• 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

1/291

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;

291 Cards in this Set

  • Front
  • Back
Define Pathology
Study of disease

Functional, structural, biochemical alterations in the body produce disease
Define Epidemiology
WHO tends to be affected by a disease process
Define Homeostasis
Ability of body to maintain a condition
Define Allostasis
"Other", Differing, Varrying = maintain some degree of stability resulting from a change
Define Allostatic Overload
When continued stress is over-whelming and allostasis can't be upheld
T/F: Pathological specimens can tell us information about a period of time within an organism
FALSE. Pathological specimens represent a "snap shot" in time of body structure
What are the two main divisions of pathology?
Anatomic and Clinical
List some examples of Anatomic Pathology
Surgical Pathology
Cytology
Autopsy/Forensic Pathology
List some examples of Clinical Pathology (Laboratory Medicine)
Microbiology/Virology
Clinical Chemistry
Blood Bank/Transfusion Medicine/Hematology
HLA
What is the central dogma of molecular biology?
DNA --> RNA --> Protein
Define what is meant by risks in the central dogma of health and disease
Risks = Stressors that challenge Homeostasis (for ex. when chemicals in smoke cause cancer, they become etiological)
When do we want to intervene with patients?
Before risks cause disease, because after that time we have to TREAT disease = we want to stay away from having to treat
What are the two big causes of disease and Differential Diagnoses?
Nature (Genetics) and Nurture (Environment)
What are the categories of disease?
VINDICATE

Vascular
Infectious/Inflammatory
Neoplastic (primary/secondary)
Drugs
Idiopathic/Latrogenic (=we don't know)
Congenital/Developmental/ Inherited
Autoimmune/Allergic/ Anatomic
Trauma
Endocrine/Metabolic/Environmental/ Occupational
What types of cellular responses to stimuli are Pathologic? What types of cellular responses are physiologic?
Pathologic:
Injury
Death (Apoptosis or Necrosis)
Accumulations

Physiologic:
Adaptations
Detail the events if a cell is exposed to a stressor for a prolonged period of time
Cell exposed to stressor

Cell Adapts to respond to stressor

Cell structure and function are altered

Cell Injury and ultimately cell death may occur
What are the 4 major types of Adaptations cells undergo?
"HHAM"

Hyperplasia (Inc. Cell Number)
Hypertrophy (Inc. Size of Cell...think of a "big" Trophy)
Atrophy (Dec. Size of cell)
Metaplasia (change from one mature cell type to another...think "meta"morphosis)
What is the goal of cellular adaptation?
to adjust to new conditions/demands

Reflects dynamic ability of cells to alter their cell cycle activity
Define Hyperplasia
Inc. Number of Cells

Requires DNA synthesis and cell division
Why might Hyperplasia occur Physiologically (i.e. normally)?
Hormonal = If we need more hormones

Compensatory = we need to regain some functional capacity after losing some cells
Why might Hyperplasia occur Pathologically?
Cells might proliferate for protection...

For Ex., Epithelial Hyperplasia might occur to thicken epidermis to protect us
Give some examples of Physiological Hyperplasia and Pathological Hyperplasia
Physiological = partial liver removal = remaining liver undergoes hyperplasia growth
Endometrial thickness bc of estrogenic stimulation (proliferative phase)

Pathologic = Endometrial Hyperplasia (i.e. Endometriosis!)
Psoriasis or Lichen Simplex = epidermal hyperplasia
Define Hypertrophy
Increase in the size of an organ because the cells increase in size (bc of increased protein synthesis)
T/F: Hypertrophy generally occurs in cells that would not typically divide
TRUE (muscle cells, cardiac cells)
What happens when the heart undergoes cardiac hypertrophy?
Cells in heart inc. in size = Left Ventricle Thickens = it can't hold as much blood = Decreased volume capacity of Left Ventricle = we'll need to inc. Heart Rate to compensate
Define Atrophy
A decrease in cell size because of a loss of cellular substance

Occurs because the cellular environment can't support the cell's current size, so it shrinks (This is what occurred in the movie "Honey, I Shrunk the Kids")

Decreased cell size = decreased function
What are the causes of atrophy?
"DDRIP"

Decreased workload (disuse)
Denervation (loss of nerve supply)
Reduced endocrine stimulation
Ischemia
Poor Nutrition
Define Metaplasia
Change of cell from one mature type to another

For example: squamous epi --> Columnar epi
What is thought to cause Metaplasia?
Reprogramming of gene expression of stem cells in tissue

Generally is protective to noxious stimuli (change from susceptible cell type to more resistant/protective cell type)
T/F: Typically, Metaplasia is Irreversible
FALSE.

Metaplasia is typically REVERSIBLE
Define Dysplasia
If injurious stimulus continues to affect cell, progressive changes may result in dysplasia = one step before cell becomes neoplastic and pre-cancerous
List the 7 causes of cell Injury
"PIINCH - G"

Physical Agents (mechanical agents like temp. change)
Infectious Agents
Immunologic Reactions
Nutritional Imbalance
Chemicals/Drugs
Hypoxia (= diminished O2 delivery vs. Ischemia = Impaired blood flow)

Genetic Mutations
What are the underlying mechanisms of cell injury?
Ox. Phos. (dec. ATP)
Ca2+ Homeostasis
ROS (Reactive Oxygen Species)
Cell Membranes (inability to maintain membrane integrity)
What are the consequences of Ox. Phos. Disruption?
For ex. Ox. Phos. disruption may occur because of ischemia.

dec. Ox. Phos. = Mito less effective = Dec. ATP = many, many cellular processes affected
What are some of the pathological effects of ROS build up?
ROS react with:
1. FAs --> disruption of plasma membrane
2. Proteins --> Loss of activity, abnormal folding
3. DNA --> mutations, breaks
List some of the Antioxidant mechanisms that contribute to removal of free radicals
1. SOD (O2--> H2O2)
2. Glutathione (OH--> H2O and O2)
3. Catalase (H2O2 --> H2O and O2)
T/F: The severity and duration of causes of cell injury determine reversibility of cell damage
TRUE
T/F: most cells are already dead by the time we see the effects of the cell death
TRUE
T/F: Cell damage is reversible along a period of time as stressor continues
FALSE. Cell damage is reversible up to a specific point as stressor continues, but beyond that point cell damage is irreversible
What are some examples of reversible cell injury?
Cell swelling
Fatty change (accumulation of lipid vacuoles)
Irreversible cell injury leads to what?
Cell Death
Whats the difference between necrosis and apoptosis?
Necrosis = ONLY PATHOLOGIC. membrane becomes leaky = leakage of cellular proteins and constituents = can further damage neighboring cells.

Apoptosis = CAN BE PHYSIOLOGIC = NORMAL. programmed cell death. Cellular fragmentation occurs
How can Reperfusion injury cause cell death?
Generating ROS
Inflammatory Mechanisms
How will DNA damage between apoptosis and necrosis look different on a DNA gel?
Apoptosis = cellular fragmentation = DNA in-tact, just in different size = gel will look like a ladder

Necrosis = DNA gel will show a continuous smear of DNA b/c DNA fragments are not distinct sizes.
T/F: Nuclear chromatin condensation occurs in both apoptosis and necrosis
TRUE
T/F: Nuclei Shrink, fragment, and dissolve in apoptosis and necrosis
FALSE.

DNA shrink and fragment in both, but its only in necrosis that DNA dissolve
T/F: Membrane integrity is upheld in apoptosis
TRUE

Membrane-bound cell fragments "bleb" off
Which involves a larger area of tissue, apoptosis or necrosis?
necrosis

Apoptosis involves isolated cells or small groups of cells
Which involves an inflammatory response, necrosis or apoptosis?
necrosis

Apoptosis does not involve an inflammatory response (makes sense, since it occurs naturally = programmed cell death)
What are the 4 types of necrosis?
Coagulative
Caseous
Liquefactive
Fat Necrosis
Define Coagulative necrosis
Denaturation of cytosolic proteins

Basic cell outline preserved = OVERALL ARCHITECTURE MAINTAINED

Shrunken, increased eosinophilia (dec. pH)

Pyknosis (nuclear shrinkage/darkening)
Karyolysis (nuclear dissolution)
Karyorrhexsis (fragmentation of nucleus)
Nuclei dissapear within a few days
Define Liquefactive Necrosis
Complete enzymatic digestion of dead cells

ex. = bacterial and fungal infections

Pus. ew.

Tissue Architecture LOST (no cell outlines when looking at in under microscope)
Define Caseous Necrosis
Cheesy, white/yellow crumbly (friable) material produced

No Tissue Architecture

Surrounded by "granulomatous" inflammation

Seen in tuberculosis
What types of necrosis preserve cell architecture? (Coagulative, Caseous, Liquefactive, Fat Necrosis?)
Coagulative
Define Fat Necrosis
Destruction of Adipocytes in fat tissue

Large scale destruction can lead to soponification = where soap is created = deposition of calcium salts in tissue (as fat cells die, they release Ca2+)
Whats the difference between "dry" and "wet" Gangrenous Necrosis
Dry = Coagulative necrosis secondary to profound ischemia

Wet = Liquefactive necrosis from inflammatory necrosis superimposed on ischemic necrosis (secondary to bacterial infection at site of ischemic damage)
List some Physiological examples of Apoptosis in the body
Involution of structures during development

Elimination of immune cells

Involution following hormonal withdrawal (ex. = menstruation)

Cytotoxic T cell mediated elimination of infected cells or neoplastic cells
List some Pathological examples of Apoptosis in the body
"Insults" i.e. radiation, drugs, DNA damage

Viral Infections

Autoimmunity

Neoplastic cells
Where do both the intrinsic and extrinsic stimulus pathways lead in apoptosis?
They lead to the common execution pathway, via Caspase Enzyme Cascade
What are the two ways Apoptosis can occur, and list some examples
Intrinsic = radiation, toxins, ROS free radicals, withdrawal of growth factors

Extrinsic = Death receptors: Fas (CD95), FasL (Fas Ligand CD95L) TNFR1 and TNF, Cytotoxic T-cell-mediated = Granzyme B

Note: Pretty sure this slide is backwards in terms of intrisic and extrisic.
What are some examples of Pro-Apoptotic Bcl Family mediators?
Pro-Apoptotic - Kill Cell

Bax (think of Katie Goodin's Dog, Bax, who killed rabbits for fun, bastard)

Bak
What are some examples of Anti-Apoptotic Bcl Family mediators?
Anti-Apoptotic = Cell Lives

bcl-2

bcl-x
What does P53 do?
Its a tumor suppressor gene; part of the INTRINSIC Apoptosis pathway

Arrests cell in G1 to repair DNA

If unsuccessful repair - apoptosis
How do you alter the Bcl family of proteins to FAVOR Apoptosis?
Favor Apoptosis = Inc. Pro-Apoptotics =

Inc. Bax and Bcl
Dec. bcl-2 and bcl-x
How do all stimulus pathways get cells to die via apoptosis?
Caspases (enzyme cascade)

Disruption of the balance of pro-apoptosis and anti-apoptosis can lead to disease
Too Little Apoptotic activity can lead to what?
Cancers
Autoimmunity
Too Much Apoptotic activity can lead to what?
Neurodegenerative disorders (for ex. Parkinson's)

Ischemic Injury

Death of virally infected cells
T/F: Accumulations can be Intracellular or Extracellular
TRUE
What are accumulations a manifestation of?
metabolic derangement
What are some of the normal Accumulations in cells?
Fatty Acids (Abnormal = steatosis = fatty liver from alcohol toxicity)

Cholesterol (atherosclerosis, xanthomas)
Is Steatosis (=fatty liver) reversible?
Yes
What are Xanthelasmas?
Lipid-laden macrophages

50% associated with hypercholesterolemia
List some other accumulations
Proteins
Glycogen
Pigments (Exogenous ex. = carbon dust, coal, smog)

Endogenous ex. = Melanin, Lipofuscin (wear/tear pigment that does NOT stain with iron), Hemosiderin (Hemolytic break down product)
What is Lipofuscin?
A "wear and tear" pigment that accumulates intracellular

Not Pathological
What is Hemosiderin Pigment?
Iron-containing pigment that is a breakdown product of Hb

We can see it via microscope with iron stain
What are some Pathological Calcifications?
Dystrophic Calcification

Metastatic Calcification
Define Dystrophic Calcification
Ca2+ deposited abnormally in tissues (normal systemic Ca2+ levels)

Associated with dying or degenerating cells

Ex. = calcific atherosclerosis and valvular disease (aortic stenosis bc of lots of cell death in that area = stimulus for dystrophic calcification), fat necrosis, certain neoplasms (Psammoma body)
What are some causes of Metastatic Calcification?
Inc. PTH (for various reasons) = Hypercalcemia = inc. serum Ca2+

Bone destruction (bc of tumors, other reasons) = inc. Ca2+

Too Much Vitamin D
T/F: Metastatic calcification may occur widely throughout the body
TRUE

Typically involves Gastric Mucosa, Kidneys, Lungs, Blood Vessels
Another name for cellular aging is?
Cellular Senescence
Whats the concept of replicative senescence?
cells have a limited ability to replicate (telomeres etc.)
T/F: As cells age, there is declining function of the proteosome
TRUE

= cellular machinery that eliminates unwanted or abnormal proteins within the cell
What are some Initiators of inflammation?
1. Infections
2. Trauma
3. Physical or Chemical Agents
4. Tissue Necrosis
5. Foreign Bodies
6. Hypersensitivity Immune Reactions
What are the cardinal features of inflammation?
heat, redness, swelling, pain
What are the general responses to Inflammation?
Vascular
Activation of Chemical Mediators
Migration and Activation of Cells (WBCs)
Termination/Resolution
How does acute inflammation differ from chronic inflammation?
Acute = Immediate, Rapid onset, short, sequential, innate immunity, neutrophils predominate

Chronic = Slower response/onset, cell mediated, lymphocytes/plasma cells/macrophages predominate, tissue destruction with granulation tissue and fibrosis
What are the Components of Inflammation?
1. Vasculature (Fluid, Chemical, Cells)
2. Connective Tissue (Matrix, Cells)
3. Parenchyma = the cell type that performs the major function in that tissue
When you alter the hemodynamics of a tissue, what is occurring?
Vasoconstriction and Vasodilation (redness/heat)
What is the net effect of vasodilation?
Increased Hydrostatic Pressure

Increased Interstitial Fluid (edema)
Contraction of endothelial cells leads to:
A. Decreased vascular permeability
B. Increased vascular permeability
Contraction of endothelial cells --> Increased Vascular Permeability --> Increased Interstitial fluid and cells that exit vessels (extravasation)
Define Transudate
Fluid leaking out of vessels due to increased venous pressure
Define Exudate
Increased vascular permeability due to forces that push fluid across endothelial barrier. Will contain a high protein concentration relative to transudate
T/F: A Transudate condition means increased Hydrostatic Pressure and Decreased Colloid Osmotic Pressure
TRUE

Transudate = extravascular fluid with LOW protein content (FLUID escapes vessel)

vs. Exudate = extravascular fluid with HIGH protein content (fluid AND protein escape vessel)
How do hydrostatic pressure and colloid osmotic pressure change in an exudate condition?
Decreased
What two things occur during the Exudate process to allow inflammation to occur?
Vasodilation and Stasis

Increased Interendothelial Spaces (=allows the fluid and protein to leak out)
T/F: The Transudate condition is synonymous with a non-inflammatory state
TRUE
Transudate and Exudate are associated with what vascular states?
Transudate = Non-inflammatory

Exudate = Inflammatory
Which is associated with a higher protein content, and thus a higher specific gravity, transudate or exudate?
Exudate
Define Effusion
Excess fluid in body cavities

(ex. = peritoneal, pericardial, pleural)
Define Serous
Yellow, Straw-like color, few cells
Define Serosanguinous
RBC's (red tinge)
Define Fibrinous
Large amounts of fibrin due to activation of coagulation cascade
Define Purulent
Large numbers of PMNs = white blood cells (neutrophils)
What are the origins of Endogenous Chemicals that activate and amplify the process of inflammation?
Plasma-derived

Cell-derived

ECM (extracellular Matrix)
Histamine, a principle chemical mediator of inflammation, is derived from what cells?
Mast cells, basophils, Platelets
Serotonin, a principle chemical mediator of inflammation, is derived from what cells?
Platelets
What are the newly synthesized mediators of inflammation in a cell undergoing inflammation?
Prostaglandins
Leukotrienes
Platelet-activating factor
ROS
Nitric Oxide
Cytokines
Neuropeptides
What products, newly synthesized during the inflammation process, are common targets for drugs?
Prostaglandins and Leukotrienes
Why does the body circulate inflammatory factors in their inactive form vs. their active form?
So we don't have inflammation all the time
What factors are activated from compliment activation that participate in the inflammatory process?
C3a, C5a, C3b, C5b-9 (membrane attack complex MAC)
What does Factor XII (12) activate?
Kinin System (Bradykinin)

Coagulation/ Fibrinolysis system (=breaks down fibirn clot)
T/F: The Compliment System involves 20-30 dif. proteins and 2 or 3 pathways.
TRUE

Classical - Ab-Ag Complexes
Alternative - Triggered by complex molecules = bacterial lipopolysacchardies (LPS)
What proteins involved in the complement system circulate in the blood in their inactive forms?
C1-C9
What are the functions of the complement system?
1. Phagocytosis/ Destruction
-Opsonization
-MAC
2. Vascular Inflammatory effects
-Anaphylaxis= inc. vascular permeability = vasodilation
3. Cellular Inflammatory Effects
-WBC Adhesion, activation, chemotaxis
Describe the Complement System
What are the Chemical mediators of Inflammation derived from plasma? (Hageman factor initiates what?)
Coagulation and Kinin Systems
Hageman Factor (Factor XII = 12) Initiates: "KCCF"
Kinin System
Coagulation
Complement System
Fibrinolytic System
What is the Kinin Cascade?
Bradykinin --> Increased Vascular Permeability --> Arteriolar Dilation --> Bronchial Sm. M. Contraction (it also causes pain when injected into skin)
What are the cell-derived chemical mediators of inflammation?
1. Vasoactive Amines
2. Arachidonic Acid (AA) Metabolites (from phospholipids)
3. Platelet-Activating Factor (PAF) (from phospholipids)
4. Cytokines
5. Chemokines
6. Nitric Oxide (NO = Vasodilation)
7. Lysosomal Molecules
-Acid and neutral proteases (=degrade cellular and ECM proteins)
8. Inferon (INF)
9.ROS
10. Neuropeptides (substance P = pain)
What are AA Metabolites derived from?
Phospholipids via Phospholipases

Steroids inhibit Phospholipases = thus inhibiting AA Metabolites
What is the function of the Lysosomal cell-derived chemical mediator of inflammation alpha-1-antitrypsin?
Inhibits PMN Elastase

Emphysema = Imbalance between protease and antiprotease results in alveolar wall destruction (=adjacent air spaces fuse together = very inefficient gas exchange = hyper-inflation of lungs)
Review the table of cell mediators of inflammation
What are the Acute cells of inflammation?
Neutrophils (=PMNs)
Eosinophils
Mast Cells
What are the chronic cells of inflammation?
Lymphocytes
Macrophages
Plasma Cells
Eosinophils (Mast cells)
Fibroblasts (prominet fibrosis)
T/F: Basophils can become Mast cells
TRUE
T/F: Monocytes can become macrophages?
TRUE
T/F: Lymphocytes can become Plasma cells
TRUE
What are the 3 outcomes of the inflammatory process?
1. Resolution
2. Healing by Fibrosis
3. Ongoing Chronic Inflammation
Gastric Ulcers result from chronic Acute Inflammation thanks to what bacteria?
Gastric Ulcers - Result from Chronic Acute inflammation process in stomach from H. pylori
What are the causes of Chronic Inflammation?
1. Persistent Infection or injurious stimulus
2. Prolonged exposure to injurious stimulus
-can be Endogenous or Exogenous
3. Autoimmunity
ex. = rheumatoid arthritis
Describe the process of chronic inflammation
1. Tissue infiltration by mononuclear cells (macrophages, lymphocytes, plasma cells, Histocytes are long-standing resident in tissues
2. Tissue Destruction
3. Tissue Repair
-Angiogenesis
Fibrosis

Typically results in Loss of Function
Describe the wide variety of tasks that macrophages do in terms of 1. Inflammation and Tissue Injury and 2. Repair
1. Inflammation and Tissue Injury = ROS and Nitrogen species
Proteases
Cytokines (including Chemokines)
Coagulation Factors
AA Metabolites

2. Repair
Growth Factors
Fibrogenic Cytokines
Angiogenic Factors
Remodeling (i.e. using collagen)
Define Granulomatous Inflammation
A distinct pattern of chronic inflammation

Aggregates of activated macrophages assume epitheliod appearance (ie it looks like an epithelium cell)

Some of the tissue coaleces to form a syncytium = multi-nucleated giant cell
If Necrosis is present, what do we call granulomatous inflammation? If no necrosis is present, what do we call granulomatous inflammation?
Necrosis = caseating granuloma

NO Necrosis = Non-Caseating granuloma
What are granulomas and what they do?
Granuloma = Mass of inflammatory cells

"Wall-off" offending agents (to contain them)
What are some systemic effects of inflammation?
Fever
Elevated Plasma Acute Phase Reactants (for ex. C-reactive protein = CRP, Fibrinogen
Leukocytosis (elevated circulating WBCs, such as PMNs = neutrophils and Lymphs)
Describe the phenomenon of RBC Rouleaux and ESR
RBCs overall negative charge = they repel each other = they don't clump together.

Infection = Acute Phase Proteins = As their neg. charge decreases, the RBCs can start coming together

Inc. Sed. Rate = Inc. Inflammatory Process
Describe the difference between renewal and Repair
Renewal = Normal Physiological process

Repair = When there's damage = Regeneration = a parenchymal component
Renewal and Repair begin with cell loss/injury- what are the responses to this?
Cell migration
Cell proliferation
Deposition/Reorganization/Remodeling of extracellular matix components
Give some examples of natural cell loss
Sloughed epithelia (skin, mucosa)

Balance of proliferation and apoptosis

Mechanical Loss
Give some examples of Pathologic cell loss
Injury/Inflammation destruction

Loss of Parenchymal cells

Loss of stromal cells (i.e. mesenchymal, fibroblasts, etc.) that support function
What function of natural cell loss maintains function?
Renewal
Are Regeneration and Fibrosis components of Renewal or Repair?
Repair
What are some processes involved in the repair of cells?
Regeneration of Cells

Healing - scar
Whats involved in the maintenance of parenchymal cell function?
balance of proliferative activity where cells become mature and enter into specific roles.

Cells not needed = Apoptosis
What regenerates cells in skin (hair + sweat glands), intestine, liver and corneal epithelium?
stem cells
In the liver, injury to ONLY hepatocytes is repaired how?

Injury to hepatocytes AND Matrix is repaired how?
If ONLY Hepatocytes, repair = Regeneration

If Hepatocytes AND MATRIX = Laying down of fibrous tissue
What types of fibers provide a scafolding in the liver for RBCs to percolate through and hepatocytes to attach to?
Reticular
Detail the process of repair in the liver if A. Only hepatocytes damaged B. Hepatocytes (parenchymal cells) and Scaffolding damaged
If only hepatocytes damaged, stem cells differentiate into hepatocytes

If Hepatocytes and scafolding damaged, stem cells don't have anything to attach to, so they form a regenerative nodule = there is a scar
List the Cellular components of repair
1. Inflammatory Cells
2. Parenchymal Cells
3. Fibroblasts and other Stromal Cells --> Repair Tissue (structural integrity)
4. Endothelial Cells = Angiogenesis
What are the two types of Fibrous Structural Proteins involved in the ECM?
Collagen
-Fibrillar: I (strong, late wound) II (Cartilage) III (early wound; hollow structures, V, IX
-NonFibrillar = IV (basement membranes)

Elastins
-Adhesion Proteins (CAM, Cadherins, Integrins)
-Proteoglycans and Hyaluronic Acid
Collagen, a fibrous structural protein, is made up of what types?
Fibrillar:
I = Strong, late wound
II = Cartilage (no wound-healing participation)
III = Early wound, Loose CT = flexibility
IV = Basement Membrane
What is the role of p53?
Cell cycle regulation

Tumor Supressor = prevents genome mutation

Can hold cell cycle at G1/S

Can initiate Apoptosis
In the healing of a wound, granulation tissue is eventually replaced by what tissue type?
collagen type I (=strong, late wound)
T/F: Superficial injury to the skin involves a lot of granulation tissue formation
FALSE. It involves minimal granulation tissue formation...primary goal = re-epithelialization
What is the overall goal of Healing and Fibrosis?
To maintain hemostasis = keep blood in blood vessels
List the steps in Healing and Fibrosis of wounds
Maintain Hemostasis
Inflammatory Response
Proliferation and migration of Parenchyma
Angiogenesis
Synthesis and Deposition of ECM
Tissue Remodeling
Wound Contraction
Acquisition of Wound Strength
List the sequence of events in the wound healing process
1. Hemostasis
2. Inflammation
3. Provisional Matrix
4. Granulation Tissue
5. Fibroblast Proliferation and Collagen deposition/remodeling (=get rid of collagen III, replace with collagen I)
6. Re-epithelialization
7. Wound contraction
8. Inc. in wound strength
What are the three overall phases of wound healing?
Inflammation

Proliferation

Maturation
Clot formation and chemotaxis occur in what stage of wound healing?
Inflammation
Granulation tissue involves lots of what type of collagen?
Collagen type I
Scar tissue involves lots of what type of collagen?
Collagen Type III
What are some conditions that can affect the repair of wounds during the healing process?
Location (ie ability of wound to contract less on scalp)

Amount and nature of ECM at Site of injury (older people have less collagen/ elastin)

Blood Supply (dec. blood supply = dec. healing capacity)
What are the local factors that can affect wound healing?
Infection
Mechanical Forces
Foreign Bodies
Size, Location, Type of Wound
What are some Systemic factors that affect wound healing?
Nutritional status
Metabolic status
Circulation
Hormones
What factors lead to cirrhosis of the liver?
Fibrosis with continued parenchymal and ECM damage
What are the key differences between primary skin wound healing and secondary skin wound healing?
Primary = neutrophils, clot, mitosis, macrophage, fibroblast, fibrous union

Secondary = Neutrophils, MANY new capillaries, wound contraction
Whats the consequence of chronic hepatic injury?
Formation of regenerative nodules separated by fibrous bands
If the kidney is damaged, and injury does NOT significantly damage the ECM framework, what can we expect from the renal tubular epithelium? What if there is damage to the ECM framework?
epithelium will recover

Damage to ECM framework = scarring = we lose function
T/F: Metaplastic processes are reversible
TRUE, as long as the stimulus is removed.
If the underlying stroma of the lung is damaged, what happens to the tissue?
Fibrosis = impairs gas exchange
Loss of cardiac myocytes results in what?
Fibrosis
Diffuse loss of cells in the heart leads to what?
Myocardial Infarction, following Coagulative Necrosis
T/F: The heart can only replace damaged/lost cells with a scar
TRUE
In Lung, what are our two options in terms of repair?
If damage limited to pneumocytes, that's OK bc they'll divide/replicate

I damage to Alveolar epi AND scaffold/matrix = thickened fibrosis - inefficient respiration
T/F: Axons in PNS can elongate/grow and remake synapses, while CNS axons cannot
TRUE
How is scarring accomplished by neurons?
PNS = Fibrosis
CNS = Glial cell proliferation
What are some problems with excessive wound healing?
1. Excessive scar formation

2. Keloid

3. Excessive Contraction = Contracture (palms, soles)
What is a Keloid?
Over-deposition of collagen within dermis = the keloid extends beyond the anatomic confines of normal structure
What type of excessive wound healing problem is common when someone is burned?
excessive contraction = Contracture = deformity of wounded tissue and surrounding structures

For ex., when people burn their hands, excessive contracture can make them have to hold their hands inwards, with wrist bent in.
What are some examples of Excessive Regeneration/Repair that can occur during wound healing?
Excessive Granulation tissue formation
-Pyogenic Granuloma
-Proud Flesh

Excessive Fibrosis after injury
-Desmoids
What force within blood vessels allows the vessel to hold onto or maintain fluid?
Plasma colloid osmotic pressure
What force within blood vessels allows the vessel to lose blood?
Hydrostatic Pressure
The primary insult in a hemodynamic disorder has to do with what factors?
Tissue injury and too much bleeding (or too much clotting)
A secondary insult in a hemodynamic disorder has to do with what?
secondary hemodynamic disorder = something in body is affecting blood vessels

examples = congestive heart failure, shock, liver disease (chronic alcohol abuse)
What is Edema?
excessive fluid accumulation in spaces outside blood vessels.

Know some causes of edema
What are 3 major causes of systemic edema?
Heart Failure

Mal-Nutrition (remember mal-nourished kids in DR with edema...)

Anything leading to decreased Synth. of albumin by liver, i.e. nephrotic syndrome = lose a lot of protein in urine
How does decreased plasma albumin lead to edema?
Dec. plasma albumin = Dec. Oncotic Pressure = Edema

OR Inc. Hydrostatic pressure = Edema (directly) (for. ex. Inc. Na+)

Heart Failure = Inc. Capillary Hydrostatic Pressure = Dec. Blood flow (Inc. blood vol.) = Edema
Whats the difference between dependent edema and pulmonary edema?
Dependent edema = gravitational influences on interstitial edema (RV failure predominates)

Pulmonary Edema = Congestion (LV failure predominates)
Whats the difference between Generalized edema and Cerebral Edema?
Generalized = renal dysfunction, nephrotic syndrome

Cerebral Edema = Localized; infection neoplasm, stroke
-Vasogenic = vessel damage
-Cytotoxic = Intracellular
-Interstitial = Extracellular
Whats the difference between Hyperemia and Congestion?
Hyperemia = Active, Blood flow INTO tissue

Congestion = Passive, Blood flow OUT of tissue
Define Hyperemia
Active increase of blood INTO a tissue
Define Congestion
Passive, impaired out-flow of blood OUT of a tissue (cOngestion - Out)
What are the 4 types of Hemorrhage?
"HEPP"

Hematoma = large collection of blood in tissue
Ecchymosis = Bruises
Petechiae = Pinpoint hemorrhages
Purpura = Red/purple skin discoloration
Between a Petechiae hemorrhage and a Hematoma Hemorrhage, which causes the least amount of vessel damage?
Petechiae (bc its pinpoint hemorrhages instead of a massive large hemorrhage)
Is hemostasis a pathological or physiological process?
Physiological
Are thrombosis and bleeding pathological or physiological processes?
Pathological
What are the steps of hemostasis when a injury to the skin occurs?
1. Vasoconstriction
2. Primary Hemostasis = platelet plug (subendothelial ECM, vW Factor)
3. Secondary Hemostasis
4. Localized Thrombus Formation
T/F: At the same time coagulation is going on, anti-coagulation is going on to keep coagulation localized
TRUE
What happens to the clotting process if we don't have von Willebrand Factor (Factor XII, 12)?
Adhesion is negatively affected = cells don't adhere = no clotting
What are some Pro-Thrombotic vs. Anti-Thrombotic Factors?
Pro = platelets, Coagulation, Anti-fifrinolytic (=eats frinolytic=more fibrin= pro-clotting)

Anti = Intact Endothelium, Anti-Caogulation, TFP1, Fibrinolytic
What does Protein C do in the process of inhibiting thrombosis?
Proeolysis of factors 5a (Va) and 8a (VIIIa)

Requires Protein S
What does a deficiency of Protein C or Protein S mean in terms of clotting?
Dec. Protein C or Protein S = Over-Clotting
Coagulation requires what two substrates to make the assembly substrate?
Calcium and Phospholipid
How do we test for the presence or absence (or just general amount) of clotting factors in the blood?
Give a Ca2+ kelator = Ca2+ removed = Blood inhibited from clotting = we can test presence of clotting factors that are free flowing in blood
What are 3 "natural" anticoagulants?
Anti-thrombins
Protein C and Protein S
TFPI
T/F: The Extrinsic and Intrinsic pathways lead to the common pathway in the coagulation cascade
Yep.
The Intrinsic pathway of coagulation is made up of what types of factors? (contact or tissue?)
Contact factors
The Extrinsic pathway of coagulation is made up of what types of factors? (contact or tissue?)
Tissue Factors
The bulk of lab testing of the Coagulation Cascade is performed in:
A. Primary Hemostasis
B. Secondary Hemostasis
C. Fibrinolysis
B, Secondary Hemostasis

-Pro-thrombin Time (PT), aPTT (activated partial thromboplastin time), TT (Thrombin Time
Whats the path of Fibrinolysis?
Plasminogen --> Plasmin --> cleaves Fibrin --> which leads to Fibrinolysis (one FSP = D-Dimer)
D-Dimer measurement in the lab is important in to tell us what?
How much blood clot break down is occurring
Thrombosis is a Pathological process because of what?
Endothelial Injury (*most significant)

Stasis

Hypercoagulable State
What is the most significant factor that promotes thrombosis?
Endothelial Injury
What are the causes of Endothelial Injury?
Hypertension

Turbulent Flow

Inflammation
T/F: Congestion and Turbulence, which disrupt normal laminar flow through vessels, can lead thrombosis
TRUE
How does Primary Thrombosis differ from Secondary Thrombosis?
Primary Thrombosis = Genetic

Secondary Thrombosis = Acquired (secondary to other disease conditions)
What are three Primary Hypercoaguable States?
1. Factor V (Leiden) gene mutation
2. Prothrombin Gene Mutation
3. Hyperhomocysteinemia
What is a Factor V (Leiden) Gene mutation?
Factor V is resistant to cleavage by Protein C = can't inactivate Factor 5 - its active and promotes clotting
What is a Prothrombin gene mutation?
Excessive amounts of Pro-thrombin = Hypercoaguable
What is Hyperhomocysteinemia?
Inhibition of Anti-Thrombin III and Thrombomodulin (can be acquired OR genetic)
Name some examples of Acquired Hypercoaguable states
Things associated with stasis or vascular injury = heart failure, MI, atherosclerosis, inactivity

Hepatic (Inc. Synth. of Clotting Factors, Dec. Synth. of Anti-Thrombin III)

Cancers

Heparin-induced Ab's that activate platelets

Obesity, Smoking. SO DON'T DO IT!

Antiphospholipid Syndrome
What are the fates of a thrombus, once formed?
Dissolution
Propagation
Organization/Recanalization
Embolization
What is DIC, Disseminated Intravascular Coagulation
Widespread thrombin activation with production of fibrin thrombi with microvascularture

Can result ultimately in increased bleeding (=platelet count decreases. More prominent in brain, kidney, heart, lungs, liver)
What is an embolism?
Dislodged or detached portion of solid, liquid or gas that travels through blood vessels (=Hypoxia and Ischemia)

WHERE Thrombi is determines which circulatory system affected (pulmonary or systemic) and likelihood of impairing blood flow to certain organs
What are the dif types of embolisms?
Fat
Air/Gas
Tumor
Blood Clot (Thromboembolism)
Pulmonary Embolisms are most frequently derived from what?
DVT's
What percent of cardiac thrombi are derived from cardiac mural thrombi?
80%

Most from LV MI
What percent of infarcts are due to Thromboembolic events?
95% (most occur in heart and brain)
What are the different types of Infarct?
Hemorrhagic = Red = Venous Occlusion
Anemic = White = Arterial Occlusion
Most Infarcts (except the brain) include what type of tissue cell death?
Ischemic Coagulative Necrosis

(EXCEPTION = BRAIN = LIQUEFACTIVE NECROSIS)
Development of an infarct is dependent on what?
Rate of vascular decline (if infarct is not severe, damage will occur more slowly)
What are the relative times the brain, heart, and fibroblasts are vulnerable to hypoxia?
Brain = 3-4 min
Heart = 20-30 min
Firbroblasts = Many hours
What is Shock?
Cardiovascular collapse with global hyperfusion of tissues and subsequent anoxia/ischemia

Microvascular Thrombosis
Vasodilation = inc. permeability = dec. perfusion
Immunosuppression
What are the major causes of shock?
Cardiogenic (heart failure)
Hypovolemic
Septic
Neurogenic
Anaphylactic
What causes Cardiogenic Shock
MI
Cardiac Arrhythmias
Pulmonary embolism
Cardiac Temponade
What is Hypovolemic shock?
Reduction of fluid volume some way or another

Things than can cause it:
Burns
Severe Dehydration
(diarrhea, vomiting, excessive perspiration)
Lack of fluid intake to restore blood volume
What is Septic Shock?
Bacterial Infections

Gram Negative Septicemia; bacteria cause vasodilation and hypotension
Disseminated Intravascular Coagulation (DIC)
Multiple End-Organ Failure
"Shock Lung"
Mortality of Septic Shock = 25-50%
What are some causes of Neurogenic Shock?
Anesthetic States
Brain/Spinal Cord injury
What is Anaphylactic Shock?
General Vasodilation from Type I Hypersensitivity Reactions
What are the Stages of Shock?
Stage I = Compensation (inc. sym. tone)
Stage II = Decompensation (dec. tissue perfusion)
Stage III = Irreversible (necrosis and organ failure)
What is Thrombocytopenia?
Decreased Platelets = spontaneous bleeding (reduced clotting)

Normal PT and PTT

Increased Destruction of TTP (thrombocytopenic Purpura)
What is Von Willebrand Disease?
Decreased amount of vWF

Spontaneous bleeding through mucous membranes

Normal PT, may see prolonged PTT
What are some clinical bleeding Caogulation disorders?
Hemophilia A
Hemophilia B
Acquired Bleeding Disorders (Vit. K deficiency, Liver disease)
What is Clinical Antiphospholipid Syndrome (APS)?
Ab's directed against phospholipids
Developmental Abnormalities typically result in A. Congenital B. Hereditary/Familial Disorders
A. Congenital
What are the 3 classifications of genetic disorders?
1. Chromosomal
2. Disorders related to single genes with pronounced effects
3. Complex multigenic disorders
Whats the most common genetic abnormality?
Multigenic (=variable contribution of many dif. alleles)
T/F: Of the known causes of congenital abnormalities, the majority are heritable in cause
TRUE
Define Angenesis
Absence of organ or anlage (Anlage = initial clustering of embryonic cells from which organ develops)
Define Aplasia
Persistence of Anlage or rudiment w/o mature organ
Define Hypoplasia
Reduced size from incomplete development
Define Dysraphic Anomaly
Failure of opposed structures to fuse
Define Involution Failure
Inappropriate persistence of structures beyond developmental stage
Define Division Failure
Incomplete cleavage of embryo
Define Atresia
Incomplete formation of lumen or tubular structure
Define Dysplasia
Abnormal histogenesis
Define Ectopia/ Heterotopia
Organ/Tissue situated outside normal location
Define Dystopia
Inadequate migration of organ, which remains in developmental location
Teratogenic exposure In-Utero has the most significant deleterious effects before what time point?
16 weeks

Week 3--> Week 16 = teratogens have major morphogenic affects
What are the signs/symptoms of FAS, Fetal Alcohol Syndrome?
Small Head (Microcephaly)
Epicanthal folds (=skin fold of upper eyelid)
Short Palepbral Fissure (separation between upper+lower eyelid)
Maxillary Hypoplasia
Thin Upper Lip
Poorly formed Philtrum
Cardiac Septal Defects
Cognitive/Behavior effects
What are some Infectious Biological Teratogens? (Teratogen = interferes with embryo development)
"TORCH"

Toxoplasma
Other (Syphilis, Varicella-zoster, EBV, TB)
Rubella
CMV
Herpesvirus (HSV 2)
What are some of the signs/symptoms if a baby is exposed to some of the infectious biological teratogens (TORCH)?
small head
calcifications present
small, closely-set eyes
petichiea purpura (scattered over skin/face)
What do cytogenics tell us?
Abnormalities in chromosome number and/or structure
When do chromosomal abnormalities arise?
Somatic cell division (mitosis) (Abnormalities in chromo # varies by cell)

OR

Gametogenesis (if chromo # changed here, all cells will have same # chromos)
What are some abnormal chromosome structures?
Translocations
Deletions
Inversions
What is balanced reciprocal Translocation of chromosomes?
When chromsomes mix equally (as opposed to Robertsonian centric fusion, when chromos mix unequally)
What is Robertsonian centric fusion of chromosomes?
When chromozomes mix unequally, for ex. there is one small chromo and one huge chromo created
What is Trisomy 21?
Downs Syndrome
What is Trisomy 13?
Patau's Syndrome
What is Trisomy 18?
Edward's Syndrome
What's the most common cause of congenital (=at birth) retardation?
Trisomy 21, Down's Syndrome
T/F: Aberrations of sex chromosome number typically produce less severe results than abnormal numbers of autosomes
TRUE

Ex. = Klinefelter Syndrome (testicular diagenesis)
What is Turner Syndrome?
Complete or partial loss of one X chromosome

-Sexual infantilism (poor development)

Neck "wings" out

Possible genetics: 45, X
What are some of the single gene mutations?
Point mutations
Frameshift mutations
Trinucleotide Repeat
What are some examples of Mendelian Inheritance of Genetic Mutations
Autosomal Dominant
Autosomal Recessive
Sex-linked Dominant
Sex-Linked Recessive
Codominance
Importance of Penetrance and Expressivity
T/F: Most Autosomal Dominant genetic mutations are expressed in a heterozygous state
TRUE
What is Ehler's Danlos-Syndrome?
Connective tissue autosomal dominant disorder
What is the difference between NF1 and NF2, both Neurofibromatosis diseases?
NF1 = Von Recklinhausen disease. Chromosome 17. Dark skin pigmentation (cafe-au-lait spots...think of woman at festival...)

NF2 = Central neurofibromatosis. Chromosome 22. Tumors of Schwann Cells
Whats the most common and lethal Autosomal Recessive genetic disorder?
Cystic Fibrosis (1 in 25 caucasians)

Chloride channel mutation = thick mucous

Cl- moves IN cell = Na+ moves IN cell = water follows Na+ = mucous dehydrated = mucous thick = Clogged pancreatic duct = pancreatitis = improper # digestive enzymes = No lipases = can't digest fat = fatty diarrhea (statorhea)
What is Lysosomal Storage Disease?
Defect in Lysosomal Hydrolases

Ex. = Gaucher Disease (deficiency in glucosylcerebrosidase)
What is Tay-Sach's Disease?
Ashkenazi Jews

GM2 ganglioside accumulates in lysosomes, (neurons of brain and cells of retina)

Progressive motor, cognitive deterioration
What chromosome does Cystic Fibrosis affect?
7q
What chromosome does Sickle Cell Anemia affect?
11p
What chromosome does Gaucher disease affect?
1q
What chromosome does Tay-Sachs affect?
15q
What is Fragile X Syndrome?
Most Common form of INHERITED (not congenital, which is Down's Syndrome) Mental Retardation
List some X-linked Recessive Disorders
Fragile X Syndrome
Hemophilia A (Factor VIII deficiency)
Duchenne-Becker Muscular Dystrophy
What is a trinucleotide repeat disorder?
Same 3 Nucleotides repeat over and over

The number of repeated segments often increases with successive generations
What are some examples of trinucleotide repeat diseases?
Huntington Disease

Fragile X Syndrome
What are some examples of X-linked dominant Diseases?
Vitamin D-resistant Rickets

Alport's Syndrome
What type of genetic disorders are probably the most common cause of genetic disease?
Complex Multigenic Disorders

Ex. = Atherosclerosis and Diabetes