• 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

1/222

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;

222 Cards in this Set

  • Front
  • Back
define homeostasis
a state in which all symptoms are
in balance at an ideal “set point” despite
alterations within the body
define homeostatic response
mechanistic, predictable
series of orchestrated internal events
define allostasis
addresses complexities and variable
levels of activity necessary to maintain or reestablish
homeostasis
what is allostasis carried out by?
a superordinate set of systems
that support homeostasis in light of
environmental and lifestyle changes
define stress
Stress: physical, chemical, or emotional factor
resulting in tension of body or mind
 Or an actual state that tension produces
is all stress bad? Give example
Not all stress is bad
For example, bone loss is prevented by weight or
stress on the skeleton
what are the three components of General Adaptation 'Syndrome? (GAS)
an alarm reaction
a stage of resistance
stage of exhaustion
at what stage of GAS does pathologic development occur?
stage of exhaustion
describe the alarm reaction of GAS
fight‐or‐flight response due to
stressful stimulus
describe the stage of resistance
The activity of the nervous
and endocrine systems in returning the body to
homeostasis
describe the stage of exhaustion
point where body can no
longer return to homeostasis
what is allostatic overload?
“cost” of body’s organs and tissues for an excessive or ineffectively regulated allostatic response
what are agents or conditions that can produce stress and endanger homeostasis
stressors
what are conditions or
situations that increase the likelihood of
encountering a stressor
risk factors
which body system response mediates
the flight‐or‐fight response
sympathico-adrenal
which neurohormonal mediators play an integral role in allostasis?
Catecholamines: norepinephrine and
epinephrine
what are the two adrenocortical steroids involved in homeostasis?
cortisol and
aldosterone
what are the body's natural pain relievers?
Endorphins and enkephalins: endogenous
opioids
what substances are secreted
by macrophages during stress response, thus
enhancing immune system response
immune cytokines
what are the sex hormones which affect stress responses
estrogen testosterone, and dhea (dehydroepiandrosterone)
how do the following affect strees responses
1. estrogen
2. testosterone
3. DHEA
Estrogen: “calming” effect during stress
 Testosterone: ramps up fight or flight response
(↑BP, ↑HR)
 DHEA: counteracts the catabolic effects of
glucocorticoids on bone; counteracts depression
associated with glucocorticoid secretion
How do the following affect stress responses:
1. growth hormone
2. prolactin
3. oxytocin
Growth hormone: can increase during stress to
enhance immune function
 Prolactin: similar to structure of growth
hormone; role in immune response
 Oxytocin: produced during childbirth and
lactation; associated with bonding and social
attachment; thought to moderate stress
response
what are the effects of stress response influenced by?
influenced by
genetics, culture, prior susceptibilities,
preexisting health status, allostatic state, and
ability to manage stress
what is the biopsychosocial process of change
in response to new or altered circumstances,
internal or external in origin
adaptation
what is behavioral adaptive response to a stressor using culturally based coping mechanisms
coping
what is allostatic overload? what results from it?
inadequate adaptation
mechanisms or overwhelming allostatic load;
results in inability to maintain homeostasis
 Pathophysiologic states result affects all areas of
the body and emotional systems
what is usually the first manifestation of most forms of reversible cell injury?
hydropic swelling: cellular swelling due to accumulation of water
what causes hydropic swelling?
malfunction of sodium‐potassium
pump with accumulation of sodium ions within
the cell
 “water follows salt” rule
 Any injury that results in loss of energy (ATP)
will also result in swelling
describe what hydropic swelling is characterized by
Characterized by large, pale cytoplasm; dilated
endoplasmic reticulum; and swollen mitochondria
Generalized swelling (hydropic) of cells in organs can lead to what?
increase in size and weight, indicated by “megaly”
 i.e., liver, heart, spleen
what is another cause of reversible cell injury besides hydropic swelling?
intracellular accumulations
describe intracellular accumulations
Excess accumulations of substances in cells may
lead to cellular injury due to toxicity, immune
response, and/or taking up cellular space
 Characterized by:
 Excessive amounts of normal intracellular
substance
 Lipids, CHO, proteins, glycogen
what are the four causes of intracellular accumulations
1. Abnormal metabolism (i.e.,
fat)
2. Defect in protein
folding transport (i e heat Shock Proteins)
28
i.e., 3. Enzyme deficient or
absent (i.e., inborn errors of
metabolism)
4. Accumulation of indigestible
materials (i.e., Ca+2, dust
where is a common site of intracellular accumulations?
the liver
lipid accumulation in hepatic cells would be inconsidered
intercelluar accumulation
what are the 5 kinds of cellular adaptation?
atrophy
hypertrophy
hyperplasia
metaplasia
dysplasia
when cells shrink and reduce their differentiated
functions in response to normal and injurious factors, its called
atrophy
what are some general causes of cell atrophy
Disuse
 Denervation
 Ischemia
 Nutrient starvation
 Interruption of endocrine signals (i.e., GH)
 Persistent cell injury
 Aging
.
.
when there is increase in cell mass accompanied
by an augmented functional capacity in response to physiologic and pathophysiologic
demands, it is called:
 Cell size increases/cell number remains same
hypertrophy
define hyperplasia
increase in functional capacity
related to an increase in cell number due to
mitotic division
 Usually in response to increased physiologic
demands or hormonal stimulation
 New cells
define metaplasia
replacement of one differentiated cell type with another
 Most often as an adaptation to persistent injury,
with replacement of a cell type that is better
suited to tolerate injurious stimulation
 Fully reversible when injurious stimulation is
removed
define dysplasia
disorganized appearance of cells
because of abnormal variations in size, shape,
and arrangement
 Represents an adaptive effort gone astray
 Significant potential to transform into cancerous
cells, thus referred to as preneoplastic lesions
describe necrosis
Usually occurs as a consequence of ischemia or
toxic injury
 Characterized by cell rupture, spilling of contents into extracellular fluid, and inflammation
what are the four types of necrosis?
1. coagulative
2. liquefactive
3. fat necrosis
4. caseous necrosis
describe coagulative tissue necrosis
(most common type): process that
begins with ischemia and ends with degradation
of plasma membrane
 Ischemialoss of PM ability to maintain electrolyte
gradient↑↑Ca+2 influx/mitochondrial
dysfunctiondegradation of PM and nuclear structures
describe liquifactive tissue necrosis
occurs with dissolution of dead
cells, liquification of lysosomal enzymes, and formation of abscess or cyst from dissolved dead tissue
fat necrosis involves what?
What is it usually the result of?
describe it
Death of adipose tissue
 Usually Usua y the result of trauma or pancreatitis
 Appears as a chalky white area of tissue
describe caseous necrosis
Characteristic of lung damage secondary to tuberculosis
 Resembles clumpy cheese
 White, soft, fragile tissue—dead cells walled off
what is Casper's law? (extra credit)
if all other factors are equal, then, when there is free access to air, a body decays twice as fast compared to if immersed in water and eight times faster compared to burial in earth. Bacterial decomposition is dependent on temperature.
.
.
Barrett's Esophagust is an example of
metaplasia
In GERD), there is a backflow of gastric contents through the lower esophageal sphincter. The highly acidic gastric contents cause damage to the lining of the esophagus, which is coated with normal squamous epithelium (like inside of your cheek). Over time, columnar tissue replaces the squamous epithelium.
how long does it take for rigor mortis to set in? Why does it happen?
Within 6 hours, Ca++ accumulates and with failure of the ATP pumps the actin-myosin cross-bridges (in skeletal muscle) become permanently fixed—thus the stiffness of the muscles
Cellular death in a large area of tissue
 Results from interruption of blood supply to a
particular part of the body
is called:
gangrene
dry gangrene is a form of what type of necrosis?
coagulative
wet gangrene is a form of what type of necrosis?
liquefactive necrosis - typically found in internal organs
what's another word for Cell suicide?
apoptosis
what causes apoptosis
Occurs in response to injury that does not
directly kill the cell, but triggers intracellular
cascades that activate a cellular suicide response
describe apoptosis
Not always a pathologic process
 Does not cause inflammation
 “Clean” process compared to necrosis
 Less damage, inflammation
what are the Two Types of Environmental or Extrinsic
Signals that May Induce Apoptosis
Withdrawal of “survival” signals that normally
suppress apoptotic pathways, such as observed
with cancer cells
 Extracellular signals, such as the Fas ligand,
bind to the cell and trigger death cascade
through activation of “death receptors
.
.
what is the most common cause of cell injury?
ischemia
why does ischemia injure cells faster than hypoxia alone?
because ischemia results in a Combination of disruption of oxygen supply with accumulation of metabolic waste
what are the 5 kinds of cellular adaptation?
atrophy
hypertrophy
hyperplasia
metaplasia
dysplasia
when cells shrink and reduce their differentiated
functions in response to normal and injurious factors, its called
atrophy
what are some general causes of cell atrophy
Disuse
 Denervation
 Ischemia
 Nutrient starvation
 Interruption of endocrine signals (i.e., GH)
 Persistent cell injury
 Aging
.
.
when there is increase in cell mass accompanied
by an augmented functional capacity in response to physiologic and pathophysiologic
demands, it is called:
 Cell size increases/cell number remains same
hypertrophy
give an example of apoptosis triggered by intrinsic pathways
In response to severe cell damage, a protein (p53,
which is normally low in the body) will increase
in response to cellular DNA damage, triggering
the cell’s own death
 Involves numerous intracellular signals and
enzymes
describe the etiology of hypoxia induced cellular injury
Hypoxia causes ATP
To stall, Na+/K+/Ca++
Pumps then fail
Hydropic swelling,
Ca++ buildup damages
mitochondria…
end product of glycolysis
(pyruvate) accumulates
and converts to lactate
causes lactic acidosis which damages the cell membrane
.
.
nutritional injuries to the cells may result for deficiencies caused by
Poor intake
 Altered absorption
 Impaired distribution by circulatory system
 Inefficient cellular uptake
cell injury may result form infectious and immunologic injury depending on its ____________
added injury may occur indirectly by triggering _______``
virulence
the body's immune response
Toxic chemicals or poisons can cause cellular
injury both indirectly and by becoming __________ by the body
metabolized into reactive chemicals
what are some physical and mechanical injury causes?
Extremes in temperature
 Abrupt changes in atmospheric pressure
 Mechanical deformation
 Electricity
 Ionizing radiation
what is the theory by which cellular aging occurs?
chromosomes shrink as we age and eventually the telomeres become too short to replicate
what is somatic death? describe it
Death of the entire organism
 No inflammation or immunologic response
occurs prior to death
 General features include cessation of
respirations and heartbeat
what do the nucleotide base abbreviations
G
C
A
T
U
stand for
guanine
cytosine
adenine
thymine
uracil
what are the two types of nucleotide bases?
pyrimidines: single-ring structures (cytosine & thymine)
purines: double-ring structures (guanine & adenine)
what type of bonds link the nucleotide bases to eachother?
hydrogen bonds: weak bonds that are easily broken and don't cause damage when broken
a nucleotide consists of what?
a sugar (deoxyribose), a phosphate group, and one of the four nucleotide bases.
nucleotides are joined by ____________to form long chains called ____________
repeating sugar-phospate bonds
polymers
the DNA polymers are then wound around ________ to form bead-like structures called__________
histones
nucleosomes
the nucleosomes become tightly coiled into _________ during cell division
chromosomes
what do initiator proteins do?
pull DNA strands apart at the replication origins (the starting point for replication)
during dna replication, what happens when the undwinding DNA becomes overly twisted?
topoisomerases cut nicks in DNA
 Allow it to unwind, and prevent tangling
 Ligases repair nicks
what does DNA polymerase do?
binds to the single DNA strands and begins the process of forming new complementary DNA Strands
Match appropriate base to template base
 Catalyze formation of sugar‐phosphate bonds
that form backbone of DNA strand
.
.
Replication proceeds along DNA strand from ___
end toward ____ end only
3' towards 5'
a linear sequence of DNA that codes for a particular protein is called _______
a gene
why is DNA replication said to be assymetrical?
Leading strand replicated as a continuous
polymer
 Lagging strand must be synthesized in short
sections
 Okazaki fragments sealed together by DNA
ligase to form unbroken DNA strand
87
what is needed to replicate the telomeres (chromosome ends)
telomerase
How is DNA replication self-correcting
DNA polymerase removes incorrect nucleotides
and replaces them with correct ones
cells have ____ different types of amino acids

amino acids join in a specific sequence to form a particular ________
20

protein
Proteins are composed of one or more chains of ________
amino acids
what is needed to code for each of the 20 aminio acids
three nucleotides (triplet)
how many triplet combinations are there? what are they called?
64
codons
stop codons are coded for by _____ and signal __________
three triplets
protein code
gene regulation is accomplished through control of _________________
the transcription process (polymerase binding)
what are introns
describe them
Unwanted stretches of bases that must be
removed from pre‐mRNA before it can be
translated into a protein • Removed by nucleus • Result in exons (mRNA sequence with wanted
segments) • Believed to be important in evolution of new
genetic information and gene regulation
what do the large units of ribosomes do
Catalyzes peptide bond formation between
incoming amino acids
Translation
 Find starting place on mRNA
 Set correct reading frame for codon triplets
 Move along mRNA
 Translate nucleotide sequence into amino acid
sequence
what do the ribosomal smaller subunits do?
Smaller subunit
 Binds mRNA and tRNA
where does transcription take place
in the cell cytoplasm (mRNA is transported to the cell cytoplasm through nuclear membrane pores)
Newly synthesized protein chain released from
ribosome when reaches __________`
stop codon
describe tRNA
Anticodon at one end
 Amino acid attachment at other end
 Anticodon formed by 3‐nucleotide sequence
what happens to the newly synthesized protein chain that is released from the ribosome when it reaches a stop codon?
New protein bound by chaperone proteins that
help it fold into shape``
what are the transcriptional controls in the regulation of gene expression?
Activators
 Positive controls “turn on” transcription
 Repressors
 Negative controls “turn off” transcription
which is the most abundant and diverse tissue in the body
connective tissue -
Commonly functions as a scaffold on which
other cells cluster to form organs
 Forms an elaborate extracellular matrix,
important for the maintenance of cell
differentiation
 Important in support/repair of nearly every
tissue and organ
DNA/RNA replication is
anti-parallel and complimentary
embryonic development is associated with selective gene expression that
controls what four essential processes to enable a single
cell to develop into a complex organism?
Cell proliferation
 Cell specialization
 Cell‐to‐cell interactions
 Cell movement and migration
simple squamous epithelium cells are found where?
Lining of blood vessels, pulmonary alveoli,
Bowman capsule
simple cuboidal epithelium cells are found where?
Thyroid, sweat, and salivary glands; kidney tubules
simple columnar epithelium cells are found where?
Lining of intestine, glandular ducts
pseudostratified epithelium cells are found where?
Male urethra, respiratory passages
stratified squamous epithelium cells are found where?
skin, mucous membranes
stratified columnar epithelium cells are found where?
epiglottis, anus, parts of pharynx
stratified transitional epithelium cells are found where?
bladder
loose connective tissue is found where?
Widespread locations, dermis of skin, adipose,
tissue organs
dense/supportive connective tissue is found where?
Cartilage, bone, tendons, joints, fascia surrounding
muscles
hematopoietic connective tissue is found where?
Bone marrow, lymph tissue, plasma`
myoepithelial cells are found where?
******Mammary, sweat, and salivary glands
neuroglia issue is found where?
Primarily central nervous system
describe meisosis
First division
– Two cells each with 46 chromosomes
– 2 or 3 crossover events occur
Second division
– Sister chromatids pulled apart
– Result is 4 haploid cells with 23 chromosomes
what are the two types of DNA mutation
point mutation
frameshift mutation
describe a point mutation
Single base pair (AA) substitution
• May cause affected codon (three base pairs) to
signify an abnormal amino acid
describe a frameshift mutation
Often dramatically changes genetic code
• Addition/removal of one or more bases changes
reading frame
• All remaining codon triplets have greatly altered
amino acid sequences
single-gene (Mendalian) disorders are due to:
Due to DNA mutation that codes for a particular
protein
what is the #1 reason for chromosomal abnormalities
Aneuploidy: abnormal number of chromosomes,
either > or < 46 in humans
 Nondisjunction: failure of pairs to separate properly
during 1st or 2nd meiotic division
 Resulting germ cells have 22 and 24 chromosomes,
respectively
 Combination of abnormal and normal germ cells produce
fertilized cell with 25 or 27 chromosomes
what is anaphase lag
1 chromosome left out of newly
formed cell nucleus
what is monosomy
Monosomy: daughter cell with a deficiency of
1 chromosome
 Not compatible with life
what is polysomy
daughter cell with too many
chromosomes
 May result in viable fetus
 Nearly always associated with severe disability
 Those involving extra/missing sex chromosome not
as debilitating
what are the possible causes of chromosomal abnormalities?
what causes increase risk?
Poorly understood
 Radiation, viruses, chemicals implicated
 Increased risk
 Advanced maternal age
 Abnormalities in parental genetic structure
what is usually the cause of abnormal chromosome structure?
Usually due to breakage and loss or rearrangement
of chromosome pieces during meiosis or mitosis
 Meiosis
 Crossing over errors: chromosome portions lost,
attached upside‐down, or attached to wrong
chromosome
 Mitosis
 Opportunities for chromosomal breakage and
impairment
what is translocation
Exchange of DNA pieces between nonhomologous
chromosomes –incorrect separation
what is reciprocal translocation?
no lost genetic material,
sometimes no symptoms; increased risk of
producing abnormal gametes
what is a Robersonian translocation?
exchange of 1 long chromatid arm for 1 short arm
what are isochrosomes?
2 identical short chromatid arms
and 2 long chromatid arms remain together
 due to incorrect separation at centromere
what is an inversion?
Removal and upside‐down reinsertion of a
chromosome
 No net loss/gain of genetic material
 Often no significance to individual, but may affect
offspring
 Inverted chromosome may not pair up properly during
meiosis, resulting in duplication or loss of genes
what is deletion? and its cause
Loss of chromosomal material
 Caused by break in arm of a single chromosome
 Results in DNA fragment with no centromere
 Piece lost in next cell division
 Associated with some forms of cancer
 Deletions at both ends may form a ring
chromosome
what is duplication?
Results in extra copies of a portion of DNA
 Less severe consequences
what does "autosomal chromosomal disorder" mean?
means not related to the sex chromosomes, but
one of the other 22 homologous pairs
what is the most common chromosomal disorder? Describe it?
Trisomy 21 (Down Syndrome)
Extra 21st chromosome
 Leading cause of mental retardation
 Protruding tongue, low‐set ears, epicanthal folds,
poor muscle tone, short stature; congenital heart
deformities, increased susceptibility to respiratory
infections, leukemia
 Majority of fetuses either stillborn or aborted
why do fathers pass X or Sex Chromosome disorders on only to their daughters?
because the disorders are linked to the X chromosome, not the Y - since boys receive only Y's from father, they don't inherit the X-linked disorder from dad
why do males always inherit an X linked disorder from their mothers?
because the mother can only pass on an x chromosome to their sons, and if there is no additional x chromosome to override the defective x chromosome (which there isn't in boys) the disorder will express
nearly all x-linked disorders are recessive so females only express the disorder when:
both x chromosomes have the mutation
hemophilia A is an example of
a sex linked disorder
describe Klinefelter syndrome
Common genetic disease
of the sex chromosomes
 Usually 1 extra X
chromosome; sometimes
more than 1
 Lack of secondary sex
characteristics during
puberty
Lack of testosterone
 Testicular atrophy; infertility
 Feminine hair distribution/gynecomastia
 Tall stature, long arms/legs
 High‐pitched voice
 Impaired intelligence
.
.
Describe Turner Syndrome
Monosomy X: 1 normal X
chromosome; no Y chromosome
 Female phenotype with no ovaries
S dX h i i
Sex Chromosome Disorders
 Second X chromosome missing or
structurally abnormal; usually due to
father’s advanced age
 Rarely survive to birth
 Short stature, webbed neck, fibrous
ovaries, sterility, amenorrhea, wide
chest, congenital heart defects
.
.
what do multiple x females exhibit?
Menstrual abnormalities; retardation tendency
with more than 4 X chromosomes
double y males are generally
taller than average
Mendelian Single -gene disorders result from
alterations or mutations of single genes
Affected genes may code for abnormal enzymes,
structural or regulatory proteins
 Transmission follows principles of Medelian
Genetics
 Random transmission, but chances of transmission
can be calculated using the Punnett Square
autosomal dominant disorders have a _____% chance of passing on with one affected and one unaffected mate
50%
Unaffected individual to do not ever transmit autosomal dominant disorders. Why?
b/c if you don't have the disorder, you are not a carrier
describe Marfan Syndrome
It is what type?
autosomal dominant disorder
Connective tissue disorder
 Typically tall, slender; long, thin
arms/legs; long, thin fingers
(arachnodactyly)
 Cardiovascular lesions most
threatening
 Aorta tends to be weak, susceptible to
dilation/rupture
 Traced to mutations in fibrillin 1 gene
on chromosome 15
describe Huntington Disease
what type?
autosomal dominant
Primarily affects neurologic function
 Symptoms appear after age 40
 Mental deterioration, involuntary arm/leg
movements
 Parent may transmit to offspring before becoming
aware of defective gene
describe autosomal recessive disorders
Mutation of autosomal recessive gene
 Males/females equally affected
 Usually not apparent in affected individuals; both
parents carriers of mutant recessive gene
unaffected individual may transmit an autosomal recessive disorder to offspring. What are the chances
Two carriers have 1:4 chance of having affected
offspring and 2:4 chance of having carrier offspring
what are some common autosomal recessive disorders
Albinism
 Phenylketonuria
 Inborn error of metabolism
 Cystic fibrosis
 Most common
Describe nonmendelian Single-gene disorders
Caused by long triplet repeat mutations, such as
fragile X syndrome
 Due to mitochondrial DNA mutations
 Associated with genomic imprinting
 Do NOT follow Mendelian Principles
 Random inheritance, etc
describe multifactorial (polygenic) disorders
“Run in families”
 Thought to be produced by an interaction of many
genes
 2 or more mutant genes act together
 Also present range of severity
 Difficult to predict based on family history
 Very common (high blood pressure, cancer,
diabetes)
 Height, weight, intelligence
8
what is the period of most vulnerability of feus to teratogen exposure
3rd to 9th week: embryo very susceptible to
teratogenesis (especially 4th and 5th weeks during
organ development
after the 3rd month, teratogens affect what?
growth or injury
to already‐formed organs
cancer cells proliferate despite lack of what?
growth‐initiating signals from the
environment (paracrine, endocrine, autocrine)
cancer cells escape _______________signals
apoptic
cancer cells are considered "immortal" b/c they are capable of
unlimited replication
why are cancer cells considered genetically unstable
b/c they evolve by accumulating new mutations at a much faster rate than normal cells.
how do cancer cells operate?
invade and overtake their local tissue
•migrate from their site of origin to colonize distant sites where they
do not belong
malignant tumors do not closely resemble the tissue type of origin, the greater the degree of anaplasia indicates what?
the more aggressive the malignancy
how is a benign tumor different from a malignant tumor?
Does not have potential to kill host, but may be life threatening because of its location
 Does not invade adjacent tissue or spread to distant sites
 Many are encapsulated
 More closely resembles original tissue type
 Grows more slowly, little vascularity, rarely necrotic, often retains original function
“‐oma” suffix indicates
benign tumor
“‐carcinoma,” indicates
malignant tumor of epithelial origin
(adenocarcinoma)
"sarcoma" indicates
malignant tumor of mesenchymal origin
 Mesenchymal~ nerve, bone, muscle
Statistics:
Cancer is ____ leading cause of death in the U.S.
 Most cancer deaths occur in individuals over age ______
 Men have ____ risk of developing cancer; women have _____
risk
 5‐year survival rate: ____
 what proportion of cancer‐related deaths may be attributable to
lifestyle factors
2nd
55
1:2
1:3
66%
One third
what are some examples of lifestyle factors
 Tobacco use, Nutrition, Obesity, Sun exposure (skin cancer),
Sexual exposure to HPV (cervical cancer)
which type of cancer is the leading cause of death in men and women and has the worst survival rate?
lung cancer
what are the two types of carcinogens in tobacco
Initiator (causes genetic damage)
 Promoter (promotes tumor growth)
malignant tumors do not closely resemble the tissue type of origin, the greater the degree of anaplasia indicates what?
the more aggressive the malignancy
how is a benign tumor different from a malignant tumor?
Does not have potential to kill host, but may be life threatening because of its location
 Does not invade adjacent tissue or spread to distant sites
 Many are encapsulated
 More closely resembles original tissue type
 Grows more slowly, little vascularity, rarely necrotic, often retains original function
what is a proto-oncogene?
a normal cellular gene that is growth promoting and usually inhibited in nonproliferating cells. When erroneously activated, it becomes an onco gene and promotes cancer
what is an onco-gene?
a gene associated with the initiation of cancerous behavior in cells
what is contained in the proto-oncogene?
the code for cell growth-activating pathways
what are the two major categories of critical cancer genes involved in the regulation of cellular growth and whose dysfunction is a factor in most forms of cancer
proto-ocogenes
tumor suppressor genes
(generally, both overactivity of proto-genes leading to oncogenes and failure of tumor-suppressor genes is necessary to achieve mallignant transformation)
what do tumor suppressor genes do?
inhibit cell proliferation = cancers may arise when tumor suppressor genes function is lost
What is 'HEMOPHILIA' defined as?

What 2 types are there?
Type A- LACK OF FACTOR VIII (8) (ANTIHEMOPHILIC GLOBULIN)

Type B- LACK OF FACTOR IX (9) (CHRISTMAS FACTOR)
How do proto-oncogenes code for growth factors
excessive autocrine or selfstimulated
growth. Small cell−manufactured peptides
secrete into extracellular space, diffuse to nearby cells, interact with receptors on target cell surface
How do proto-oncogenes code for growth factor receptors
cause expression of receptors that shouldn't be there at all, increase the number of growth factor receptors, or increase the growth receptors' affinity
How do proto-oncogenes code for cytoplasmic signaling pathways
excessive abnormal
intracellular growth‐signaling/Mutant proto‐oncogene
canactivate without external stimulation
 Involve numerous enzymes and chemicals that normally function to transmit signals from activated receptors at
cell surface to cell nucleus
How do proto-oncogenes code for nuclear transcription factors:
mutations cause overproduction of transcription factors (which initiate S phase in DNA replication) or interfere with the normal mechanisms for keeping them in check.
proto-oncogenes become activated oncogenes when
mutations alter their activity so that proliferation-promoting signals are generated inappropriately
what are the four ways that proto-oncogenes are activated
1. oncogenes may be introduced into the host cell by a retrovirus
2. a proto-oncogene w/i the cell may suffer a mutagenic event that changes its structure and fxn
3. a DNA swquence that normally regulates proto-oncogene expression may be damages or lost and allow the proto-oncogene to become abnormally active.
4.an error in chromosome replication may cause extra copies of the proto-oncogene to be included in the genome (amplification)
what are the three cancers thought to be associated with retroviruses
HIV
 Kaposi’s sarcoma
 Epstein‐Barr virus
 Burkitt lymphoma
 Human T‐lymphocyte virus type 1
 Adult T‐cell leukemia/lymphoma
How do tumor suppressor genes contribute to cancer?
Contribute to cancer only when not present
 Induce apoptosis, inhibit cell proliferation
 When not functioning or not present, inappropriate cell proliferation prevails
describe the genetic component of tumor suppressor gene's role in cancer
Both copies of tumor suppressor genes are inactivated
when cancer develops
 Both copies, meaning one copy of the gene from each
parent
 One can inherit a defective copy of tumor suppressor
gene from 1 or both parents
Typically its at the level of _____________ that mutations begin with oncogenes
transcription
what is the Rb Gene?
a tumor suppressor gene
Named for its role in retinoblastoma
 Codes for large protein in cell nucleus (pRb) that is the
“master break” for the cell cycle
 Blocks cell division
 Binding transcription factors
 Inhibits T factors from transcribing genes that initiate cell cycle
an inactivating mutation of the RB gene removes ______________ on cell division
1 major restraint
what is the most common tumor suppressor gene defect identified in cancer cells?
lack of a functional P52 tene
More than ____ of all types of human tumors lack functional
p53
1/2
what does the P53 gene do when functioning normally?
Gene inhibits cell cycling/mediates apoptosis
 Accumulates only after cellular (DNA) damage
 Binds to damaged DNA and stalls division
 Stalled division allows DNA to repair (Allows genetically damaged/unstable cells to survive and continue to replicate
 If DNA does not repair then it triggers apoptosis
 Very little p53 in normal cells
The P53 gene may direct cell to initiated __________
apoptosis
describe P53's role in chemotherapy/radiation
Chemotherapy/radiation
 Do not directly kill cells
 Instead, they create enough damage to the target cell to
trigger p53‐mediated cell death
 Cancer cells that lack functional p53 may be resistant to
chemotherapy/radiation
what are the three steps of carninogenesis?
Initiation
 Promotion
 Progression
define metastasis
Process by which cancer cells escape their tissue of origin
and initiate new colonies of cancer in distant sites
 Specialized enzymes and receptors enable them to escape their tissue of origin and metastasize
describe the 4 steps of metastasis
1. Tumor cells cut cell-to-cell attachments; allows
Movement toward basement membrane
2. Enzymes are released to break down membrane
3. Tumor cell begins to migrate via use of laminin/
Fibronectin receptors
4. Cell pulls through; autocrine signaling enhances
mobility
cancer cells generally spread via what?
circulatory and lymphatic systems
what are ways to identify parent tissu of cancer origin?
Tumor markers help identify parent tissue of cancer origin
 Rely on some retention of parent tumor characteristics
 Some released into circulation
 Others identified through biopsy
 Enzymes typically used as tumor markers
 Help track tumor activity
describe angiogenesis
 Process by which cancer tumor forms new blood vessels
in order to grow
 Usually does not develop until late stages of development
 Triggers are not generally understood
 Inhibition of angiogenesis is important therapeutic goal
what are the warning signs of cancer in adults
Change in bowel or bladder habits
 A sore that does not heal
 Unusual bleeding or discharge
 Thickening or lump in breast or elsewhere
 Indigestion or difficulty swallowing
 Obvious change in wart or mole
 Nagging cough or hoarseness
.
.
what are the warning signs of cancer in children
Continued, unexplained weight loss
 Headaches with vomiting in the morning
 Increased swelling or persistent pain in bones or joints
 Lump or mass in abdomen, neck, or elsewhere
 Development of whitish appearance in pupil of the eye
 Recurrent fevers not caused by infections
 Excessive bleeding or bruising
 Noticeable paleness or prolonged tiredness
.
.
describe cachexia
Overall weight loss and generalized weakness
 Loss of appetite (anorexia): secondary to toxins released by
malignant cells
 Increased metabolic rate: TNF‐alpha, high levels of cytokines
 Nausea/vomiting
how do cancers affect the immune system?
cause deficits
Immune system suppressed by cancer cell secretions
 Some cancers can elude immune system detection
what happens when cancer suppresses the immune system through bone marrow suppression?
anemia, leukopenia, and
thrombocytopenia
 Due to invasion and destruction of bone marrow cells, poor nutrition, and chemotherapy
what is anemia
deficiency in circulating red blood cells
what is leukopenia?
what is it primarily caused by?
Deficiency in circulating white blood cells
 Primary cause
 Malignant invasion of bone marrow
 Contributing factors
 Malnutrition
 Chemotherapy
what is thrombocytopenia?
why is it life-threatening?
Deficiency in circulating platelets
 Important mediators in blood clotting
 Predispose to life‐threatening hemorrhage
by what can anemia, leukopenia, and thrombocytopenia all be managed?
blood replacement therapy
what are some examples of paraneoplastic syndromes
Hypercalcemia: over‐secretion of PTH‐related protein by cancer cells
 Cushing syndrome secondary to ACTH secretion
 Hyponatremia and water overload secondary to excess ADH secretion
what is the primary source of cancer pain and anorexia and can cause a portal for infection?
Mucositis caused by chemotherapy and radiation therapy