• 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

1/517

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;

517 Cards in this Set

  • Front
  • Back
LIGHT MICROSCOPE or bright field microscope
Resolution: minimum distance between 2 closely positioned objects
where 2 objects detected as separate entities (quality of the glass prisms will
impact resolution) – the ability to distinguish two distinct objects as two distinct objects.
a. Wavelength of illuminating source
b. Numerical aperture (NA)
2. Pathway of Light
a. ROYGBIV
What's the path of a Light Microscope
Light source is the illuminator --> condenser (focuses)  sample (stage is where the specimen is located) --> passes through/absorbed --> objective lens (magnify 4x-100x) --> ocular lens --> retina
What microscope is good for living cells and tissues?
Phase contrast microscope
Differential Interference Contrast Microscope
similar to phase contrast, but uses additional prisms and polarizers to generate contrast in fixed or living specimens; 3D image
What's a good microscope for studying proteins?
Fluorescent Microscope
Birefringence
refractive index dependent on the polarization/direction of light
What microscope is good for oriented molecules such as collagen and HAP?
Polarization Microscope
Fluorochromes
labeling fluorescing compounds – usually a tagged antibody is used for this.
1. Fixation
a. Preservation of structure
b. Cross link macromolecules together --> freeze tissue in near native configuration
c. Formalin (37% formaldehyde): cross-linking of amino acids without precipitation
d. Artifacts: shrinkage
Describe.
2. Decalicification
3. Dehydration
4. Clearing
2. Decalcification: removal of calcium
3. Dehydration: removal of water by alcohols more and more % alcohol up to 100%
4. Clearing: replacement of alcohols with xylene (more complex organic solvents), etc. tissue becomes transparent in xylene
5. Embedding
paraffin wax block with tissue inside, plastic resins
6. Sectioning
using a machine called a microtome, or cryostat (freezing microtome) – tissue is slowly moved further out and get sliced and put onto a slide.
Rehydration
(reverse the process and decrease percent alcohol step by step to prep for staining)
Basophilia Stain
Hematoxylin stain negatively charged things like DNA and RNA - bluish tint known as basophilic.
Acidophilia
1. Tissue stains more readily with acid dyes like eosin Binds to positive charged items (E): cytoplasm
2. Acid dye carries (-) charge which binds to (+) charged molecules
Metachromasia
change of dye color when reacting with tissue components – components that stain purple with toluidine blue are known as Metachromatic.
Sudan black
for lipids
PAS
glycoproteins and other carbohydrate-rich molecules forms a magenta precipitate.
Direct Method
Indirect Method (uses a fluorescent antibody)
Direct and indirect methods of immunocytochemistry. Left, An antibody against the antigen was labeled with a fluorescent dye and viewed with a fluorescent microscope. The fluorescence occurs only over the location of the antibody. Right, Fluorescent-labeled antibodies are prepared against an antibody that reacts with a particular antigen. When viewed with fluorescent microscopy, the region of fluorescence represents the location of the antibody.
IMMUNOFLUORESCENT MICROSCOPY
1. Fluorescent molecules attached to antibodies
2. Excitation and emission of light waves
What Microscope:

1. Light source: electrons from heated tungsten filament
2. Electromagnetic coils: lenses
3. Sample Preparation: limited ability of electrons to penetrate biological molecules
a. Fixation
1) Glutaraldehyde: cross links proteins
2) Osmium tetroxide: stabilizes lipids and proteins
b. Use of uranium and lead (stains): increase contrast
 
This is coupled to a computer
Transmission Electron Microscope (TEM)
SCANNING ELECTRON MICROSCOPE (SEM)
Fixed sample coated with thin film of heavy metal atoms (like uranium and lead to stain the tissue to withstand the electron beam.)
Electrons pass over surface
3. Good to study Cell surface features of bone, enamel, dentin: cell surface plus metal
FREEZE FRACTURE (CRYOFRACTURE) EM
1. Rapid freezing – liquid Nitrogen and fracture the surface
2. Fracturing tissue
3. Coating fractured tissue with platinum
4. Study of metal replica – good for studying membrane type structures
AUTORADIOGRAPHY - PULSE CHASE STUDIES
1. Use of radioisotopes
2. Trace chemical pathways in cells: good for studying metabolism of proteins, carbohydrates, lipids, nucleic acids. Measure the amount of radioactivity.
TISSUE CULTURE
1. Organ and cell culture
2. Use of microelectrodes: ion concentrations, injections of antibodies and fluorescent dyes
3. Cell differentiation
RECOMBINANT DNA TECHNOLOGY
1. Manipulation of proteins, DNA, etc.
2. Replacement of damaged genes
What is the embryonic origin of epithelium?
Mainly from Ectoderm and Endoderm, but also Mesoderm

Ectoderm– Skin and its appendages (hair and sweat glands), mammary glands
Endoderm–Liver, pancreases, GI and respiratory tracts
Mesoderm–Reproductive system, Blood vessels and body cavities
Epithelium comes As a sheet of closely apposed cells, separating a space (lumen) from underlying tissue.
but also as __________ of invaginated epithelium cells
gland
What are the basic functions of epithelium?
Protection (Skin)
Absorption and Transcellular transport (intestinal epithelium)
Secretory (glandular epithelium)
Sensation (olfactory neuroepithelium)
Contractibility (myoepithelial cells)
What tissue is found here:

pulmonary alveoli
blood and lymph vessels
pleural
peritoneal cavities
simple squamous
What tissue is found here:

ducts of many glands
Ovary
Some kidney tubules
simple cuboidal
What tissue is found here:

digestive tract
Gallbladder
large ducts of glands
simple columnar
esophagus
stratified squamous non keratinized
ducts of the sweat glands
stratified cuboidal
Goblet cells (mucus-secreting cells) are commonly found in? columnar and pseudostratified epithelium
columnar and pseudostratified epitheliu
Stratified transitional epithelium
Ureters, Bladder,
Proximal urethra-organs
Collectively, microvilli borders: brush-like or striated by light microscopy.

Increase intervillar spaces and facilitate?
absorption/secretion
Cilia have 9 microtubule doublets + 2 central microtubule “spokes” that make up what is known as?
Ciliary Axonemes and Cilia is found in the lungs, oviduct, and vestibular apparatus and are hair-like Propelling substances over the surface of the epithelium via rapid rhythmic oscillations Or sensention
Stereocilium
Found in the epididymus. Increase surface area and facilitating the movement of molecules
Keratinization
Protection from Abrasion or Desiccation waterproof ie. palm of hand
What are some functions of intercellular Junctions?
Seals (occluding junctions - zonula occludens) claudins - Seals to prevent the flow of materials between the cells

Adhesion (adhesive or anchoring junctions)

Communication (gap junctions)
What adheres epithelial cells to Basal Lamina?
hemidesmosome

desmisomes have intermediate filaments
What comprises a junctional complex?
A junctional complex:

a zonula occludens,

a zonula adherens and

a desmosome.
A gap junction has connexons and what are they comprised of?
6-connexins
Basal Lamina
Extracellular material between the epithelial basal surface and underlying connective tissue

Also found between adjacent epithelial cells
Reticular fibers secreted by connective tissue cells (reticular lamina) may be closely associated with the?
basal lamina.
What are the components that make the basal lamina?
Composed mainly of type IV
collagen, laminin and
heparin sulfate (a proteoglycan

Note. BL are only visible by electron microscopy
What is the function of Basal Lamina?
Barrier: Filtration –Kidney

Cell-to-cell interactions:
Attachment to connective tissue
Receptors (Integrins) attach to BL components

Compartmentalization or separation

Polarity

Tissue scaffolding: development and regeneration
The basement membrane is a thicker structure visible by light microscopy which is composed of either:
two fused basal laminae ( or a?
basal lamina and a reticular lamina
In the Kidney:
BM of several tubules and of the structures within single glomerulus Function to?
Support and filter
Focal basal cell layer disruptions occur in?
cancers/ carcinomas
Metaplasia
is the reversible replacement of one differentiated cell type with another mature differentiated cell type. The change from one type of cell to another may generally be a part of normal maturation process or caused by some sort of abnormal stimulus. In simplistic terms, it is as if the original cells are not robust enough to withstand the new environment, and so they change into another type more suited to the new environment.
Glands
Formed by epithelial cells that secrete a fluid of different composition than blood or intercellular fluid.

Secreted substances can be ions, secretory polypeptides or proteins, lipids or glycoproteins.
Secretory Granules
secreted are generally stored in the glandular epithelia
in small membrane-bound vesicles
Ex. Parotid salivary gland
cytokines
cell to cell communication

Distance between secretion and target:


Autocrine: target self
Paracrine: target cell in vicinity
Endocrine: far, blood/lymph
Autocrine
target self
Paracrine
target cell in vicinity
Endocrine
Far, blood/lymph
What are the two glandular secretion pathways?
Constitutive: make and release no signaling molecules
Regulated: concentrate and store signaling for releasing (Neural/endocrine)
Exocrine glands
have ducts
What are the two structural types of multicellular exocrine glands?
Simple glands: one unbranched duct. : Secretory portion may be tubular, coiled tubular, branched tubular, or acinar (Flask-shaped).

2. Compound glands: more than one duct that branches repeatably. Secretory portion may be tubular, acinar or tubuloacinar.
Merocrine
Exocrine gland that functions via exocytosis w/o loss of cytoplasm. It's the most common form of secretion.
what regulates exocytosis in salivary acinar cells?
cAMP and Ca
Holocrine exocrine gland functions by?
the whole cell is a discharge
Sebaceous gland
Sebaceous gland
simple, branched, acinar glands
holocrine mode of secretion
Apocrine
Apocrine: secretory product and a portion of the cytoplasm
Ex. mammary glands and specialized sweat glands
Serous producing cells
Serous producing cells- watery secretions; usually intensely stained with eosin; nuclei usually rounded or oval.

Glycoproteins:
N-linked to the β-amide of asparagine .
Mucous producing cells
Mucous producing cells- viscous, slimy secretions; appear empty by routine H/E; nuclei often flattened against base of cell

Glycoproteins: :
O-linked to the hydroxyl groups of serine or threonine
What cell surounds the acini?
Myoepithelial cells
Anesthetic Diffusion
Absorbed by non-neuronal tissue
Diluted by interstitial fluid
Removed by capillaries and lymphatics
Hydrolyzed-inactivated
Teeth is embedded in alveolar bone a type of?
CT and by PDL another CT
What are the functions of CT?
Connects, Cushions, Supports, Fills space
Protects
bone protects underlying organs
mast cells-inflammation
plasma cells- antibodies
phagocytes- engulf foreign substances
Barrier under epithelium – protects from the outside world. Anus/esophagus epithelium
Contains nerves- sensation
Contains blood vessels, lymphatics
nutrient, waste, gas exchange
CT comprises?
Cells
Fixed and wandering
Fibers
collagen, elastic
Ground substance
GAGs, proteoglycans, adhesive proteins
Tissue fluid
Biological Considerations
Mesenchyme
Mesenchyme cells --> these give rise to the fiberblasts which are the work horse and they produce and gives rise to adipocytes, etc.
Fibroblast
Synthesize extracellular matrices such as:
Fibers
Collagen
Elastin
They synthesize the Ground substance such as:
Glycosaminoglycans
Proteoglycan
Synthesize growth factors
influence growth and differentiation
Lots of interactions between CT and epithelium
What are the differences between a fibroblast and a fibrocyte?
Fibroblast- active
abundant, irregularly branched cytoplasm
ovoid, large pale staining nucleus
well developed RER, golgi – because these are making proteins that they are going to secrete
Fibrocyte- inactive (quiescent)
it becomes spindle shaped, few cell processes
it becomes smaller, darker, elongated nucleus
only a small amount of RER
Fibroblasts make?
collagen fibers
What is the major cell type in CT?
fibroblast - thin elongated cell - spindle shaped
Adipocyte
It Stores lipids
Energy supply
Offers Padding, protection, shock absorber
Insulation
Generation of heat (brown fat)
Visceral fat has a Endocrine function – have to do a lot with inflammation
Adipocytes develop from?
Mesenchyme --> lipoblast -->
multilocular/unilocular adipocyte
Unilocular is a?
white fat cell
One lipid droplet (white-yellow, carotenoids)
polyhedral shaped (50-150 um) Large
signet ring cell- lipids dissolved in routine histology cell with vacuole and eccentric flattened nucleus

More overweight someone is the larger the adipose cells become
Multilocular is?
a brown fat cell


many lipid droplets and mitochondria
color due to blood vessels and mitochondria with cytochromes
polygonal, smaller than unilocular
heat producing
newborn
adults


Newborns don’t have a lot of protection so they need a way to survive and the brown fat is heat producing to keep themselves warm

Recently brown fat found in Adults, which set up a race with pharmacompanies to figure it out and what they could do with it.

PET scan inject radioactive compound … analog of glucose. Taken up by actively metabolizing cells. PET machine can then detect this activity
Fat as an Endocrine organ.
Macrophages surrounded by fat cells are secreting a number of biologically active materials/fa
Endocrine function of fat changes with an increase in obesity. The cells give off a different cascade of molecules. Leptin is up. The fat cells produce a great number of bioactive material Ex. Leptin

TNF alpha lowers adiponectin??

Endothelial cells let more macrophages in
Q: T/F In the model Model linking periodontitis and obesity with inflammatory related chronic diseases, if a patients RER/Golgi had a mutation that impaired secretion of newly synthesized proteins, this would not effect periodontitis.
FALSE
Q: Your patient is borderline obese and has high levels of obesity related inlfammaotory cytokines. You recommend intense periodontal therapy. Based on our proposed model of obesity and periodontal disease, this recommendation does what?
aims to lower biologies that relate both obesity and periodontal disease.
Macrophage
Phagocytic
antigen presenting
long living
fixed in the tissues and wandering/can wander and travel in the blood stream
Mononuclear Phagocyte System – very important in the normal signaling of adipose tissue
What is the precursor to the macrophage?
monocyte - will move from the blood stream to the tissue and will differentiate into a macrophage
Kupffer cell
liver
microglia
nervous tissue
Langerhans cells
Skin - Antigen presentation
Phagocytosis
Lysosome will fusen with a phagosome and the lysosomal enzymes will kill the microbes
Opsinization
Phagocytosis increased when the foreign material/microorganism is covered by antibodies or its antigens bind complement.

This enhances the ability of it to be phagocytized.
Q You have decided to be an academic dentist who will study the link between obesity and periodontal disease. You receive a link from Life Line Cell Tech that they are selling pre-adipocytes. In the lab, if they are differentiated into adipocytes and in the presence of oral bact. Secrete obesity related inflammatory cytokines, will they be a good model for you to use?
Yes
Giant cell
A giant cell is a mass formed by the union of several distinct cells (usually macrophages). It can arise in response to an infection, such as from tuberculosis, herpes, or HIV, or foreign body.
Name a cell that is metachromatic.
Mast cell
Mast Cell
oval to round
IgE on surface
filled with metachromatic secretory granules that are filled with:
histamine
heparin
neutral proteases
ECF-A (eosinophil chemotactic factor of anaphylaxis)
NCF (neutrophil chemotactic factor)
leukotrienes
prostaglandins
cytokines
How do the secretions of granules in Mast cells work?
Antigen binds to IgE receptor
a Calcium event occurs and there is a fusion of these granules and a release

ECF for eosinophils
NCF for neutrophils, drawing neutrophils to the area

Other pathway converts Arachidonic acid and secretes Leukotrines and Thromboxanes and Prostaglandyns
What happens during anaphylactic shock?
all of the bodies in mast cells degranulate and you have a real life and death situation
Plasma Cells arise from?
arise from B-lymphocytes
ovoid, basophilic
well developed rer
juxtanuclear golgi secretes antibodies
Leukocytes - white blood cells
migrate from blood
increase in inflammation
Neutrophil – kill bacteria by phago
Eosinophil – kill parasitic infection
Basophil – analogous to mast cells
lymphocyte
monocyte – precursor to macrophage
Eosinophil
WBC that helps with parasitic bacteria
Pericyte
surface of capillaries and veinules
rounded cell that wraps around
contractile- involved in blood flow control
very important in wound healing- gives rise to connective tissue and blood vessels (i.e undifferentiated stem cell-like)
Various Stem Cells
MSCs- multipotent mesenchymal stem cells
bone marrow, adipose tissue, umbilical cord blood
Give rise to different cell lineages
Regenerative medicine, therapy
Dental MSCs
Name the following multi potent stem cells:

DPSCs
SHED
PDLSCs
SCAP
DFPCs
DPSCs- post natal dental pulp stem cells
SHED- stem cells from exfoliated deciduous teeth
PDLSCs- periodontal ligament stem cells
SCAP- stem cells from apical papilla
DFPCs- dental follicle precursor cells

Rootword for formation potential: Osteogenic, dentinogenic, cementogenic, adipogenic, chondrogenic, myogenic, neurogenic
What are the cells found in Areolar CT 1?
Fibroblasts, Mesenchyme, Adipocyte
What are the cells found in Areolar CT 2?
Mast cells, Macrophage, lymphocyte, Plasma cell
Name the 3 CT fibers.
Collagen, Reticular, Elastic
Name Collagen that forms long fibrils.
Types 1,2,3,5, and 11
Fibril associated collagens do what?
bind collagen fibers together.

Types 9, 12, 14
Network forming collagens
forms basal lamina
type 4, 8, and 10
Type 7 collagen forms what?
anchoring fibrils. binding collagen fibers to basal lamina
Structure of a collagen fibril.
overlapping array
Osteogenesis Imperfecta
Change in one nucleotide gene for Type 1. spontaneous fractures
Scurvy
lack of Vit C --> ulceration of gums, hemorrhages
Reticular fibers are what type of collagen.
Type III


thinner and more carbohydrate than type I fiber
stains black with silver stain
produced by reticular cell
they produce a scaffolding network
Elastic Fibers
Can stretch 150% of length
Fiber composed of
Outer sheath- microfibrils of fibrillin (glycoprotein)
Surrounds Inner core- amorphous core of elastin

Elastic fibers are required in areas that need to expand and contract Ex. Blood vessels
Marfan Syndrome
mutation in fibrillin, defects in a protein
ECM interactions regulating TGF beta
TGF beta gets up regulated. – disregulation of growth factor – gets embedded in the CT can become a sink or a source for growth factors.

They put mutation into the mouse they were able to treat the mouse with pharmaceuticals, they were able to reverse the pathology of Maphan Syndrome, which led to the next phase: clinical trials.

Thrombospondin
Ground Substance
are complex of macromolecules
they fill the space between fibers and cells
organizes tissue topography, supports cell migration, orients cells, induces cell behavior
Can bind growth factors, cytokines-reservoir
3 classes of compounds
Glycosaminoglycans
Proteoglycans
Multiadhesive glycoproteins.

Fibronectin is a G.S. protein
GAGs
repeating dissacharide of uronic acid and sugar
hydrophilic polyanions (can bind cations such as sodium)
part of Proteoglycan
very important in regulating osmotic pressure leads to increased water content- firmness, flexibility
Proteoglycans
Protein core + GAGs (coming off of them can attract water)

GAG molecules are found within CT Ex. Hyaluronic acid
What is the major protein of basement membranes?
Type IV Collagen
Mulitadhesive glycoproteins
is a globular protein with carbohydrate
Very functional in cell-cell and cell-substrate interactions
What are the Two major multi adhesive glycoproteins?
Fibronectin – can bind cells, collagens, and GAGs
laminin - can also bind cells, collagens, and GAGs, basement membrane
Fibronectin - Laminin
binding domains
cells
collagens
GAGs
Tissue Fluid
similar to blood plasma in ions
similar to blood plasma in diffusable substances
contains low molecular weight plasma proteins
circulation to feed cells and remove waste
Arterial side, when the hydrostatic pressure is greater than the Osmotic pressure, water then goes where?
leaves the venules to the arterial side. It works both ways and goes back and forth
Tissue Fluid Movement
hydrostatic pressure from blood
water leaves capillary

colloid osmotic pressure- plasma proteins
water enters capillary
Edema
increase in tissue fluid, i.e. swelling
Areolar CT
Fills space between muscle, supports epithelial tissue, sheaths lymphatics and blood vessels
fibroblasts and macrophages most numerous- contains numerous other all cell types
flexible, well vascularized- loose consistency
Name the two types of Dense CT.
Irregular collagen fiber bundles- no orientation
3-d network- resists stress all directions

Regular collagen fiber bundles - in a pattern
collagen fibers aligned linearly with fibroblasts
Dense CT
less flexible & more resistant than loose
offers resistance and protection
fewer cell types
mostly collagen
What CT might you find in tendon?
Dense regular CT
Elastic CT
bundles of parallel elastic fibers
between is thin collagen fibers, fibroblasts
yellowish color
occurs infrequently
found in some ligaments
vertebral column, penis
Reticular CT
loose CT of reticular fibers and specialized fibroblasts (reticular cells)
forms a 3-d network to support cells
reticular cells cover fibers and ground substance producing sponge-like structure
produces microenvironment for hematopoietic and lymphoid organs (found in bone marrow, lymph nodes, spleen)

**Stains silver - lymphoid tissue, spleen
Mucous CT
in umbilical cord; pulp of young teeth
abundance of ground substance
primarily hyaluronic acid
Wharton‘s jelly (umbilical cord)
cells- mainly fibroblasts
Adipose Tissue
White (unilocular) and brown (multilocular)
cells are packed and subdivided into lobules with loose CT sheaths
make lipoprotein lipase- transported to luminal surface of endothelial cells to hydrolyze chylomicrons
endocrine organ
Lipid Transport
Adipocyte <----> Capillary
Can epithelium be influenced by underlying CT?
Yes it can.

Heterotransplants of epithelium/CT- normal
tongue ep/buccal CT- tongue-like differentiation
Heterotransplants of epithelium/CT- altered
palate ep/buccal CT produces buccal-like keratinization (differentiation)***
changed, it didn’t stay palate went to buccal
palatal or buccal ep/tongue CT produces tongue-like epithelium
Wound Healing Surgical Incision
Incisional space fills clotted blood, fibrin, blood cells; dehydration forms scab
<24 hrs- neutrophils appear (kill bacteria)
by 72 hrs- macrophages replace neutrophils
injest proteolytic debris
provide growth factors to stimulate fibroblast growth, collagen secretion, neovascularization
When do the neutrophils show up in wound healing?
The 1st 24 hours to kill bacteria.

by 72 hrs- macrophages replace neutrophils
injest proteolytic debris
Inflammation
Complex reaction to injuries
fluid and leukocyte leakage; systemic responses
Leads to repair
Protects
When inflammation goes wrong --> Basis of many chronic diseases
rheumatoid arthritis; hypersensitivity

Blood vessels get leaky during inflammatory response. Increase in blood flow. Neutrophils get drawn in in case of bacterial infections. The body is set to draw in the types of cells it needs in order to repair itself.
When inflammation goes wrong it is the Basis of many chronic diseases. Name one.
rheumatoid arthritis; hypersensitivity
What's the difference between acute and chronic inflammation?
Acute- short duration; leakage, extravazation

Chronic- longer duration; cell based; neovascularization, fibrosis, necrosis
What is the differentiating pathway of an Osteoprogenitor?
O.P. --> Preosteoblast --> Osteoblast --> Ostocyte
Osteoprogenitors
OSTEOPROGENITORS
1. Extensive proliferation: growth, repair, re-organization
2. Near free bone surfaces, few cell layers from active osteoblasts
3. Alkaline phosphatase negative
Pre-osteoblasts
PRE-OSTEOBLASTS
1. Limited proliferation (some mitotic ability) and this leads to a more differentiated cell
2. Alkaline phosphatase positive
In an Osteoclast the ruffled border represents what?
where the acids and enzymes are released.
Intramembranous Bone Formation
1. Mesenchyme -->lead to osteoblasts
2. Woven bone - is primary spongiosa – weak bone
3. Mature spongy bone - secondary spongiosa --> part of this then becomes --> CB
4. Compact bone
What process will get rid of
the hyaline cartilage and woven
bone to make way for the newly
synthesized spongy bone?
Absorption process
OSTEOBLASTS (mature cell) – do not undergo mitosis at all.
1. Synthesize bone (collagen type I & non collagenous proteins)
2. Strongly positive for alkaline phosphatase (bone formation index)
3. Functions
a. Secretion --> bone matrix (osteoid) as well as beginning of osteoid degradation
b. Osteoid (uncalcified bone matrix) resorption
c. Osteoclast stimulating factor(s)
Bone lining cells are?
“resting” osteoblasts – resting cell. – more elongated flattened cells lying on the outside of trabelcula.
Osteocytes
1. Osteoblasts that become trapped in their own matrix
2. Important mechanosensory (mechanotransduction) role, maintain bone matrix
3. Form a syncytium through gap junctions between their processes: called connexins
Osteoclasts
1. Bone resorption by osteoclast tightly coupled with bone formation by osteoblast
Large and multinucleated (4-20 nuclei)
3. Bone marrow-derived (monocyte-macrophage lineage / CFU-GM)
5. Numerous large lysosomes, abundant mitochondria, well developed Golgi, some RER
What cell type is found in Howship's Lacuna?
Osteoclasts.


7. Sealing/adhesion /circumferential zone: attachment to mineralized bone matrix
8. Ruffled border: cell membrane thrown into many folds representing site of active resorption
9. Receptors for calcitonin, but not for PTH, Vitamin D3, or cytokines
What is the osteoclasts mechanism?
Osteoclast mechanism: GMP/CFU-GM  MCS-F binds to receptor on macrophage --> osteoclast precursor  induction of RANK on precursor -->RANK interacts with RANKL produced by stroma cells --> RANKL binds to RANK receptor on osteoclast precursor -->fully activated osteoclast --> bone resorption
OPG (Osteoptrotogera) prevents what?
Osteoclast formation
RANK receptor + RANK =
Osteoclast formation
RANK + OPG =
prevents osteoclast formation
Osteocytes
"professional" mechanosensory cells
Osteoclasts
have calcitonin receptors
What is:

Large, multinucleated
Monocyte precursor
Howship’s lacunae
Ruffled border
Sealing zone
Calcitonin receptors
Bone resorption
Osteoclast
Zone of rest or reserve
chondorcytes random
Zone of proliferation
mitosis, isogenous groups, beginning of matrix synthesis
Zone of maturation
completion of matrix synthesis, partial glycoprotein breakdown
Zone of hypertrophy
– enlargement of lacunae
Zone of provisional calcification
matrix mineralization
Calcium brought into hyaline cartilage matrix through periosteal buds --> calcifies --> limited diffusion of nutrients --> apoptosis (death) of hypertrophic chondrocytes
b. Vertical septae fully calcified, whereas, transverse septae do not calcify
Zone of degeneration
cartilage resorption
Zone of Bone Formation
periosteal buds bring in mesenchymal cells --> osteoblasts --> form bone on cartilage remnants: primary spongiosa (woven bone) --> resorption and disappearance --> secondary (lamellar) spongiosa
Secondary Ossification center
Epiphyseal plate
What ends secondary growth?
When the epiphyseal plate disappears
Cartilage is elongated as a result of?
Mitotic activity
How does the osteoid layer become ossified?
Ca with osteoblasts so that gets calcified first.
Bone remodeling of Spongy and Compact with Rubin's Path ARF (ARRFR)
1. Process by which bone grows and is turned over
2. No net gain or loss in bone mass
3. ARF Sequence Phases (ARRFR)
a. Activation (1 week)
b. Resorption (2 weeks) - osteoclasts go to work
c. Reversal (1.5 weeks) – osteoclasts go away and then the osteoblasts come in.
d. Formation (13.5 weeks)
Monocytes in BVs give rise to?
osteoclasts
Modeling is
net gain in bone
What are the bone remodeling regulation players?
Calcium and phosphate
PTH – stimulates osteoclasts to resorb bone – acts on the kidney tubules to increase absorption.
Vitamin D – calcium shortage in children - rickets
Calcitonin – stimulates bone formation
Growth and thyroid hormones – they affect all growth processes in the body.
Bone morphogenic proteins
Cytokines
Cell-cell communication
b. Interleukins (IL-1, IL-6): osteoclast recruitmen
FGF
FGF: neurovascularization and wound healing, stimulation of osteoblast precursor cells
d. PDGF: wound repair stimulates bone resorption and osetoblast precursors
e. IGFI and IGFII: ↑ bone collagen matrix and ↓ collagenase levels
BMPs
are osteoinductive
TGFS
stimulates precursor cells of osteoblast lineage and on bone collagen formation
Insulin has a direct effect on what bone cell?
Osteoblast
Glucocorticoids have what effect on bone?
inhibition of bone resorption and collagen degradation
coupling
– formation with resorption ie osteoblast/osteoclast
Long bone is completely made by
hyaline cartilage which is later replaced by bone
nucleus pulposis
is the intervertebral disc of a spinal cord
What are the functions of bone?
Functions: skeletal support, skeletal muscle attachment, protective (skull around brain), metabolic, hematopoietic organ, dynamic material
When one part of the mandible is shorter than the other.

Device is screwed into the bone and it is fractured and the screws adjust to pull the bone apart to allow for more and more growth to occur...this is known as?
Distraction Osteogeneis
Long bone: epiphysis
wider spherical ends
Long bone: diaphysis
shaft
Long bone: metaphysis
cone shaped transitional region
Epiphyseal plate
responsible for growth of long bone
What are the classifications of bones?
BONE CLASSIFICATION
1. Long bones
2. Short bones
3. Flat bones
4. Irregular bones – sphenoid, ethmoid
5. Sesamoid bones – associated with tendons and ligaments giving additional strength. Patella
5. Haversian/Compact/Dense/Cortical bone:
external part of bone
6. Spongy/Cancellous/Trabecular bone:
lattice of branching bony spicules or trabeculae
7. Articular hyaline cartilage:
ends of long bones
9. Periosteum (covers outer surface) and Endosteum: lines?
inner surface: soft CT
10. Skull:
compact bone-inner and outer plates/tables; spongy bone between: diploe
8. Periosteal (external surface) and Endosteal?
(internal surface)
By volume____________ bone here is predominant
By weight ______________ bone is predominant
Spongy; compact
What are the two bone types in long bone?
spongy and compact
Periosteum has what two layers?
a. Outer layer: fibroblastic
b. Inner layer: osteogenic

Also contains Sharpey's fibers
Endosteum
thin single layer of flat squamous-like cells lining the walls of all cavities in the bone
4. Periosteal and Endosteal Circumferential Lamella:
lamellae around the outer and inner circumference of the shaft
5. Osteons/Haversian Systems:
What parts make up a system?
basic structural unit of compact bone
a. Central/Haversian Canal: diameter: 22-110 :m
b. Surrounded by Haversian Lamellae
c. Lacunae: contain osteocytes, filled with extracellular fluid (ECF)
d. Canaliculi: thin channels containing osteocyte cytoplasmic processes
6. Volkmann’s Canals:
transverse/oblique channels without lamellae --> Haversian canals
7. Interstitial Lamellae:
angular fragments of previous concentric and circumferential lamellae
8. Cement lines
a. Arrest lines -
bone formation resumes after an interruption
Cement Lines: b. Reversal lines -
bone formation follows bone resorption.
9. Spongy/Cancellous/Trabeculae bone:
not penetrated by blood vessels: NO osteons
Osteoprogenitor cell -
A mesenchymal cell that differentiates into an osteoblast. Also called preosteoblast.
What is found inside a bone lacuna?
Osteocyte
TYPES OF BONE
1. Primary/Immature/Woven Bone:
very cellular, no organization of collagen, less ground substance and mineral
2. Secondary/Mature/Lamellae/Trabeculae Bone: less cells, highly organized collagen arrangement, more ground substance and mineral
Types of BONE
2. Secondary/Mature/Lamellae/Trabeculae Bone:
less cells, highly organized collagen arrangement, more ground substance and mineral
Interdental septum contains what kind of bone?
spongy bone
Synarthrotic joints - limited or no movement. Please name the various types.
a. Synostosis - bony tissue sutures
b. Synchondrosis - hyaline cartilage
c. Syndesmosis - pubic symphysis
d. Gomphosis – PDL – more of a shock absorber
Diarthrosis joint is freely movable, what layers does it have?
a. Fibrous layer
b. Synovial layer - synoviocytes
What comprises Bone Matrix?
1. Inorganic matrix
a. Hydroxyapatite crystals
b. Hydration shell
c. Mineralization process – nucleation cores
d. Measurement of calcification
1 Osteomalacia
2 Osteitis fibrosa cystica
2. Organic matrix
a. Type I and Type V collagen
b. NCPs
What is the mineral portion of the bone matrix?
1. 65% of dry weight
a. Slender rod-like hydroxyapatite [Ca10 (PO4)6 (OH)2]
b. HAP responsible for hardness and resistance
c. Hydration shell
2. Mineralization Process
3. Measurement of Calcification: fluorescent antibiotic tetracycline
a. Osteomalacia – occurs in Ricketts
b. Osteitis fibrosa cystica -
4. Certain soft tissue susceptible to calcification: ectopic
What is the organic portion of the bone matrix?
ORGANIC MATRIX: 35% of matrix
1. Collagen
a. Type I collagen: 85 to 90% of bone total proteins
b. Bone Type I collagen: triple helical, super coiled, typical 67 nm cross banding
c. Type I collagen in bone greater number of cross-links than in soft tissue
2. Noncollagenous Proteins (NCPs)
a. 10 -15% of total bone protein
b. Bone cells synthesize and secrete as many molecules of NCP as it does collagen
What are the bone attachment molecules?
1 Fibronectin (FN): mediates cell attachment and bone cell spreading; produced by osteoblast
2. Thrombospondin (TSP): mediates adhesion, not spread; binds calcium
3. Osteopontin (OP): role in osteoclast attachment via integrin receptor; binds calcium; produced by osteoblasts and osteoclasts
4. Bone sialoprotein (BSP)
a. In osteoblasts, osteocytes and recently in osteoclasts
b. Bone sialoprotein: role in osteoclast attachment? Role in binding calcium
1 Fibronectin (FN):
mediates cell attachment and bone cell spreading; produced by osteoblast
2. Thrombospondin (TSP): mediates?
adhesion, not spread; binds calcium
3. Osteopontin (OP): role in?
osteoclast attachment via integrin receptor; binds calcium; produced by osteoblasts and osteoclasts
4. Bone sialoprotein (BSP)
a. In osteoblasts, osteocytes and recently in osteoclasts
b. Bone sialoprotein: role in osteoclast attachment? Role in binding calcium
PROTEOGLYCANS
1. Macromolecules that contain GAG side chains attached to a central core protein
2. Chondroitin sulfate: attached to 3 separate core proteins - Versican - Decorin - Biglycan
3. Heparin sulfate: facilitates interaction of osteoblast with extra-cellular macromolecules
2. Chondroitin sulfate:
attached to 3 separate core proteins - Versican - Decorin - Biglycan
3. Heparin sulfate:
facilitates interaction of osteoblast with extra-cellular macromolecules
OSTEOCALCIN [3 (-CARBOXY GLUTAMIC ACID (GLA) CONTAINING PROTEINS)
1. Produced by osteoblasts
2. Regulated by 1,25 dihydroxy vitamin D3
3. Gla confers Ca2+ binding properties
4. Important signal in bone turnover and may act as a chemo-attractant to osteoclast precursors
GROWTH FACTORS
1. Factors synthesized by cells of the osteoblast lineage: fibroblast growth factors (FGFs), insulin-like growth factors (IGFs), and transforming growth factor-ß molecules (TGF-ßs), bone morphogenetic proteins (BMPs), possibly platelet derived growth factor (PDGF)
2. Also synthesized by cells intimately associated with bone, such as chondrocytic cells, endothelial cells, and fibroblasts: PDGF, FGFs, IGFs
3. Predominant growth factors in bone: IGF-II, TGF-$, IGF-I
1. Alkaline phosphatase
a. Used as an index of bone formation in serum
b. May also function in mineralization
2. Osteonectin:
phosphorylated glycoprotein
a. High affinity for binding ionic calcium and hydroxyapatite and collagen
b. May play a role as modulator of mineralization and function in tissue remodeling/bone remodeling
Only cell inside bone is?
an osteocyte
Steps to fracture repair.
1. A fracture hematoma forms
2. A fibrocartilaginous (soft) callous forms
3. A hard bony callous forms
4. The bone is remodeled
What are the 3 types of cartilage?
Hyaline, Elastic, and Fibro
Cartilage
INTRODUCTION
1. Special form of C.T.
2. Semirigid supporting tissue
3. Avascular and aneuralar
4. Types: hyaline, elastic, fibro

Low oxygen type of tissue. Diffusion transfers the nutrients through the gel-like framework.
Where are chondrocytes embedded?
in lacuna
Hyaline and Elastic is what type of collagen?
Type II
Fibro is what type of collagen?
Type I
Hyaline Cartilage
articulating cartilage. Long bone ends (articulating cartilage), Trachea, ribs/costal cartilages, Primary Bronchi and secondary bronchi. Supporting type of tissue as you exhale and inhale.
Elastic Cartilage is found where?
the ear, and epiglottis – it bends
Remnant of the notochord forms the Nucleus pulposis – forms the soft-gelatinous central part of the intervertebral disc. Anulus fibrosis surrounds it and it is?
a fibrocartilage.
Fibrocartilage is located?
between the vertebrae, helps to make up the intervertebral disks. Pubic Symphysis held together by fibrocartilage.
Perichondrium
mitotic layer: outer fibrous and inner chondrogenic layer – surrounds the tissue and acts as a capsule.
chondroblast.
It traps itself in a lacuna, then changes its name to Chondrocyte. Similar to Osteoblast --> Osteocyte

Chondroblasts (chondrogenic cell/chondrocyte)
a. Precursor to and smaller than chondrocyte
b. Well developed organelles
c. Basophilic cells
5. Chondrocytes in lacunae
a. Large cell, well developed organelles
b. Isogenous groups: progeny of one cell
c. Synthesis and maintenance of tissue
Hyaline Cartilage
1. PAS+ and metachromatic with Toluidine Blue
2. Considerable amount of collagen: Types II (dominant), IX, X, XI
GAGs – proteoglycans – aggrecan – chondronectin – the ones with the Sulfur
Gives off the metachromatic properties
4. Water (60-80% wet weight) → for resilience cushioning – in synovial fluid
Is Hyaline Cartilage metachromatic?
Hyaline cartilage is metachromatic, but it can also be stained with PAS because they are high in carbohydrates. Hyaline has tightly and loosely bound water, which is needed for movement areas and it is released into synovial cavity. Because of the sulfur in the cartilage, it doesn’t stain blue it stains reddish.

Mast cells are also metachromatic and will stain blue.
What type is the strongest cartilage?
Type I

Hyaline is Type II, but can resist a intermittant pressure a certain kind of pressure
Besides Type II collagen, what other types are found in Hyaline?
Type IX helps stabilize the cartilage matrix.

Type X as well in bone formation. Major type is Type II though
Pericellular capsule
surrounds the cell, surrounds every lacunae
Difference between Inter-territorial and Territorial matrix.
Inter-territorial matrix is found between the isogenous groups and territorial matrix is within the isogenous groups.
Chondronectin is a?
typical glycoprotein that attaches the chondorcyte to a collagen fibril. They hold things together.
Chondorgenic cells give rise to?
chondroblasts --> matrix is laid down, cell becomes trapped and then you have a chondrocyte.
Elastic cartilage
1. Withstand repeated bending
2. Similar to hyaline cartilage except presence of elastic fibers
3. External ear, external auditory meatus, eustachian tube

Elastic fibers have a different refractive index than the rest of the tissue
Fibrocartilage
FIBROCARTILAGE
1. Poorly defined limits
2. Transition tissue
3. Chondrocytes arranged in rows
4. No perichondrium
5. Good tensile strength
6. Intervertebral disc: nucleus pulposus + annulus fibrosis; pubic symphysis; tendon-bone attachments
What are the Two types of growth of Cartilage?
appositional growth and Interstitial growth

Interstitial growth: expansion from within – we have chondrocytes in the pc of cartilage in side increases the internal dimension. This does not happen in bone except for hyaline cartilage in epiphyseal plate ex. Long bones.

Appositional growth: increase in width of bone, perichondrium required – adding on cartilage from the outside
How is cartilage formed?
1. Mesenchyme --> chondroblasts --> chondrocytes
Interstitial growth:
expansion from within – we have chondrocytes in the pc of cartilage in side increases the internal dimension. This does not happen in bone except for hyaline cartilage in epiphyseal plate ex. Long bones.
Appositional growth:
increase in width of bone, perichondrium required – adding on cartilage from the outside
DEFICIENCIES in the following cause:

1. Vitamin A
2. Vitamin C
3. Vitamin D
1. Protein/Vitamin A --> reduction in epiphyseal plate thickness
2. Vitamin C: scurvy --> epiphyseal plate distortion
3. Vitamin D: rickets --> absence/poor calcification
Skeletal muscle
strong quick discontinuous, voluntary contraction
Cardiac muscle
strong, quick, continuous involuntary contraction
Smooth muscle
Weak, slow, involuntary contraction
What are the 3 types of skeletal muscle fibers?
Red Fibers – Slow Oxidative
White Fibers – Fast Glycolytic
Intermediate – Fast Oxidative/Glycolytic
Sarcosomes
- mitochondria
Myofibrils
– contractile elements
Define the following skeletal CT coverings:

1. Epimysium
2. Perimysium
3. Endomysium
Connective Tissue Coverings:
Epimysium – dense CT around muscle
Perimysium – dense CT around fascicle
Endomysium – delicate CT around fiber
Development of Skeletal Muscle
from Myoblasts
Satellite Cells- source of stem cells in adult muscle that can repair damage
Hypertrophy
increase in cell size
Following damage the myofiber, what happens?
satellite cells are activated to enter the cell cycle and proliferate allowing for expansion of the myogenic cell population (B),
These activated satellite cells are characterized by high expression of MRFsMyoD and Myf5.
The proliferative phase is followed by Myoblast terminal differentiation and characterized by the upregulation of the MRF’sMyogenin and Mrf4 (C)
new myofiber formation (upon innervation) (D)
Sarcomeres
repeating contractile units
A Band
– anisotropic, dark band, composed of thick filaments, myosin
I Band
-- isotropic, light band, thin filaments, actin
Z Line
-- in the center of the I-band, sarcomere is from Z-line to Z-line
H Zone
– in the center of the A-band
M Line
-- in the center of the H Zone
F Actin
– filaments of actin embedded in the Z-line
Tropomyosin
– filamentous protein that fills the groove in F-actin and binds Troponin.
What are the 3 types of Troponin
Troponin
TnT—Troponin subunit that binds tropomyosin
TnC– Troponin subunit that binds calcium
TnI—Troponin subunit that inhibits actin-myosin interaction
α-Actinin
—Actin linking protein located at the Z-line,
Desmin
– Intermediate filament located in muscle
Titin
is the largest known molecule and functions as a molecular spring and is responsible for passive elasticity in SK muscle. It connects the Z-line to M-line
Nebulin binds to
actin and is thought to set the length of the thin filament
Sarcoplasmic Reticulum
—special SER in muscle. Sequesters and releases calcium
What are the components of the SR?
Components
Terminal Cisternae – I band, calciquestrin
Tubular Channels – A band
H Sacs – H zone
T-tubules—penetrations of the cell membrane deep into the muscle cell cytoplasm.
Terminal Cisternae
– I band, calciquestrin
Tubular Channels
– A band
H Sacs
– H zone
T-tubules
—penetrations of the cell membrane deep into the muscle cell cytoplasm.
Contraction of Skeletal Muscle
Contraction
Sliding Filament Mechanism
Motor End Plate
Acetylcholine Receptors – sarcolemma
Role of collagen in Cardiac muscle
collagen I and III that attach to the cardia muscle cells and keep them registered.
Intercalated Disc
At sarcomere ends, connect myocytes

There’s a vertical and horizontal component. It connects one cell to the other cell via a fascia adherens. It replaces the Z-line, which is also made of actin. Then it goes through the gap junctions and goes through another z-line.

Principle intermediate filament for muscle is desmin
The principle intermediate filament for muscle is?
desmin
Perkinji cells.
are 5 – 6 times bigger than a cardiac muscle cells.
close to endocardium, specialized cardiac muscle cells
Cardiac Muscle
– Muscle cells
Cytoplasm– Full of organelles, myofibrils
Striations – not as distinct as in skeletal muscle
Nuclei – centrally located
Myofibrils- not as uniformly distributed as skeletal
Mitochondria– abundant and interspersed in cytoplasm
Branching and Anastomosing of Fibers
Sarcoplasmic Reticulum – less defined than in skeletal muscle
Myoendocrine
Cardiodilatins (CDD) or atrial natriuretic polypetides – vasodilation
Location of Smooth Muscle
Location – tubular organs
Form – Spindle shaped with centrally located nucleus
Similar cells-- myoepithelial cells, pericytes, myofibroblasts.
Light Microscopic Structure
Elongated
Nonstriated
Overlapping
Smooth Muscle Filaments
Thin Filaments – 30-80 Å, composed of actin
Dark Patches – cytoplasmic dense bodies
subplasmalemma dense plaques
Anchoring Filaments – 100 Å, desmin IF,
links together bodies and plaques
Thick Filaments – 120-160 Å, composed of myosin
Sliding Filament Mechanism – calmodulin dependent
Smooth Muscle contraction
1. SM contraction is the result of Actin-Myosin interactions that result in sliding filaments. However, the stimulus for SM contraction can be:

2. Neural, Stretch, Gap Junction electrical, or Humoral .

3. SM in different locations are stimulated by different mechanisms.
Smooth Muscle-like cells
Myoepithelial cells- have the properties of epithelial cells, but they are also contractile. Located in sweat, mammary and salivary glands

Myofibroblasts- have the properties of fibroblasts, but they are also contractile. Important in wound healing and scar contraction.
Myoepithelial cells
- have the properties of epithelial cells, but they are also contractile. Located in sweat, mammary and salivary glands

Myofibroblasts- have the properties of fibroblasts, but they are also contractile. Important in wound healing and scar contraction.
Myofibroblasts
- have the properties of fibroblasts, but they are also contractile. Important in wound healing and scar contraction.
Smooth Muscle and Disease
Asthma- SM in Bronchi contracts and limit aid flow.
Atherosclerosis- SM proliferation contribute to lumen narrowing

Myofibroblasts –Fibrotic diseases like cirrhosis or lung responses to smoking
Myoepithelial cells- Lactation failure due to lack of response to oxytocin.
Asthma
Asthma- SM in Bronchi contracts and limit aid flow.
Atherosclerosis- SM proliferation contribute to lumen narrowing
What are the general properties of the collagen gene family?
Most abundant connective tissue protein in vertebrates (ca. 25% by weight – mostly Type I)
Confers the property of tensile strength
Primary determinant of tissue form
Serves as a scaffold for cell attachment and mineralization
The collagens comprise a family of?
28 extracellular matrix proteins.
Each is a product of a unique genetic loci
Therefore, a substantial portion of the human genome is devoted to the collagen gene family
Common features of collagen.
1. All of them are made up of Oligomers of 3 polypeptide chains (α-chains)

2. They Contain regions (domains) of triple helix

3. 1º amino acid sequence of the triple helical domain is repeating 3 amino acids [gly-X-Y]n
X=proline usually, Y usually hydroxyproline

4. Unusual post-translational modifications of hydroxyproline and hydroxylysine (also present in elastin, acetylcholinesterase, osteocalcin (bone protein), and complement component C1q)
What are the Fibril forming collagens?
e.g. types I-III, V and XI
What are the Network forming collagens that form mesh-like arrays?
e.g. collagen types IV, VI, VII, VIII and X
What are the Fibril associated collagens with interrupted triple helices (FACIT)?
e.g. collagen types IX, XII, XIV and XVI (“chimeric” proteins)
What are the Transmembrane collagens that link cells with ECM?
collagen types XIII, XVII (aka BPAG2)
Type II collagen has how many alpha chains?
only 1. Hyaline cartilage – all 3 alpha come off the same chain
Type IV collagen
has 5 alpha chains and is the major collagen of most basement membranes
Type VII collagen
has 1 alpha chain and is the anchoring fibril of the basement membrane
Type XII collagen
has 1 alpha chain. Tendons, ligaments, muscle insertions in association with collagen type I. Non-collagenous domains contain fibronectin and von Willibrand clotting factor-like domains.
What is the structure of collagen?
Made up of 3 alpha chains – need glycine and every 1st position, which allows each alpha chain to form a tight right handed alpha helix. Helps with tensile strength.
Because glycine is at every 3rd position, the R groups are outward projecting, which protects the peptide bond.
The X in the triple helix type I collagen domain is usually?
proline
The Y in the triple helix type I collagen domain is usually?
Hydroxyproline
What do tight turns of the Type I collagen helix do?
force the functional groups of the amino acids in X and Y positions to radiate from the peptide bond back bone of the α chains, hence collagen is resistant to most proteases (steric hinderance).
That is the precursor to collagen
procollagen


The cell secretes propeptidases that cleaves off the propeptides of the procollagen and then they self assemble into a collagen fibrils.
Type I collagen C and N telopeptides
C- and N-telopeptides: Short sections at the carboxy and amino termini of the type I collagen protein that do not assemble into the collagen triple helix

During degradation of the type I collagen protein both C- and N-telopeptides are locally released. Levels can be measured in urine, serum or gingival crevicular fluid as a measure of collagen degradation
How is the collagen molecule stable?
1. Repeated glycines allow the formation of a tight α-helix and maximal intra chain hydrogen bonding

2. Proline, an imido acid, limits rotation around the peptide bond

3. Hydroxy-proline and lysine allow intrachain hydrogen bonding

4. Inter-molecule covalent bonds between derivatives of lysine and hydroxylysine formed extra-cellularly
Why do collagen fibrils have a banded appearance?
1/4 staggered arrays

The ¼ region fits a crysallite of hydroxyappitite just perfectly inside the space, giving significance in regards to mineralization
During Collagen biosynthesis, where do the post translational modifications take place?
Occurs in the cisternae of the ER or the leaflets of the golgi
Why do we need a procollagen form?
C and N propeptides prevent intracellular fibril formation
C propeptides, register the three α chains within the cisternae of the ER, allowing formation of?
the triple helix in a C→N direction
What are some post translational modifications that occur with collagen?
Disulfide bond formation
Hydroxylation of proline and lysine by prolylhydroxylase. Under-hydroxylation results in scurvy
Glycosylation of hydroxylysine
Extra-cellular processing of C and N propeptides (Ehlers-Danlos syndrome)
Covalent crosslinks between derivatives of lysine and hydroxylysine (Lathynism)
Prolyl hydroxylase in the cisternae of the ER requires ascorbate (vitamin C), Fe++, O2 and α-ketoglutarate for activity. The enzyme can only act on nascent procollagen α-chains prior to formation of the triple helix. Under hydroxylation of newly synthesized collagen results in?
scurvy
The inability to synthesize ascorbate (vitamin C) is due to a mutation in?
gulonolactone oxidase gene
Lysyl oxidase, which is involved in the biosynthesis of collagen is present in the cisternae of the ER and requires Cu++ as a cofactor. Failure to extract copper from the diet results in?
Menke’s disease.
Lysyl oxidase, which is involved in the biosynthesis of collagen, can be inhibited by β-amino proprionitrile (found in sweat peas) resulting in?
lathyrism.
Aldehyde derivatives of lysine are essential to the formation of?
extracelluar collagen cross links.
Procollagen α1(I) gene is divided into approximately 50 exons, most exons are 54 bp in length and the remaining are a multiples of?
9 bp
What are the cues for exon/intron genome organization?
1. FACIT collagen domains containing non-collagenous sequences have been “spliced in” from other genes such as fibronectin and von Willibrand’s clotting factor in the case of type XII collagen.

2. Chromosome location. The genes for α chains for heterotrimeric collagens (e.g. types I, V, IX) are always found on separate chromosomes.

3. Nucleic acid sequence. Each exon begins with the triplet codon for glycine and ends with the codon for a “Y” amino acid.
For collagen, the smallest number of [gly-X-Y]n that can form a thermodynamically stable triple helix at 37°C is?
54 bp long. (6 triplets X 3 nucleotides/codon X 3 codons = 54 nucleotides)
Most abundant ECM protein is?
collagen. Collagens impart tensile strength
Other desirable physical properties:
Tissue pliancy
Volume for tissue growth and cell migration
Linkage of ECM proteins into a unified matrix
Cell matrix interactions
Others
Guanidine is used in protein extraction and collagen extraction, what agent is guanidine?
a chaotropic agent. It induces chaos…
Non Collagen Protein: Elastin
1. Ultrastructure
Amorphous component
Microfibrillar component

2. Properties
Rubber-like, high molecular weight protein
Insoluble in most solvents

3. Composition
33% glycine, 10-13% proline or hydroxyproline, 40 hydrophobic amino acids (very few hydrophobic amino acids in collagen)
Contains desmosine, a post translational derivative of lysine
We Can isolate precursor of elastin by?
growing animals in a copper deficient diet and extract a 72 kDa protein without crosslinks (= tropoelastin)
More recently the elastin gene has been sequenced
What Two domains have been described?
Small domains with lysine-derived crosslinkages (desmosine)
Large domains enriched in hydrophobic amino acids
Why is elastin an elastic molecule?
With long sequences of hydrophobic amino acids and inside the body they are trying to make their surface area as close as possible. When pulling them outward and letting go they’ll recoil because of the aqueous environment. That is why it is an elastic molecule.

We also have a mean high blood pressure that is relatively high and this correlates with the number hydrophobic amino acids in elastin
Fibronectin
1. Abundant serum and ECM protein

2. Functions: decorates collagen, fibrin and other ECM molecules during development and wound healing

3. Facilitates cell migration both in wound healing and development (eg neural crest cell migration)
Cell migration is facilitated for cells that express cell adhesion molecules (eg some classes of integrins) that specifically bind the RGD sequence repeats of fibronectin.

RGD is comprised of what AA's?
arginine, glycine, aspartic acid
Collagen is turned over pretty quickly. Is the half-life of collagen the same in all tissues?
No

Skin: 50.4 days
Palate 21.4 days
***Periodontal Ligament 8.0 days***
Ginviva 11.4 days
Periodontal ligament is richly supplied with vasculature implying what?
a high metabolic rate
Fibrillar collagens are highly resistant to most?
proteases
Bacterial collagenases (e.g. P. gingivalis collagenase) cleave fibrillar collagens at each?
glycine residue (done to invade tissue)
During degradation, Vertebrate collagenases bind to the?
“hinge region” of type I collagen and make a single cut in each α-chain generating 2 fragments ¼ and ¾ the length of intact type I collagen (ends start to fray and denature become gelatin)

Other proteases then complete the degradation of the partially denatured collagen molecule
The hinge region is between what two AAs?
leucine and glycine
Periodontal ligament fibroblasts both synthesize and degrade?
collagen fibrils
Vertebrate collagenases are members of what family?
Matrix Metalloproteinase family.
More than 23 MMPs characterized in the human
Most share a homologous 27 kD active site (don’t need to know the size)
MMPs require what at their active site in order for proteolysis to occur?
Require divalent Zn++ as a co-factor at the active site for proteolysis to occur. ***NEED TO KNOW.

Also Require Zn++ and Ca++ for enzyme conformational stability
MMPs, Most are synthesized as proenzymes
and are Are inhibited by?
TIMPS - Tissue Inhibitors Metalloproteinases (in the ECM) or systemically by serum α2-macroglobin

Can be divided by substrate specificity into: collagenases, gelatinases, stromelysins and matrilysins
MMP 1 and 8
Collagenases

1 - CT
8 - Inflammatory cells
MMP 14
A membrane type MMP - PMN
Most MMPs are synthesized as a proenzyme, locally activated by proteinases such as?
plasmin or trypsin
local proteinases are activated by other proteinases, such as?
plasminogen activator, released in sites of inflammation and wound healing
MMP expression can be up-regulated by increased transcription, eg. pro-inflammatory cytokines (TNF-α, IL-1, IL-6) increase especially MMP-8
MMPs are inhibited locally by issue inhibitors of metaloproteinases (TIMPs 1-4) and systemically by α2-macroglobin
MMP expression can be up-regulated by?
increased transcription, eg. pro-inflammatory cytokines (TNF-α, IL-1, IL-6) increase especially MMP-8
MMPs are inhibited locally by?
issue inhibitors of metaloproteinases (TIMPs 1-4) and systemically by α2-macroglobin
MMP1 has a higher affinity for Zinc than?
MMP8. Tetracycline will bind to the active site of all MMPs, but at low dose can bind to MMP8 and inhibit it.

Periostat (doxacycline) is a low dose form of a derivative of tetracylcine – pill form
Thin actin and thick myosin filaments overlap w/ each other in a hexagonal array in cross section.
What is their ratio?
????
How is contractile force generated?
- myosin backbone swings its cross-bridge arms + globular heads out to bind to the thin actin filament - the arm bends, pulls actin towards center, Z-lines move closer, sarcomeres shorten, fiber shortens
this is happening in opposite directions on both ends of the myosin rod. grab, pull, release... grab again, pull, release... grab again... like climbing a rope remember that there are 6 actin thin filaments surrounding every 1 myosin thick filament
Where does the ATP energy come from to create contractile force?
- Myosin heads have a high affinity for ATP in the resting state - Myosin heads have ATP-ase enzymes in them
1. myosin head + ATP (resting state), what happens next?
2. myosin head + ATP grabs on to actin --> forms a cross-bridge of actin + myosin + ATP
3. ATP hydrolysis --> actin + myosin + ADP --> allows the myosin arm to bend and pull the actin filament
4. ADP released --> actin + myosin “Rigor Complex”
5. but ATP is also needed to break the cross-bridge b/t actin and myosin to relax the muscle
6. if no ATP is available --> actin-myosin cross-bridge is
locked --> rigor mortis
• Tropomyosin =
blocks the actin active site where myosin would bind for contraction.

Unblocked = contractile state
Blocked = relaxed state
Troponins. There are 3.
- M = binds to Tropomyosin
- I = intermediate linked
- C = binds Ca++
During contraction = Ca++ binds to?
Troponin --> Troponin conformation change --> Tropomyosin unblock --> cross-bridge formed
During relaxation = there is no Ca++ bound to?
Troponin --> Tropomyosin blocks actin’s binding site
What regulates Ca++ in the cytoplasm?
at rest [Ca++] = 10-7 M
activated [Ca++] = 10-5 M
equilibrium constant = 2 x 10-6 M meaning that 50% of Ca++ binded + 50% not binded
Contraction occurs during what Ca concentration?
- contract = if [Ca++] is higher than 2 x 10-6 M --> Ca++ will bind to Troponin-C --> Tropomyosin unblocked
Relaxation occurs during what Ca concentration?
- relaxed = if [Ca++] is lower than 2 x 10-6 M --> Ca++ will not bind to Troponin-C --> Tropomyosin blocks
no significant amt of Ca++ comes into the cytoplasm from the extracellular fluid b/c?
there are too many myofibrils blocking
Sarcoplasmic Reticulum
Sarcoplasmic Reticulum
- stores Ca++ (bound to Calciquestrin)
- has ligand-gated calcium channels that bind to Ca++ from T-tubule system for initiation of contraction
- Ca++/ATP-ase pump = cytoplasmic Ca++
T-tubule
T-tubule system
- different from the SR system
- continuous w/ the extracellular fluid
- weaves in bt myofibrils (straws)
- closely approaches the terminal cisternae of the SR, but never connects / make contact with it
What is the signal that triggers cytoplasmic Ca++ influx?
1. depolarization of the sarcolemma (there are very few voltage-gated Ca++ channels here)
2. action potential transfer to the T-tubule system
3. T-tubule membrane opens up its voltage-gated Ca++ channels
4. allows Ca++ into the narrow space b/t T-tubule and SR
5. Ca++ binds to receptors in the SR
6. Ca++ channels in the SR open and release Ca++ into cytoplasm --> 100x increase in [Ca++]
When Ca binds to Troponin C, what happens next?
7. Ca++ binds to Troponin-C due to equilibrium constant > 2x10-6 M

8. Tropomyosin moves out of the way to unblock the Actin binding site (relaxed state)

9. Myosin heads + ATP form a cross-bridge w/ Actin

10. ATP hydrolysis allows the Myosin arms to bend and pull the Actin filaments --> contraction

11. ATP is needed to break the Myosin-Actin cross-bridge --> relaxation

12. SR Ca++/ATP-pase pump removes Ca++ from the cytoplasm back into the SR --> relaxation
What is the Series Elastic Component of muscle for locomotion?
muscle --> tendon (the series elastic component) --> bone

1. Actin-Myosin System generates the tension force
2. Tendons demonstrate elastic elongation that is proportional to the force imposed on it by muscle

• As long as the Actin-Myosin System is producing tension, tendons will continue to stretch and exert progressively increasing force on the bone until it becomes enough to actually move the bone
TWITCH =
the actual tension force produced by a muscle in response to one action potential.

how much tension force is produced depends on how many Myosin-Actin cross-bridges are formed

reduced tension force results from either stretching or contracting too much... beyond maximum stretch yields no overlap of thick and thin filaments anymore beyond maximum contraction = thin actin filaments from opp. Z-Lines overlap

Because of this, thin filaments are no longer at an optimum distance from thick filaments for good overlap and formation of cross-bridge
TETANUS =
the constant full expression of tension produced by the Actin-Myosin System that is only possible when the rate of action potentials is above the “critical frequency”

Side comments...
- this is not associated to the disease Tetanus
- under normal conditions, your body cannot produce a tetanus state
- in both cases, rate of action potentials is below the critical frequency
- tetanus state is not physiologically possible & can only be artificially generated - producing a tetanus state is the only accurate way to measure muscle potential
Active Tension + Passive Tension =
total tension (tetanus)
Skeletal muscle cells
- large size = surface area... requires lots of SR for Ca++ storage in prep for contraction (Calciquestrin)
- contraction depends on SR Ca++ --> into cytoplasm to initiate pre-contractile events
(if the skeletal muscle depended on Ca++ coming from ECM (like in nerve transmission),
it would be impossible for the Ca++ to diffuse through all the stacked layers of muscle fibers)
Smooth Muscle cells
- small size = increased surface area... does not depend as much on SR for Ca++ storage (Calmodulin)

- contraction depends on ECM Ca++ --> into the cytoplasm to initiate pre-contractile events (like in nerve) • does not have a Parallel Elastic Component that skeletal muscle has
Cardiac Muscle Cells
- intermediate in size, surface area, complexity, uses both sarcolemma + SR for cytoplasmic Ca++ influx

- muscle fibers are connected to e/o by Gap Jxns (connexons) --> forms a network / “Syncitium”

- action potentials involve a large amount of voltage-gated Ca++ ion channels voltage-gated Na+ ion channels open = influx of Na+ --> fast depolarization
voltage-gated Ca++ ion channels open = influx of Ca++
Blood
Properties:
bright to dark red color
viscous
slightly alkaline (pH = 7.4)
Accounts for approx. 7% of the total body weight
Average total blood volume = 5 L / 5.5 kg
70 ml per Kg body weight
Clot =
Formed elements + serum
serum
plasma without the clotting factors
Name some anti-coagulants
heparin, EDTA, citrate
Blood in a centrifuge will have what three layers?
Bottom – red cells
Upper – plasma
Middle – buffy coat: white cells and platelets
Plasma
Fluid component of blood comprising about 55% of total blood volume
Pale yellow aqueous (90%) solution of
proteins (7-8%)
electrolytes and ions (0.9-1%)
organic molecules (1-2%)
Plasma Proteins
1. Albumin - most abundant and maintains colloid pressure.
2. Globulins. Gama = antibodies secreted by plasma cells. Alpha and Beta are non-imune globulins
3. Fibrinogen = blood clot
Other plasma components
Electrolytes: Na+, Ca2+, K+, Mg2+, Fe2+, Cl-, HCO3-, PO43-, SO42-
Regulate osmotic balance and maintain pH
Imbalance of electrolytes – serious medical condition
Nutrients: amino acids, glucose, lipids
Building blocks of macromolecules and production of energy
Enzymes: alkaline phosphatase (ALP), creatinine kinase (CK), aspartate transaminase (AST), lactate dehydrogenase (LDH)
Hormones
Blood gases: O2, CO2, N2
Waste products: urea, uric acid, creatinine, ammonium salts
transported to kidneys for removal from the body
Erythrocytes
Most numerous (5 x 1012 / L of blood)
Red color because of hemoglobin
Survival in circulation: 120 days
Transport O2 from lungs to tissues
and CO2 away from tissues
Platelets or Thrombocytes
Second most numerous cell type in blood (250 x 109 / L of blood)
Produced by fragmentation of cytoplasm of megakaryocytes (large multinucleate cells) in bone marrow
Survival in circulation: 10-12 days
Role in hemostasis
Leukocytes (5 x 10^9 /L of blood)
1. Neutrophils - Bacteria
2. Eosinophils - Parasites
3. Basophils - Anaphylactic and inflammatory
Monocytes
mature into macrophages and produce cytokines for inflammatory response.
Lymphocytes 33% of WBCs
T-Lymphocytes - cell mediated
B-Lymphocytes - humoral immunity
Order of most to least WBCs
Neutrophils, Lymphocytes, Eosinophils, and Basophils
CBC
Complete Blood Count
Hematocrit
packed cell volume (PCV) - %
amount of space (volume) RBCs take up in blood
mean corpuscular (cell) volume (MCV) – fL (10-15)
average volume of RBCs
measure of RBC size
(Hct / RBC #) x 10
mean corpuscular (cell) hemoglobin (MCH) – pg
average weight of Hb in an RBC
measure of RBC weight
(Hb / RBC #) x 10
mean corpuscular (cell) hemoglobin concentration (MCHC) – g / dL
average concentration (relative to cell size) of Hb in an RBC
measure of how many RBCs in a certain amount of fluid
(Hb / Hct) x 10
red cell distribution width (RDW) - %
measure of size variation of RBCs
Mean Platelet Volume (MPV) – fL
measure of the average amount (volume) of platelets
What are the 3 Romanowsky's Blood Stains?
Wright’s stain – simpler method
Giemsa stain – more complex stain; for delicate staining characteristics and detection of blood parasites
May-Grünwald stain – good for routine work
Wright’s stain
– simpler method
Giemsa stain
– more complex stain; for delicate staining characteristics and detection of blood parasites
May-Grünwald stain
– good for routine work
RBC
no DNA/nucleus 120 days in the circulation
1 circ cycle 20 sec.
Reticulcytes
immature RBCs and still has rRNA - no nucleus
high reticulocyte is found in?
Anemia


*bleeding and premature destruction of RBCs
Low reticulocyte count causes
vitamin / mineral deficiency
BM malignancy
radiation (chemotherapy)
chronic infection
drugs
Hb
RBCs packed with hemoglobin = O2-carrying protein
hemoglobin = large protein composed of 4 amino acid chains each bound to an iron-containing heme group
the heme group consists of an iron (Fe) ion, which is the site of O2 binding
Hb also carries CO2 that binds to N-terminal groups of the globin chains

8 globin genes
Where is heme synthesized?
in mitochondria and cytoplasm of immature RBCs
Normal Hb: globin chains a, b, g, and d (z and e 3 -10 wks gestation)
Adult : Hb A - HbA1: a2b2 (97%); HbA2: a2d2 (2-3%); HbF (1%)
Fetus: Hb F - a2g2
oxyhemoglobin: Hb carrying O2
carbaminohemoglobin: Hb carrying CO2
Abnormal Hb:
carboxyhemoglobin:
methemoglobin:
Abnormal Hb:
carboxyhemoglobin: binds to CO instead of O2
methemoglobin: Fe2+ to Fe3+; cannot bind O2
Increased RBC # is called?
erythrocytosis or polycythemia
A decreased RBC # is called?
decreased number: anemia, erythropenia (erythrocytopenia)
anisocytosis
same or variable RBCs
RBC sizes and colors
normal – normocytic
small – microcytic
large: macrocytic/megalocytic

color – blood smear; [Hb]
normal – normochromic;
pale – hypochromic
darker: hyperchromic
poikilocytosis
shape; normal biconcave vs. different and oddly shaped cells
Blood smear distribution:
rouleaux
Red cell agglutination
distribution in the blood smear

rouleaux – like stack of coins; due to increase in high MW plasma proteins

red cell agglutination – due to antibody on surface of RBCs
Red cell inclusions
Pappenheimer bodies – small basophilic inclusions in cell periphery

basophilic stippling - small basophilic inclusions throughout cell

Howell-Jolly bodies – large round densely stained inclusions on edge of cell; nuclear remnant
Polycythemia
Characterized by increased Hct
Polycythemia Rubra Vera PV
chronic myeloproliferative disease
due to increased red cell proliferation in bone marrow (BM)
rare; most prevalent in Ashkenazi Jews
Acute blood loss in Anemia is caused by?
trauma
surgery
symptoms and signs related to hypovolemia (decreased blood volume); hypotension, palpitations, tachycardia, fainting, shock
Chronic Blood loss in Anemia
gastrointestinal bleeding due to
ulcers
gastritis
cancer
hemorrhoids
non-steroidal anti-inflammatory drugs (e.g. aspirin, ibuprofen)
heavy menstrual bleeding
childbirth
symptoms: fatigue, pallor, dyspnea, (shortness of breath), dizziness
Iron Deficiency anemia
Insufficient iron stores due to
poor diet
poor iron absorption / utilization
body iron stores are depleted by prolonged bleeding
Common in menstruating and pregnant women
CBC: low reticulocytes, MCV, MCH, ferritin (iron-storing protein
Sideroblastic Anemia
abnormal incorporation of iron into the heme group of Hb
toxic accumulation of iron in mitochondria = ringed sideroblasts
classified as:
hereditary – enzyme deficiency (genetic); deficiency of enzyme involved in heme synthesis
secondary - drug-induced or alcohol-induced, lead poisoning; inhibition of enzymes involved in heme synthesis
idiopathic – cause not known; primarily in elderly
Anemia of Chronic Disease
CBC:
low or normal MCV
normal to decreased serum iron levels
normal to increased serum ferritin
blood smear: marked rouleaux formation due to increased plasma protein concentration
Alpha Thalassemia is caused by what deletion?
1, 2, 3, or 4 a globin genes
Beta Thalassemia is caused by?
caused by mutations in the b globin gene cluster
B-Thalassemia is also known as?
Cooley's Anemia

most severe form; both b-globin genes are defective
reduced or absent Hb A1; increased Hb A2 and Hb F (up to 98%)
Lab findings:
CBC: low Hb, Hct, MCV
Beta Thalassemia minor
mildest form; one b-globin genes is defective
moderately reduced Hb A1; increased Hb A2 (4-8%) and Hb F (2-5%)
Lab findings:
CBC: normal Hb, low MCV, high RBCs (distinct from iron deficiency)
alpha Thalassemia
deficient or no synthesis of a-globin chains
severity of disease depends on number of a-globin chains deleted
If 1 alpha-globin chain is deleted what A-Thalassemia do you have?
"silent" carrier

asymptomatic; only identified by pedigree and DNA analyses
If 2 alpha-globin chain is deleted what A-Thalassemia do you have?
A-thalassemia trait

a-thalassemia trait: mild hypochromic microcytosis
If 3 alpha-globin chain is deleted what A-Thalassemia do you have?
hemoglobin H disease


Hemoglobin H disease: production of Hb H (b4)
moderately severe microcytic anemia (due to reduced Hb synthesis and hemolysis); hepatosplenomegaly
If 4 alpha-globin chain is deleted what A-Thalassemia do you have?
hemoglobin Barts hydrops fetalis


most severe form of a-thalassemia;
complete absence of a-globin chains; hemoglobin Barts (g4)
incompatible with life - stillbirth by 3rd trimester or shortly after delivery
Hydrops fetalis: fluid buildup in multiple organs, cardiac failure, massive hepatosplenomegaly
Toxemia in mother carrying fetus with Barts hydrops fetalis
Hydrops fetalis
fluid buildup in multiple organs, cardiac failure, massive hepatosplenomegaly
Toxemia in mother carrying fetus with Barts hydrops fetalis
Macrocytic Anemias
elevated MCV
impaired DNA synthesis but normal RNA synthesis
usually due to B12 and folic acid deficiency
other causes
gastrointestinal disorder or surgery
chemotherapy
chronic alcoholism
hypothyroidism
liver disease (hepatitis)
splenectomy
common; commonest in elderly
Macrocytic anemias – B12 deficiency
Causes:
inadequate dietary intake (in vegans)
intestinal malabsorption due to lack of intrinsic factor (gastrectomy, pernicious anemia)
increased requirements during pregnancy
Years to develop deficiency because of sufficient body stores
Macrocytic Anemias - Folic Acid deficiency
Causes:
Poor diet (no fresh fruits/vegetables) or overcooked food
intestinal malabsorption due to alcoholism, gastrectomy, Crohn’s disease
increased requirements during pregnancy
certain drugs (anticonvulsive) impair intestinal folic acid absorption
deficiency develops fast; body stores only for 3 months
Lab findings, Signs & Symptoms: same as in B12 deficiency
Treatment: Folic acid supplement
Hemolytic disorders
reduction in RBC lifespan and compensatory increase in rate of erythropoiesis
Causes: incompatible blood transfusion, cancer, drugs, etc.
intravascular or extravascular hemolysis:
extravascular: most common form of hemolytic anemia
premature destruction of RBCs by macrophages in spleen, liver, BM
intravascular: abnormal breakdown of RBCs within blood vessels
Hemolytic Disorder Pathophysiology

Anemia – frequently absent because of increased erythropoiesis
jaundice – increased RBC breakdown products; increased bilirubin
cholelithiasis – gall stones; due to increased bilirubin
splenomegaly
hemoglobinuria – passing of Hb in urine
hemoglobinemia – too much Hb exceeds binding capacity of haptoglobin and cannot get cleared from body
growth retardation and delayed puberty
hypertrophic skeletal changes – due to BM expansion
G6PD deficiency
Glucose-6-phosphate dehydrogenase - red cell enzyme
Role: protects red cell proteins from endogenous or exogenous oxidant stress
Mechanism: converts glucose to ribose-5-phosphate - NADPH production
NADPH prevents building up of free radicals within cells
G6PD deficiency – hereditary enzyme deficiency; malaria immunity
increased susceptibility of RBCs to oxidant stress - hemolysis
most common enzyme deficiency
X-linked recessive disorder affecting more males than females

CBC: low RBC count and Hb, high reticulocyte count
In G6PD what are the triggers of hemolysis?
ingestion of drugs (antimalarial, aspirin) - induce oxidant stress on RBCs
ingestion of fava beans – divicine
viral infections: lung infection, hepatitis - activated neutrophils
Reason: production and release of free radicals
Sickle Cell Disease
Genetic disorder - change in hemoglobin structure due to mutation in the Hb gene
Normal Hb: Hb A; substitution of glutamic acid by valine – Hb S

Result: distortion of RBC shape from biconcave to half-moon (sickle) shape - Sickle cell disease

Carriers are resistant to malaria

low MCV, Hb; high WBC, neutrophils, lymphocytes, platelets, reticulocytes; Hb S
Diapedesis:
migration of WBCs through vessel walls to tissues
What are the granulocyte WBCs?
Neutrophils – normal value : 2000-7500/ml; 60-70% WBCs
Eosinophils - normal value : < 450 / ml; <4% of WBCs
Basophils - normal value : < 200 / ml; <1% of WBCs
What are the Agranulocyte WBCs?
Lymphocytes – normal value : 1000-4000/ml; 20-35% WBCs
Monocytes – normal value : < 900 / ml; 5% of WBCs
Both granulocytes and agranulocytes possess lysosomes
Neutrophils
Function: defense against acute bacterial and fungal infection
Move to site of infection by chemotaxis
Engulf invading microbes – phagocytosis
Kill by releasing toxic substances stored in their granules
Neutrophils have what 3 types of granules?
Azurophilic (primary) – lysosomes; largest and fewest; bactericidal
Specific (secondary); smallest and most numerous; bactericidal
Tertiary – facilitate diapedesis
Hypersegmented neutrophils:
6 or more lobes; Vitamin B12 or folic acid deficiency; interferes with DNA synthesis
Hyposegmented neutrophils:
defect in chromatin synthesis e.g. Pelger-Huet anomaly; myelodysplasia
Eosinophils
produced in BM; 4-5 hours in bloodstream
bilobed nucleus; pale blue cytoplasm with large orange-red granules
Specific granules – contain proteins with cytotoxic effect, neutralization of histamine
Azurophilic granules – lysosomes: enzymes destroy parasites


respond to chemotactic stimuli, phagosytose and kill
Function: defense against parasitic infestation, dampen allergic reaction
Basophils
lobed S-shape nucleus with large purple-staining cytoplasmic granules
Specific granules – histamine, slow-reacting substance-A (SRS-A); vasodilation
Azurophilic granules – lysosomes
Basophil function
Function: initiators of inflammatory processes, involved in anaphylactic, hypersensitivity, and inflammatory reactions
antigen triggers release of IgE
IgE binds to basophil surface receptors
cell degranulates and releases inflammatory mediators (histamine) to surrounding tissues
There are 3 types of lymphocytes based on what?
3 types depending on
size
amount of cytoplasm
presence or absence of cytoplasmic granules
Small - most numerous, dense homogeneous nucleus; N/C >90%
Medium - less dense, somewhat heterogeneous nucleus; N/C 80%
Large - (granular or agranular) lymphocytes; N/C 40-60%
What are the 3 types of Lymphocytes?
T cells – differentiate in thymus; (75-80%); cell-mediated immunity
B cells - differentiate in BM; (15%); plasma cells; humoral immunity
Natural killer cells (NK) - programmed to kill foreign, virally altered cells, some types of tumor cells; (5-10%); cell-mediated immunity
larger than B and T cells
contain large cytoplasmic granules – large granular leukocytes
Monocytes
in blood stream for a short time, then migrate to tissues where they mature into actively phagocytic macrophages
Function: body’s defense against bacterial and fungal infections; also ingest and break down dead and dying body cells
during inflammation
monocyte leaves blood vessels at site of inflammation,
transforms into a tissue macrophage, and
phagocytoses bacteria, other cells and tissue debris
What is the most common type of Leukopenia?
Neutropenia -Susceptibility to bacterial & fungal infections – reduced phagocytosis
Neutrophila
Causes:
Acute infections (bacteria, viruses, parasites, fungi)
noninfectious: inflammatory conditions (rheumatic fever, gout)
metabolic conditions (ketoacidosis, uremia)
cancer, MI, burns, drugs (steroids), stress / strenuous exercise
Leukocytosis - Monocytosis
Causes:
inflammatory disorders (granulomatous disease)
infections (TB, syphilis)
autoimmune disorders (lupus, rheumatoid arthritis)
connective tissue disorders (ulcerative colitis, collagen vascular disease)
cancer (leukemia, Hodgkin’s Disease)
Infectious Mononucleosis
cause of lymphocytosis
also known as “mono”, “glandular fever” or “the kissing disease”
spread primarily by saliva; sneezing and coughing
virus-induced – Epstein-Barr virus (EBV); type of herpes virus
EBV infects epithelial cells and B cells
95% of population exposed to EBV by 40 – most no symptoms
virus lingers in inactive form in WBCs until gets reactivated - IM

**Atypical Lymphocytes
Leukemias
malignant disorders of blood characterized by uncontrolled proliferation of hematopoietic cells
Acute Leukemia
Acute: uncontrolled proliferation of immature blood cells – blasts
rapid, usually fatal, survival less than 6 months
impaired BM function
Chronic Leukemia
Chronic: uncontrolled proliferation of well-differentiated mature blood cells
long-term disease, often long survival
incidental findings during routine exam
Acute Lymphocytic/Lymphoblastic Leukemia (ALL)
Proliferating cell – primitive lymphoid cell
Most common leukemia in children <15 years (60-70%)
Principal cause of cancer deaths in children; peak incidence age 4
Treatable and potentially curable – half the children (age 2-10) are cured
Poor prognosis for adults
Classified according to lymphocytes and state of maturation
Early B cell
Pre-B cell
Mature B cell
Early T cell
Mature T cell
Acute Myeloid / Myelogenous Leukemia (AML)
Proliferating cell – primitive myeloid cell
Occurs at all ages, from neonatal to adult
incidence increases with age; most common acute leukemia in adults
20% of acute leukemia in children
In blood smear cytoplasm contains inclusions, Auer rods, diagnostic

****Auer Rods****
Chronic Lymphocytic Leukemia (CLL)
proliferating cells – mature but incompetent lymphocytes, don’t make antibodies in response to antigens; 95% B lymphocytes
accounts for 2/3 of chronic leukemias ; most common over 60 yrs
male : female ratio 2:1
early stages asymptomatic; later s fatigue, weight loss, lymphadenopathy, hepatosplenomegaly
Lab findings:
CBC: leukocytosis 5-10x higher WBC count, low Hb and platelets
Chronic myeloid / myelogenous Leukemia (CML)
results in myeloid marrow hyperplasia
accounts for 1/3 of chronic leukemias ; young adults (10-20 yrs) and middle age (50-60 yrs)
due to Philadelphia chromosome mutation; translocation between chromosome 9 and 22
Chronic myeloid / myelogenous Leukemia (CML)
chronic: 3-5 yrs; asymptomatic
acute: 2-4 months; increased blasts (>30%) in blood and BM
No response to treatment – blast crisis
fever, weakness, fatigue, anorexia, weight loss, splenomegaly, anemia, infection
Lymphomas
Solid tumors that arise in lymphoid tissue but then spread to other solid tissues, blood and BM
3rd most common malignancy in children
2 types:
Hodgkin’s Lymphoma / Disease:
Non-Hodgkin’s Lymphoma:
Hodgkin's Lymphoma
2 types: classic and nodular
95% have classic form
presence of rare malignant Hodgkin’s Reed-Sternberg cells
Unknown cause; genetic and environmental e.g. EBV
Non Hodgkin's Lymphoma
associated with chronic inflammatory diseases e.g. rheumatoid arthritis
often develops after organ transplantation in immunosuppressed patients
Causes: unknown
Genetic – chromosomal translocations
Viral - HIV, EBV
90% of NHL B-cell lymphomas; 10% T-cell lymphomas
Bence-Jones Proteins that are excreted in urine are diagnostic for?
Multiple Myeloma

plasma cells produce abnormal proteins (e.g. Bence-Jones). plasma cells accumulate in bone marrow – pressure on walls – bone pain, fractures
Platelets
Also known as thrombocytes – thrombos (Greek) = clot
Smallest blood cell (2-4mm diameter) - 200,000 – 400,000/mL
life span: 10-12 days
derived from cytoplasm fragmentation of megakaryocytes (giant multinucleated cells) residing in BM
promote blood clotting and help repair gaps in the walls of blood vessels, preventing blood loss
What has the following characteristics:
small, disk-shaped, non-nucleated cell fragment
peripheral light blue-stained transparent zone, the hyalomere, and central darker zone with purple granules, the granulomere
Platelets
Hyalomere
Microtubules – allow platelets to maintain their discoid shape
Actin / myosin – contraction – platelet movement & aggregation
Surface-opening system – take up and release molecules
Dense-tubular system – calcium storage, prevent platelet stickiness
Granulomere
Mitochondria
Glycogen deposits
Enzymes – catabolize glycogen, consume O2, and generate ATP
Granules – 3 types: a, d, l
This granule deals with vessel repair, blood coagulation, and platelet aggregation.
A-granules
This granule contains Ca, ADP, ATP, histamine, serotonin
and facilitates platelet adhesion and vasoconstriction.
Delta granules
This granule contains lysosomal enzymes
and clot resorption
gamma granules (lysosomes)
Heomstasis
clotting mechanism of blood; Sequence of events leading to cessation of bleeding by formation of a stable plug (clot)
What are the stages of hemostasis?
Vascular wall injury and exposure of sub-endothelial collagen
Vasoconstriction
Formation of platelet plug
Coagulation cascade
Formation of fibrin clot (plug)
Healing and fibrinolysis
What are the platelet functions
Adhesion to the injured surface
Shape change
Release of granule contents
Aggregation
What is the mechanism of Hemostasis? Part I
1. blood vessel wall injured, collagen fibers exposed at damaged site
2. release of endothelin (vasoconstrictor) and von Willebrand factor (vWF) which binds exposed collagen fibers
3. Platelets adhere to exposed collagen by binding to vWF and they get activated (platelet activation)
Platelet shape transformation
Platelet degranulation
Platelet aggregation
- Primary hemostatic plug (loose)
Mechanism of Hemostasis Part II
4. conversion of fibrinogen to fibrin
Platelet cross-linking by fibrin
Secondary hemostatic plug (solid)
Blood coagulation

5. Clot shrinks
Clot Retraction

6. Clot dissolves after fibrin degradation by proteolytic enzyme plasmin
Clot Removal - Fibrinolysis
Vessel wall repaired
Intrinsic Clotting Pathway
1. Begins with activation of F XII by
surface contact
2. F XII interacts with F XI
3. F XI activates F IX → (F IXa)
- calcium required
4. F IXa forms complex with F VIII
5. F VIII converts F X → F Xa
Calcium and phospholipid required
Extrinsic Clotting Pathway
Predominant pathway
1. Depends on release of tissue factor (TF) - thromboplastin (F III)
2. TF released from endothelial cells at damaged site
3. TF binds F VII
- calcium required
4. F VIII converts F X → F Xa
What is the Common Clotting Pathway?
1. Activation of F X → F Xa
2. F Xa in presence of calcium, phospholipid and F V converts
prothrombin (F II) → thrombin (F IIa)
3. Thrombin converts
fibrinogen (soluble) → fibrin (insoluble)
(Fibrinogen – F I)
4. Fibrin polymerizes to a gel
5. Thrombin converts F XIII → F XIIIa
6. F XIIIa cross-links fibrin
- solid clot
Main Steps to Hemostasis**** KNOW THESE
1. Activation of F X → F Xa
2. F Xa in presence of calcium, phospholipid and F V converts
prothrombin → thrombin
3. Thrombin converts
fibrinogen (soluble) → fibrin (insoluble)
4. Fibrin polymerizes to a gel
5. Thrombin converts F XIII → F XIIIa
6. F XIIIa cross-links fibrin
- solid clot
Prothrombin time and Thrombin time
Prothrombin time (PT) – time sample takes to clot; normal 10-14 sec


thrombin time (TT) - normal 9-13 sec
ability to form clot from fibrinogen in presence of thrombin
Thrombocytosis (reactive)
Increased platelet count - >500,000 / mL
Causes:
genetic (JAK2 mutation)
infection / inflammation
iron deficiency
malignancy / chemotherapy reaction
splenectomy
Essential Thrombocythemia (primary)
Malignant disorder – increased platelet count >1,000,000 / mL
Rare in children and young adults; diagnosis at 60 yrs
Causes: as in thrombocytosis
Lab findings:
Blood smear: platelet clumps, variation in size, shape, granulation
Thrombus
Thrombus: clot that develops and persists in an unbroken blood vessel
May block circulation, leading to tissue death
Blockage of coronary artery – Myocardial Infarction
Embolus:
a thrombus that becomes dislodged or fragmented freely circulating in the blood stream; thromboembolism
Disseminated Intravascular Coagulation (DIC)
triggered by potent stimuli that activate both F XII and tissue factor
form ation of microthrombi and emboli throughout the microvasculature
Thrombosis and hemorrhage
consumption of coagulation factors and platelets, generation of FDPs that have antihemostatic effects
Hemophilia
sex-linked recessive inherited genetic disorder
caused by deficiency or defect in a clotting factor
severe bleeding even after mild injuries e.g. skin cut, and may bleed to death after more severe injuries
spontaneous hemorrhages in body cavities, e.g. major joints, intracranial, urinary tract
Hemophilia A defect?
defect in factor VIII

only males are affected by hemophilia A (transmitted from their mothers)
females have one defective X chromosome
females develop hemophilia only when they have the abnormal gene in both X chromosomes - rare
Hemophilia B defect?
Hemophilia B - defect in factor IX (Christmas Factor)
von Willebrand’s disease (vWD)
defects in concentration, structure, or function of vWF
defective platelet plug formation
autosomal dominant trait with varying penetrance
most common blood clotting disorder – 1:8,000-10,000 persons
females and males are equally affected in a heterozygous state
Signs & Symptoms:
easy bruising
prolonged bleeding after surgery or dental procedure
epistaxis (nose bleed)
mucosal bleeding, soft tissue hemorrhage
Thrombocytopenia
Platelet count < 100,000 / ml; very serious when < 50,000 / ml
Bleeding from small vessels, skin, GI tract
Purpura and petechiae – purple spots and patches on skin
autoimmune disease: antibodies are formed to one's own platelets and destroy the platelets
Caused by
Decreased platelet production
Increased platelet destruction
Idiopathic Thrombocytopenic Purpura (ITP)
caused by accelerated antibody-mediated platelet consumption
platelets are destroyed in spleen by macrophages
acute and self-limiting (2 to 6 weeks) in children
cause of death: Intracerebral hemorrhage
secondary to another disorder e.g. SLE, HIV
Ontogeny of Hematopoiesis
2 – 10 weeks gestation: yolk sac
2 weeks post-conception: “blood islands”; erythroid precursors
6 weeks gestation: granulocytes and megakaryocytes
7+ weeks gestation: lymphocytes in lymph sacs
CFU-GEM give rise to?
myeloid cells (granulocytes, erythrocytes, monocytes, megakaryocytes (platelets)) - myelopoiesis
CFU-L give rise?
to lymphoid cells (B, T, NK cells) – lymphopoiesis
CSFs
Growth factors or colony-stimulating factors (CSFs) affect blood cell proliferation and differentiation
stimulate proliferation of immature (progenitor and precursor) cells
support the differentiation of maturing cells
enhance the functions of mature cells
CSFs also stimulate cell division and differentiation of progenitor cells of the granulocytic and monocytic series
Growth Factors: Proliferation of stem cells
Stem Cell Factor (SCF)
Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF)
IL-3 and IL-7
Epo:
kidney-secreted hormone produced in response to hypoxia
stimulates production of globin, the protein component of Hb
What are the stages in Red Blood Cell formation?
1. Proerythroblast
2. Basophillic erythroblast
3. Polychromatic erythroblast
4. Orthochromatic erythroblast (not capable of Mitosis)
5. Reticulocytes (not capable of Mitosis)
Granulopoiesis
CFU-GM →neutrophils
CFU-Eo →eosinophils
CFU-Ba → basophils
Neutrophilic Metamyelocyte and Stab/Band Cell
have no cell division
Megakaryoblast → Megakaryocyte under hormonal stimulation by?
thrombopoietin