety of cancers and is caused by a defect in p53, preventing the cell from activating p21 to stall S-phase entry and repair defective DNA.
Retinoblastoma
- A childhood tumor of the retina caused by unregulated cell proliferation because of defects in the Rb protein responsible for maintaining the inactivity of S-phase cyclins.
Nuclear lamina
- network of lamin filaments directly below the nuclear envelope (provides structural integrity)
- composed of 3 lamins. Lamin A/C are from the gene (different C-terminus splicing), B is a separate gene
- connects to the cytoskeleton, anchoring the nucleus in the cell via Sun/KASH complexes (overlap in the lumen of the nuclear envelope, Sun links to the lamina, KASH to cytoskeleton)
Transport through the nuclear pore complex
- to travel into the nucleus proteins are tagged with a nuclear localization signal which is recognized by a signal complex (α&β subunits) to form a cargo receptor complex. The receptor complex interacts with the NPC and passes through.
- Inside the nucleus Ran-GTP binds and releases the cargo from the complex which then exit via the NPC. Back in the cytosol the Ran-GTP is hydrolized to Ran-GDP, dissociating the carrier and α&β subunits
Chromatin packaging
- nucleosome: DNA is wrapped twice around histone octomers (H2A,2B,3,4) then associated with histone monomer (H1). Histone tails in these complexes are available for modification (important for chromatid structure)
- fibrils: nucleosome strand is coiled on itself into 30nm fibril
- Fibrils are then arranged in loops that are condensed into the chromosome
- Chromatin is most dense when packed in the mitotic chromosome
- Transcriptionally active "euchromatin" is less dense and central in the nucleus. Not active, "heterochromatin" is more dense and peripherally located
Histone Code and DNA remodeling/regulation
- Modifications to histone tails that are associated with certain chromatin states (i.e. availability for reading). Locally impact chromatin structure and recruit transcription factors
- possible modifications include acetylations, methylations, and phosphorylations
- histone modifying enzyme works in conjunction with chromatin-remodeling complex to make DNA available for transcription
- modifications to chomatin structure are collective known as the epigenome
Chromatin as a target for chemotherapy
- histone deacetylase (HDAC) inhibitors: block removal of acetylations from histone tails. Increase acetylation can disrupt gene expression, stopping cancer progression
- clinically seems to be effective in some leukemias
Models of pathology in laminopathies
- the lamina seems to be critical for nuclear/cellular integrity, particularly in cell undergoing mechanical stress (like muscle). Mutations therefore cause weaker adhesions (esp. in Sun/Kash complexes) and preferentially effect muscle due to high mechanical stress.
- Lamins and other nuclear envelop proteins interact directly with chromatin (and modying factors, TX factors). These interactions may influence gene expression, and mutation therefore could lead to altered expression and phenotype
- Emery dreyfus muscular dytrophy and other diseases result from mutated lamins
Factors influencing animal and organ size
- cell number (division vs death)
- cell size (synthesis vs. degradation of molecules/proteins)
- each of these factors is influenced by genetic patterns of expression and access to nutrients
Neufield experiments on cell division vs. growth
- alternately over expressed E2F (increased division) and Rb (reduced division) in embryos. Neither changed overall organism size relative to the control
- Showed that growth is not a consequence of division
BUT; division is sensitive to growth: starvation slows replication, growth must be "upstream" of division
In vivo determinants of growth
- intrinsic: pathways that act within a tissue to regulate its
growth down a preprogrammed developmental path
- Extrinsic: mediate the ability of a tissue to communicate
with other tissues and auto-regulate size
- Environmental: nutrient availability (sugars, amino acids) & use (e.g. muscle in bodybuilders)
SRY gene is...
the master regulatory gene in testes development
Metcalf's experiments of local & systemic control of cell proliferation
- Ist exp: transplanted multiple thymuses into a single host. Each grew to a full size demonstrating "intrinsic" growth control
- 2nd exp: transplanted multiple spleens into 1 host. Each grew to a fractional size proportional to the number in the body, demonstrating "extrinsic" growth control
insulin-like growth factor (IGF-1) pathway
- controls cell growth by increasing the biosynthetic capacity of the cell
- triggers production of ribosome subunit S6P and activates elF-4e which is a mRNA receptor and transcription factor
- Not found in yeast (no extrinsic signalling)
- Defects in ligand (IGF-1) or receptor (IRS-1) lead to fewer, smaller cells
- PTEN ("brake" in pathway) results in more, larger cells
- Dog size is a result of IGF activity
TSC/TOR pathway (tuberous sclerosis complex/Target of rapamycin)
- - controls cell growth by increasing the biosynthetic capacity of the cell
- triggers production of ribosome subunit S6P and activates elF-4e which is a mRNA receptor and transcription factor (same as IGF-1)
- TOR is nutritionally sensitive, activates in response to nutrient availibility
Upstream open reading frames and growth dependent cell replication
- cylin mRNA has 2 ORFs, one is upstream of the coding sequence and does not code for protein.
- Ribosomes will bind to the upstream ORF but fall off before the coding sequence.
- In high nutrient media, ribosomes are abundant due to TSC/TOR pathway, so more ribosome will reach the coding sequence and more cylin will be transcribed, moving the cell towards division.
Myostatin as an extrinsic growth factor
- TGF- β protein that inhibits muscle differentiation and growth. (External signal that stops muscle growth: extrinsic)
- Produced and secreted exclusively by skeletal muscle, circulated in blood
- Excess myostatin associated with muscle wasting, myostatin-mutants have 20-30% increase in muscle mass (Belgian Bull)
Necrosis
- passive, uncontrolled cell death that takes place in response to injury
- When undergoing necrosis, a cell will first swell, and then its membrane will disintegrate such that the cell contents are released into the local environment. This normally triggers a large inflammatory response
Apoptosis
- active, specific, highly regulated, molecular-mediated cell death
- requisite for normal development and tissue function
- in apoptosing cells, both the chromatin and nucleus will condense and fragment, and the cell will shrink. In the final stages, a process called “membrane blebbing” occurs in which portions of the cell protrude and bud off, forming apoptotic bodies. These bodies undergo phagocytosis by neighboring cells or by macrophages. Phagocytosis typically occurs quickly, to avoid an inflammatory response.
- development apoptosis helps to: sculpt tissues, delete structures, adjust cell numbers, eliminate harmful/injured/abnormal cells
Biochemical signalling for apoptosis
- Apoptotic stimuli (DNA damage, elevated p53, radiation, etc) activate BH3 (normally inhibited by Bcl-2) which promotes pore formation in mitochondria.
- Cytochome-C leaks out of mitochondria, bind with Apaf-1 and form the apoptosome.
- apoptosome cleaves the pro-segement in initiator caspases which activate effector caspases, which carry out apoptosis
Extrinsic and intrinsic control of caspase activity
- Extrinsic: pro-death ligands bind to death receptors which cleave and activate initiator caspases, which activate effector caspases
- Intrinstic: intrinsic death signals (radiation, DNA damage, mutations, etc) activate p53 pathway which activate BH3 to create pores in the mitochondria, allowing cytochrome-C to leak out, bind Apaf-1 and form the apoptosome which ativates caspases
Oncogenes
- when overactive these genes promote excess cell number or growth
- only one mutational event is required to gain dominant function an achieve cancerous phenotype
- Ex: Ras gene: if mutated to be permanently on, will increase growth and inhibit apoptosis
Tumor suppressor genes
- genes that would promote excess cell growth or number if inactivated
- Two mutational event needed to destroy tumor suppressor function (both gene copies), causing a recessive loss of function
Caretaker genes in cancer
- genes that maintain the integrity of the genome
- inactivation of these genes increases the chance of a cell to acquire mutations that are cancer promoting.
- two mutational events are required (knock-out both gene copies)
Clonal evolution of human cancers
= the idea that the compounded effects of multiple acquired mutations over time result in an aggressive cancer phenotype.
- Cancer is therefore a multi-step(mutation) process that "selects" for the most aggressively dividing cells
- Ex: stages of colon cancer
66 acquired characteristics requisite for cancer development
- self-sufficiency in growth signals
- insensitivity to anti-growth signals
- tissue invasion and metastasis
- limitless replicative potential (i.e. prevent telomere degradation)
- sustained angiogenesis
- evading apoptosis
Viral origins of cancer (Rous chicken experiment)
- Rous injected ground up tumor cell into healthy chicken, which then developed cancer. Therefore something in the tumor promotes tumorigenesis
- Rous sarcoma virus (RSV) found to carry mutated SRC oncogene that was incorporated into the host genome
Mechanisms of oncogene hyperactivation
- mutation: locked in a active or inactive state
- amplification: so much signal produced that it overwhelms regulatory mechanisms (Ex. Her2/neu receptor in breast cancer, activates Ras)
- translocation of the gene to an area of a strong enhancer/promoter (ex: Bcl-2 in Bcell lymphoma, inhibits apoptosis)
- translocation to create fusion protein with new function
- proviral insertion
Discovery of tumor suppressors (Stanbridge experiment in 1976)
- Clones of Hela tumor cells were implanted into mice.
- When normal human fibroblast cells (or hybrids with the Hela cells) were injected, no tumor observed--tumor suppressor active in normal tissue
- When chromosome 11 ejected from hybrids, tumor reformed--tumor suppressor on chromosome 11
Knudon's "2-Hit" hypothesis for cancerous mutations in tumor suppressor genes, especially Rb
- Loss of function mutations must occur in both gene copies (hence 2 hits) to get cancer
- In retinoblastoma predisposition for the disease is inherited, not the actual disease
--children with family history likely inherited 1 mutation, so achieved the second mutation earlier (and more likely in both eyes)
--in children w/o family history (sporadic), accumulated both mutations, so tumors later and mostly in 1 eye only
Premises underlying the use of simple organism as models in medical research
- most of the important biological processes have remained essentially unchanged throughout evolution (conserved in humans and simpler organisms)
- Conserved processes are easier to unravel in simple organism than in humans--humans are crappy model systems (expensive, old, complex)
Hippo/Mst-2 Tumor suppressor pathway
- highly conserved in flies and humans (associated with human cancer)
- When Hippo is deleted, tumors arise in flies.
-Crb (Crumbs) is an upstream regulator of the Hippo/Mst-2 pathway which also may be regulated by AIB-1 (amplified-in-breast-cancer-1)
Translating cancer research in flies to treatments in humans
- identify candidate targets for rational drug design
- facilitate development of targeted vs. general therapies
- genotype tumors to diagnose dependence of pathways and thus responsiveness to drugs
- tailor drugs to cancer genotypes
Loose (areolar) connective tissue (organization, location, function)
O: few cells, dispersed between disorganized fibers and abundant ground substance
L: beneath epithelia, between epithelium and muscle, serous membranes
F: holds blood supply, media for nutrient supply, supports epithelium, area for immune action
Dense Irregular connective tissue (organization, location, function)
O: bundled collagen fibers w/o specific orientation. Less cells and ground substance
L: capsules of organs (liver, spleen, lymph nodes, gonads), dermis
F: provide mechanical support and integrity
Dense regular connective tissue
(organization, location, function)
O: abundant collagen fibers, arranged in the direction of tension, cells are scarce w/ flattened nuclei
L: tendon, ligament, cornea
F: mechanical coordination and continuity between bone/muscle and bone/bone joints
Fibroblast
- "fiber forming cell"
- most abundant cell type in CT
- synthesizes ECM: collagen, elastin, proteoglycans, associative proteins
- organizes fibrils via membrane extensions surrounding them
- repairs damaged tissue (via metalloproteases)
Reticular Cell
- specialized fibroblast, produces reticular fibers of collagen III
- Reticular fibers form an elaborate network which interstitial fluid and wandering cells pass through.
- Reticular cells in lymph CT also participate in antigen presentation
Unilocular adipose tissue
- main adipose tissue in adults, often yellow
- has one central lipid droplet that occupies most of the cell. Nuclei are flattened and peripheral
Multilocular adipose tissue
- limited distribution in adults, common in newborns and hibernating animals, brown due to lots of mitochondria and blood
- multiple lipid droplets, central nucleus, lots of mitochondria
- used to produce heat by uncoupling ETC in mitochondria
-
Ground substance/Proteoglycans
- viscous, gel like substance in CT, water and ion reservoir
- fills the space between CT fibers, provides tissue resilience, lubrication/barrier, modulates fiber assembly and cell signaling
Proteoglycan composition and function
- Central protein core with several side chains of glycosaminoglycans, which vary in number and type
- The sugars forming the polymers can be modified. These modifications increase the negative charge of the side chains
-Negative charge of the side chains allow the complex to sequester water and ions and provides a repelling force that restores tissue shape when compressed
Collagen Fibers
- most abundant protein in the body
- confers tissue strength and rigidity
- many types, some form fibrils, others form meshes or associate and aid bundling of fibrils
- ex: CI in bone, tendon, skin, fibrous cartilage; CII in cartilage, CIII in loose CT, organs, blood vessels (elastin); CIV basement membrane
- multiple diseases associated with collagen
Collagen structure
- composed of 3 α helical subunits coiled around each other.
- in each sequence, Gly is every 3rd residue, which interact in the center of the helix complex, stabilizing it
Collagen synthesis
- synthesized as a monomer in the RER, modified by lysyl or hyroxylases (Vit C, Fe, O2 dependent) in ER lumen to form alpha helices
- interactions at the C-terminus ends drives assembly of the triple helix (disufide bonds) in the rest of molecule
- assembled subunits are secreted, C and N terminus domains are cleaved, remaining central rod spontaneously assembles into fibrils with others (stabilized by cross-linker, lysyl oxidase (Cu dependent))
- in fibers, subunits are staggered, leaving a lacunar region (impt for bone mineralization), creating banding
Reticular fibers
- made from collagen III, thin and branched
- stains with silver
- forms stable framework in organs
Elastic fibers
- confers elasticity and resilience to tissues, esp arteries, lungs, skin and ligaments/tendons
- elastin monomers are hyropobic and so resist expansion, will recoil
- lysyl residues are crosslinked by lysyl oxidase and condensed into desmosine crosslinks
- elastic fibers are polymerized on the fibrilin scaffold secreted by fibroblasts. Microfibrils surround elastin, and limit stretching
Associative proteins in CT
- glycoproteins which associate with CT fibers in the extracellular matrix and to cellular receptors (integrins)
- abundant in the basement membrane
- ex: fribronectin, laminin, tenascin, entactin
Basement membrane/basal lamina
- structural attachment site for overlaying epithelia and underlying connective tissue
- synthesized by epithelial cells
- rich in associative proteins binding to integrins
- Function: cell bonding & scaffolding, barrier & filtration, polarity cue fro epithelia, compartmentalization
Integrins
- transmembrane receptors. Extracellular domain anchors proteins to the cell's actin cyroskeleton.
- cells are mechanically connected to the ECM
Function of Connective tissue
- support and packing
- transport
- storage
- defense
- repair
Pathophysiology for CT diseases
• Haploinsuficiency: stop codon, null allele leads to loss of function, reduced secretion of wild type protein.
• Dominant Negative effects: mutations alter interaction with other protein, reduce stability or affect function.
- Enzymatic defects: compromises morphology and strength. Ex: mutations in enzymes involved in collagen synthesis, Brittle Bone syndrome
• Clinical consequences depend on function and location.
Osteoblasts
- bone forming cells, found on bone surface
- synthesize and secrete organic components of bone matrix and release factors aiding in mineralization
- become incorporated into bone as osteocytes
- when inactive, called bone lining cells
Osteocytes
- bone cells
- housed in cavities called lacuna with projections (canaliculi) to other cavities
- cell membrane extensions (processes) overlap in the canaliculi and transfer nutrients and signals via gap junctions
Osteoclasts
- bone reabsorbing/refoming cells
- multinucleated, migrate from blood vessels to bone, form a ruffled border to increase surface area.
- secrete HCl and proteases to decalcify and breakdown bone matrix
- creates Howship lacunae
Bone matrix mineralization
- composed of 70% mineral. Stores calcium as hydroxyapatite
- can occur directly, or by replacement of collagen precursors (endochondrial ossification)
- minerals released by osteoblasts (and chondrocytes, if in cartilage) as well as enzymes to disable inhibitors and recruit calcium
- minerals occupy the gap sites in collagen first then progress outwards