Shared Flashcard Set

Details

Midwestern Summer Biochemistry
genetics/blood for Nathan
280
Biochemistry
Graduate
07/31/2012

Additional Biochemistry Flashcards

 


 

Cards

Term
Hemophilia B
Definition

Gene: Factor IX

Type of mutation: point mutation

 

Gene is located on the x-chromosome

Term
Tay-Sachs disease
Definition

Gene: HexA

Type of Mutation: Point mutation in the splicing site

 

Lysosomal storage disorder

Term
Sickle-cell anemia
Definition

Gene: (beta) B-globing

Type: Missense Point mutation in coding sequence

 

High frequency in African-Americans

Term

Neurofibromatosis Type 1

 

 

Definition

Gene: NF1

Type of mutation: nonsense point mutation in coding sequence

 

Highly variable: cafe-au-lait spots and neurofibromas

Term
B-thalassemia
Definition

Gene: B-globin

Type: Different types of point mutations

 

Decreased production of B-globin

Term

Tay-Sachs

(most common variant)

Definition

gene: HexA (same as normal)

Type: Small insertion of 4 base pairs in coding sequence

 

GM2 build up in neuronal lysosomes

Term
Cystic Fibrosis (most common variant)
Definition
Gene: CFTR
Type: small deletion of 3bp in coding sequence

Over 1000 mutations reported
Term
(alpha) a-thalassemia
Definition

Gene: a-globin

Type: Large deletion from aberrant recombination

 

Decreased production of a-globin

Term
Charcot-Marie-Tooth
Definition

Gene: PMP22

Type: large insertion from aberrant recombination

 

Most Common inherited neurological disorder

Term

Defination of and

examples of New (de novo) Mutations?

 

 

Definition

Dominant acting mutation in a child with no previous family history

 

Achondroplasia

NF Type 1

Marfan Syndrom

 

Term
Defintion of and examples of Germline mosaicism?
Definition

Germline consists of more than on distinct population of cells

 

Osteogenesis Imperfecti

Term

Delayed age of onset diseases?

 

Definition

Usually do not manifest until at least 30 years old (usually after they have children)

 

Huntington disease

Hereditary hemachromatosis

Term

Defintion of Reduced penetrance?

Examples?

Definition

Individuals with the mutated genotype do not manifest symptoms at all

 

Retinoblastima

Fragile X syndrome (in some women)

Term
Variable expression defintion and examples of diseases?
Definition

Degree of severity is different

 

NF type 1

 

 

Term
What is the difference between Reduced penetrance and variable expression?
Definition

Those with reduced penetrance will display no symptoms.

 

Those with variable expression with always show symptoms, just in different severity.

Term

What is pleiotropy?

Examples of diseases

Definition

Gene mutation exerting effect on several tissues/organs instead of just one

 

Cystic Fibrosis

Diabetes

Marfan Syndrom

von Gierke disease

Term

What is heterogeneity and examples?

 

Definition

Disease resulting from mutations in different genes

 

Osteogenesis Imperfecti

Term
What is genomic imprinting and examples?
Definition

Transcriptional inactivation of genes. Results in different syndroms depending on sex of the parent

 

Angelman syndrome (from mother)

Prader-Willi (from father)

Term
What is the concept of anticipation and examples of diseases?
Definition

Earlier onset or more severe expression of the disease in the more recent generations.

 

Myotonic dystrophy

Fragile X syndrome

Huntington disease

 

Often due to DNA polymerase slippage ("here it comes" video)

Term
What is consanguinity and what are the diseases?
Definition

Mating between related individuals

 

Leads to rare autosomal recessive diseases

 

It is estimated that each person carries 5 recessive genes in heterozygous form that would present in a lethal phenotype if present in a homozygous state. This is termed our genetic load.

Term

Characteristics of Autosomal dominant inheritance?

 

Examples

Definition

Both sexes are equally affected

Vertical Transmission

Father-son transmission

Affected individuals

[image]

Examples: Huntington disease, achondroplasia, NF type 1, Marfan syndrome, Familial Hypercholesterolemia

Term
Characteristics of Autosomal recessive inheritance and examples?
Definition

Both sexes equally affected

Usually no prior family history

Consanguinity

Mating between two carriers transmits trait to ~25% of offspring.

[image]

examples: hurler syndrome, hereditary hemochromatosis, cystic fibrosis, sickle-cell anemia, PKU, B-thalassemia, tay-sachs

Term
What genes are silenced in Angelman syndrome and Prader-Willi?
Definition

Angelman syndrome: UBE3A

Prader-Willi: SNRPN among others

Term

Summary table for patterns of inheritance...

 

Understand where diseases fall under these categories:

 

Vertical transmission yes/no

Father-son- transmission yes/no

 

in some cases:

is there father-daughter transmission/ are females mostly affected?

Are there multiple cases/males mostly affected?

Definition
Term

What are the mitochondrial diseases?

 

Which parent is the mtDNA inherited from?

Definition

CPEO

LHON

MELAS

MERRF

Kearns-sayre syndrome

Leigh syndrome

Pearson syndrome


----------------------

The mother, only females can transmit the disease to their offspring, and will do so 100% of the time.

 

Term

Characteristics of X-Linked dominant diseases and examples?

 

Definition

Vertical transmission

No father --> son

Females 2x as likely to be affected than males

Affected males cannot transmit disease to sons, but will 100% of the time to daughters.

ex: Fragile X syndrome

[image]

Term

X-Linked recessive characteristics and examples?

Definition

Diseases can skip generations by carrier females

No father to son (NO x-linked diseases will be)

Males are much more frequently affected (females have a 2nd x chromosome to use instead)

ex: Hemophilia A/B

G6PD deficiency

OTC deficiency

Red-green color blind

Lesch-Nyan

Duchenne muscular dystrophy

Becker Muscular dystrophy

[image]

Term

What would a mitochondrial pedigree look like/why?

 

Definition

[image]

Males/females can be affected, but only females can transmit.

0% of offspring of men will manifest disease

100% of offspring of women will have disease

Term
Characteristics of multifactorial inheritance and examples
Definition

Additive polygenic model for quatitive traits

Threshold model for qualitative traits

Term

Huntington Disease

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Dominant

 

HD (huntington) Gene

 

Mental Retardation, choreic movements

Term

Achondroplasia

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal dominant

 

FGFR-3

 

Dwarfism

Term

NF Type 1

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal dominant

 

NF1

 

Cafe-au-lait spots, nurofibromas

Term

Marfan Syndrome

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Dominant

 

FBN1(fibrillin)

 

tall stature, long/slender fingers

Term

Familial Hypercholesterolemia

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal dominant

 

LDL Receptor

 

Elevated serum cholesterol (death by MI by age 20)

Term

Hurler Syndrome

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Recessive

 

a-L-iduronidase

 

Lysosomal storage disorder

Term

Hereditary Hemochromatosis

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Recessive

 

HFE

 

Iron Overload (increases absorption too much)

Term

Cystic Fibrosis

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal recessive

 

CFTR

 

>1000 mutations reported

Term

Sickle-Cell anemia

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Recessive

 

B-globin

Term

Phenylketonuria (PKU)

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal recessive

 

Phenyalanine Hydroxylase (PAH)

leads to a build up of substrate, need to change diet within a month of birth

 

 

Term

B-thalassemia

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal recessive

 

B-globin

 

hemoglobinopathy

Term

Tay-sachs

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

Autosomal Recessive

 

HexA

 

Lysosomal storage disorder

Term

Fragile X Syndrome

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-Linked Dominant (only one)

 

FMR1

 

Mental retardation

Term

Hemophilia A

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-linked recessive

 

Factor VIII

 

 

Term

Hemophilia B

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-linked recessive

 

Factor IX

 

 

Term

Lesch-Nyhan Syndrome

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-linked recessive

 

HGPRT

 

Self-mutilation

Term

Duchenne Muscular Dystrophy

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-linked recessive

 

Dystrophin

 

Weakness and loss of muscles (Severe)

Term

Becker musclular dystrophy

Type of inheritance?

Gene affected?

Hallmark (characteristic) of disease?

Definition

X-linked recessive

 

dystrophin

 

weakness and loss of muscles (mild)

Term

Red-green color blindness

Type of inheritance?

 

Definition
x-linked recessive
Term

G6PD deficiency

Type of inheritance?

 

Definition
X-linked recessive
Term

OTC deficiency

Type of inheritance?

Definition
X-linked recessive
Term

All initial diseases named ONLY by initial

CPEO
LHON
MELAS
MERRF

 

Type of inheritence?

Definition

Mitochondrial

 

also

Kearns-Sayre syndrome
Leigh syndrome
Pearson syndrome

Term

Coronary artery disease
Diabetes
Obesity
Alcoholism
Infantile autism
Cancer
Schizophrenia
Cleft lip/palate
Pyloric stenosis
Club foot
Spina bifida

EXAMPLES OF?

Definition
Multifactorial inheritance
Term
What are the 2 invasive techniques for prenatal diagnosis?
Definition

Amniocentesis

Chorionic Villus sampling (can do sooner, but more dangerous)

Term
What are the 2 techniques for prenatal dx that are Non-invasive?
Definition

Nuchal translucency and maternal serum screening

Ultrasonography

Term

Amniocentesis

Optimal Time?

Samples collected?

Tests what?

Conditions reflected?

Invasive?

Definition
  • 15-17 weeks
  • amniotic fluid and amniocytes
  • alpha-fetoprotein (AFP) /DNA based testing/cytogenetic analysis
  • Chromosomal abnormalities/genotype of selected genes/Trisomies (low AFP) or Neural tube disorder (High AFP)
  • Yes (.5% fetal loss chance)
Term

Chorionic Villus sampling (CVS)

Optimal Time?

Samples collected?

Tests what?

Conditions reflected?

Invasive?

Definition
  • 10-11 weeks
  • taking fetal trophoblastic tissue
  • Cytogenetic analysis/dna based testing
  • chromosomal abnormalities
  • genotype status of selected genes
  • Yes(1-1.5% fetal loss, risk of limb reduction)
Term

Nuchal translucency and maternal serum screening

Optimal Time?

Samples collected?

Tests what?

Conditions reflected?

Invasive?

Definition
  • 15-17 weeks
  • maternal serum (taking mother's blood) and ultrasounds
  • Nueral tube ( NT) / maternal AFP
  • trisomies (low AFP)/ NTD (high AFP)/ chromosomal anomalies (NT)
  • no risk
Term

Ultrasonography

Optimal Time?

Samples collected?

Tests what?

Conditions reflected?

Invasive?

Definition
  • 16-18 weeks
  • visualization
  • -
  • Congenital malformation/NTD
  • no risk
Term

Erythrocyte life span?

 

produced where?

destroyed by?

 

Definition

120 days

 

bone marrow

resident macrophages (spleen, liver, bone marrow)

Term

Development from stem cell to erythrocyte?

1 BFU-E can divide into __ RBCs?

divisions per blast stage?

Definition

Stem cell --> BFU-E -->series of blasts --> reticulocyte -->erythrocyte

 

1000s

3-5 rounds

Term

Lifespan of reticulocyte?

where does it spend its time?

 

Definition

72 hours

 

48hrs in bone marrow, 24 hours in circulation

 

in cases of anemia, can be released early, 24/48

Term

CFU-E/normoblast, Reticulocyte, Erythrocite (mature RBC)

 

present in bone marrow or blood?

 

Definition

CFU-E in bone marrow

 

Reticulocyte in bone marrow for 48, blood for 24

 

Erythrocite in blood.

 

So Reticulocytes are useful messengers of what is going on in bone marrow

Term
What is the process for erythropoeisis?
Definition

1: Kidneys detect lower O2 in blood

2: Kidneys secrete EPO into blood in response

3: EPO stimulates erythropeiesis (by ligand binding)

4: Increased O2 carrying capacity of blood

5: Relieves initial stimulus that triggered EPO secretion

Term

How does EPO exert its affects?

 

Definition

Increases division

Inhibits cell death of blast cells, so make more

increased Hb synthesis

Increased Fe absorption

Increased erythroid differentation

stimulated production and early release of reticulocytes

Term
You need a healthy kidney to produce EPO, what can be done if patient has chronic kidney failure?
Definition

Recombinant EPO can be injected regularly to prevent severe anemia.

 

Term
Nutrients needed for cell division?
Definition

Folate (folic acid)

 

Vit B12

Term
Nutrients required for Hemoglobin synthesis?
Definition

Dietary protein and Iron

 

 

Term
How do you define anemia?
Definition

abnormally low Hb concentration in whole blood samples

 

anemia is a SIGN of a disease, not a disease itself

Term
2 causes of anemia?
Definition
decresed erythropoiesis and premature blood loss
Term
RBC normal value? and value used for calculations
Definition

5,000,000/microliter

 

5 is used in math

Term

HGB?

what is it and avg?

Definition

hemoglobin concentration in whole blood

 

15g/dl

Term

HCT

what is is and avg?

Definition

volume % of whole blood occupied by rbcs

 

45%

 

45 used in calculation not .45

Term
What is the rule of 3?
Definition

RBC *3=HGB

HGB*3=HCT

 

 

in a normal person..

Term
How do you calculate MCV and what is it?
Definition

average volume or circulating RBS

80 and under microcytic

100 and up macrocytic

 

MCV= HCT/RBC  *10

Term
What is MCH and how calculate it?
Definition

amount of Hb in avg circulating erythrocyte

 

MCH= HGB/RBC   *10

Term

What is MCHC and how to calculate it?

 

what does it identify?

Definition

concentration of HGB

 

MCHC= HGB/HCT   *100

 

normochromic/hypochromic rbcs

Term
Elevated CR=?
Definition

Anemia due to blood loss/premature destruction

 

body is ABLE to produce RBCs in response to EPO

Term
CR normal or decreased?
Definition

Body unable to produce RBCs in spite of EPO increase

 

caused by decreased erythropoiesis

Term

If HGB low, means what?

and then

if CR is decreased?

and then the MCV is low?

Definition

Anemia present

due to decreased erythropoiesis

 

if MCV is low, there are microcytic cells due to impaired HGB Synthesis

Term

HGB low

and

CR low

and MCV high?

Definition

anemia present

decreased erythropoiesis

 

and the elevated MCV =macrocytic cells,

so due to limited DNA synthesis since cells aren't dividing

Term
What can cause microcytic RBC anemia?
Definition

Decreased proteing synthesis.

 

most commonly caused by chronic iron deficiency.

inadequate protein in diet can contribute

Term
an elevated MCV has what pathological cause of anemia?
Definition

macrocytic cells mean that the nucleotide supply is deficient.

the blast cell division time increases so the cells are larger

Most commonly due to B12 or folate deficiency

Term

How much iron does the human body keep in storage?

in use?

 

How does the body get rid of Fe?

Definition

4 grams total, 1 g in storage, 3 in use

 

Body cannot promote iron loss, can only lose by bleeding. So body absorbs however much it loses by skin shedding (1-2 mg/day) to maintain homeostasis

Term
Bioavailabity of iron from most available to least?
Definition

Heme>>Ferrous Fe 2+ > Ferric Fe 3+ > Elemental iron Fe 0

 

Heme is mainly found in meat fish and seafood

5-35% of iron absorbed

Term
Meal composition that enhances or diminishes iron availability affects which forms of iron?
Definition
all non heme iron
Term
What is Vitamin C's (a reducing molecules) affect on iron absorption?
Definition

Enhances it

 

Vitamin C reduces Fe 3 ferric to Fe2 ferrous. it can form complexes with ferrous iron. and iron complexes increase the bioavailability of ferrous iron

Term
What is amino acids affect on iron absorption?
Definition
enhances
Term
What is the affect of acidic foods on iron absoption?
Definition

increases absorption by increasing gastric acid production increasing bioavailability

 

Term
Affect of Phytates on iron absorption?
Definition

Form insoluble chelates decreasing mineral absorption.

 

high in cereal grains

 

decrease absorption

Term
affect of polyphenols on iron absorption?
Definition

form insoluble chelates with minerals decreasing absorption

 

in Tea and Coffee

Term
Affect of phosphates and phosphoproteins on iron absorption?
Definition

form insoluble chelates with Fe 2 and fe 3 iron

 

ex egg yolk

Term
affect of oxalate on iron absorption?
Definition

decreases it

chelate non heme iron

 

ex spinache. has a lot of iron, but only 5% is bioavailable because of high oxalate concentraion in spinache

Term
Affect of calcium or zinc on Fe absorption?
Definition
supplements can diminish iron absorption
Term
are vegetarians at an increased risk of becoming iron deficient?
Definition
No, absence of Heme iron is balance by increase in enhancer Vit C
Term
Where in the body is Iron absorbed?
Definition

Enterocytes of the duodenum and upper jejunem

 

surgical removal of any part of these will decrease iron absorption

Term
What form of iron is toxic and why
Definition

Ferrous Fe2 when it is unbound

 

it catalyzes a highly destructiv hydroxyl radical OH in the fenton RXN

 

Fe2+H2O2 --> Fe3 +OH +OH

Term

What binds to ferric iron?

What is this things name when bound to 0, 1, 2 irons?

Definition

Transferrin, can bind up to two Fe 3 atoms.

 

apotransferrin: not bound

monoferric transferrin: bound to 1

Diferric transferrin: bound to 2

Term
How does iron cross into cells?
Definition

Can't cross lipid bilayers directly, requires machinery for transport

 

All growing cells possess TfR, transferrin receptors, a plasma membrane protein that mediates iron uptake

Term

What is the protein used to store iron?

 

What is protein used to store iron during iron overload?

Definition

ferritin.

is a 24 subunit protein, never H24L0 or H0L24 unless in pathological instances

 

Hemosiderin, another form of ferritin. contains 30% more stored iron than ferritin

Term
Affect of fenton rxn on Fe in ferritin?
Definition
Iron stored in ferritin cannot be catalyzed by Fenton Reaction
Term
How much iron is in one ml of blood?
Definition

.5 mg of iron / 1 ml of blood

 

so donating 500 ml of blood is 250 mg of iron

Term

What is the laboratory value TIBC?

 

What is best test of iron stores?

Definition

Serum transferrin.

 

 

Serum ferritin is the best test of iron stores.

ferritin levels are found with ferritin too

 

Term
What does high ferritin, low Transferrin mean?
Definition
iron is high. ferritin is high to store iron. transferrin levels are low because cells have enough iron and don't need any transported to it
Term
what does low ferritin high transferritin mean?
Definition

low levels of iron

 

Ferritin is low because stores are depleted, and transferrin receptors are high because cells demand more iron

Term
What is nutrition adaptation regarding Fe?
Definition
individuals with an iron deficiency will absorb more iron than someone with adequate iron
Term

What is the most common coause of iron deficiency in adults?

 

toddlers?

Adolescent girls and premenopausal women?

pregnant women?

 

Definition

Acute or chronic blood loss (ie ulcers, tumors)

 

nutritional insufficiency

menstruation

increased requirement (3-4 mg of iron are transferred to fetus everyday)

 

Term
What are symptoms of iron deficiency?
Definition

early stages are asymptomatic

 

later stages include:

Koilonychias (concave nails)

Anisocytosis (inequality of RBC size)

Poikilocytosis (abnormal RBC shape)

Term
Are iron supplements recommended in order to prevent iron deficiency anemia?
Definition
No, iron-deficiency anemia is rarely to to diet
Term
What are the 3 enhancers of iron uptake
Definition
Meat and fish, VIT C, acidic foods
Term
Typically, macrocytic anemias are hypo or normo chromic?
Definition

Normochromic

 

Usually microcytic anemias involve hypochromic cells

Term
What is the difference between megaloblatic anermias and nonmegaloblastic
Definition

both are macrocytic, but nonmegaloblatic are usually only 100-110MCV. dna synthesis is fine, etiology is usually alcoholoism, liver disease, medications.

 

Megaloblastic are 110-130 MCV usually, but up to 160. DNA synthesis is impaired, can have oval marcrocytes and hypersegmented neutrophils

Term
How does impaired Dna synthesis affect cells?
Definition
Increases them, longer dividing time
Term
What is the cause of anemia in megaloblastic anemias?
Definition
decreased DNA synthesis leads to premature destruction of megaloblasts within bone marrow
Term

Metabolism of Folate:

Folic acid gets to Tetrahydrofolate (THF)

 

Definition
2 NADPH dependent reductions, via the enzyme dihydrofolate reductase.
Term
Folate plays a role in what 2 processes of the cell?
Definition

nucleotide synthesis

 

Methylation rxns. folate acts as a carrier for methyl groups

Term
most common cause of folate deficiency?
Definition
poor diet, body only stores enough for a few months
Term
what is methotrexate used for?
Definition

is a structural analog of folic acid. inhibits dihydroflate reductase thus reducing the ability of the cell to recycle folate.

 

Used for killing cancer

Term
B12 is an enzyme for what 2 enzymes? which are used for what?
Definition

homocysteine methyltransferase

involved in folate recycling

 

Methylmalonyl-coa-mutase

not used in folate recycling at all, good for identifying B12 or folate deficiency

Term
99% of all methyl transfers in the body use what as a methyl donor?
Definition

SAMe

s-adenosylmethionine

 

Term
B12 deficiency compromises what two pathways?
Definition

Recycling of Folate

 

Methylation of cellular proteins and nucleic acids

 

 

Homocysteine is the enzyme that gets both of those going, and B12 is the coenzyme

Term

If the folate recycling is compromised, what is the pathology?

 

If the methylation is compromised, what is the pathology?

Definition

Megaloblastic anemia

 

Peripheral neuropathy

 

Term
Why is it important to differentiate between folate and b12 deficiency?
Definition
B12, not folate will present development of neuropathy
Term
What unrelated rxn to folate is B12 involved in?
Definition

Methylmalonic acid -->succynyl Coa

 

Methylmalonic levels will be elevated in cases of b12 deficiency but normal in folate deficiency

Term

B12 Absorption

 

In the mouth, what is b12 bound to?

 

what enzyme is released?

Definition

b12 is still bound to dietary protein

 

R-Binder is secreted but does not bind yet

Term

B12 absorption

What happens to B12 in the stomach?

 

what enzyme is releases?

Definition

Stomach is acidic, B12 is released from protein and binds to R-Binder.

 

 

intrinsic factor is released

 

Term

b12 absorption

 

in the duodenum, what happens to B12-R binder complex?

 

 

Definition

There is a neutral pH, pancreatic proteases are secreted which degrade R binder

 

at this neutral pH, free B12 is able to bind to intrinsic factor

Term

b12 absorption

in the ileum what happens to B12

 

Definition

the IF/B12 complex binds to a IF receptor on surface of ileal cells.

 

the complex is endocytosed and b12 released on basal side into circulation, bound to TRANSCOBALAMIN II

Term

B12 absorption

in the portal blood what happens to b12

Definition

Transcobalamin II transports b12 to the tissues

In the tissues the transcobalaminII/B12 complex binds to cellular receptors and is endocytosed by cells

Term
Acidity of stomach affects B12 how?
Definition
the acidity converts pepsinogen to pepsin, which hydrolyzes the dietary protein B12 is bound to
Term
Pernicious anemia?
Definition

slow disease spanning decades usally age 50-80

 

simple treatments of b12

Term

decreased stomach acid and its affect on B12 absorption?

 

what aspect of absorption would be affected?

Definition

decreases uptake

 

 

can happen by achlorhydria (decreased acid production by parietal cells) or use of antacids or proton pump inhibitors

 

pepsinogen to pepsin conversion would be decreased

Term
What is blind loop syndrome?
Definition

a pocket forms in wall of small intenstine leadin gto stagnation of nutrent flow and increased bacterial growth (which absorbs b12)

 

surgery corrects this

Term

DBIL test:

measures what

Definition

both unconjugated bilirubin and conjugated

 

measures 100% of CB and 10% of UCB

 

 

Term
IBIL measures what?
Definition
90% of unconjugated bilirubin
Term
purpose of heme catabolism?
Definition
To alter structure of heme(hyprophobic and toxic) to a nontoxic molecule that can be excreted
Term
Where does heme catabolism happen?
Definition

70% in spleen

15% in bone marrow

15% in tussues

Term

resident macrophages in spleen and bone marrow:

 

what is the intermediate between Heme and unconjugated bilirubin?

Definition
Hb-->Heme-->BILIVERDIN-->UCB
Term

Enzyme involved in Heme --> biliverdin?

 

biliverdin to bilirubin?

Definition

Heme oxygenase

 

biliverdin reductase

Term

UCB/Albumin travels in blood to the liver, and only UCB enters the liver. what prevents UCB from getting back out of cell?

 

Definition
tight junctions and GST
Term

How is bilirubin conjugated?

 

What transports CB to bile duct?

Definition

In hepatocyte, UDP-glucoronyl transferase (UGT) adds 2 negative charges to bilirubin making is more hydrophilic

 

 

MRP2, a unidirection transporter

Term

in the intestine, CB is metabolized by what?

producing what?

Definition

metabolized by colonic bacteria

 

urobilinogen

Term

What % of urobilinogen is excreted?

what is its rxn?

 

What % is reabsorbed?

of that %, what % is reabsorbed by liver and waht % is excreted in urine?

Definition

90%

fecal urobilinogen is autooxidated into stercobilin(brown)

 

10% reabsorbed,

9% is reabsorbed by liver,

1% is excreted in urine as urobilin

Term
What could pale stool be a sign of?
Definition
bile duct obstruction, preventing formation of urobilinogen
Term
Reabsorption of urobilinogen can be compromised by what?
Definition
liver damage
Term
What is the bilirubin measurement method?
Definition

Diazo rxn, gets TBIL and DBIL

Total-direct =indirect bilirubin

Term
What is jaundice?
Definition

yellowish discoloration of skin nails, sclera caused by an INCREASE in amount of BILIRUBIN in the blood called hyperbilirubinemia

 

 not a disease, but a sign of a disease

Term
what are the 4 types of jaundice and their causes?
Definition

neonatal

 

pre-hepatic

       caused by hemolytic anemia

hepatic

       caused by viral infection infecting integrity of liver

post-hepatic

       caused by bile duct obstruction

Term

What causes neonatal jaundice?

 

Definition

50% of all neonates are jaundice within first 5 days of life because:

immature liver fails to produce adequate amounts of proteins necessary for bilirubin excretion

 

increased RBC turnover which results in elevated UCB since not enough enzymes to process it

Term
Treatment of neonatal jaundice?
Definition

blue light therapy. NOT UV LIGHT!

 

bilirubin is light sensitive, the blue light alters the conformation of UCB yielding more polar photoisomers. These isomers can be excreted in the urine or bile without further modification.

 

so it makes the hydrophobic UCB for ampipathic

Term

Pre-hepatic jaundice

caused by? condition of liver?

Definition

hemolytic anemia, increased hemolysis of damaged/immature RBcs in spleen or bone marrow results in elevated UCB

 

No pathology of liver or duct

 

lab results will have IBIL increased due to increased UCB

Term
What is going on with hepatic jaundice?
Definition

Liver cells are damaged by viral infection.

UCB production normal, but conjugation is decreased

 

 

DBIL is still increased though beause CB leaks through leaky tight junctions

 

IBIL increased due to decreased conjugation

Term

Post-hepatic jaundice caused by?

 

affects on test results

Definition

Bile duct obstruction results in blocked flow of CB from bile duct to intestine.

 

results in little or no urinobilogen (pale stool)

bile acid induced damage to liver cells causes leaky tight junctions. then there is a leakage of CB into circulation

DBIL is increased due to leaking CB

 

fecal/urinary urobilinogen is decreased due to CB flow obstruction

Term
What are some defining test results for jaundice?
Definition

normal IBIL --> post-hepatic jaundice

normal DBIL--> pre-hepatic, liver undamaged so no leaky cells

decreased Urinary/fecal urobilinogen --> post hepatic, bile duct obstructed, no CB

 

In hepatic jaundice, everything is increased because of decreased conjugation AND leaky junctions. only thing normal is fecal

Term
If DBIL Normal?
Definition

Pre-hepatic jaundice

liver is fine

Term

if DBIL increased, and

 

urinary urobilinogen is...

increased?

decreased?

Definition

hepatic (can't reabsorb, so the 10% will go to urine)

 

post-hepatic

Term
What affects a phenotype?
Definition
both the genotype and environment
Term

What is a point mutation?

 

types?

Definition

single base pair change in sequence of dna

 

Transitions: a puring replaced with a purine, a pyrimidine is replaced by a different pyrimidine

 

transversions: a purine is replaced by a pyrimidine and vice versa

 

Term
what is a missense mutation?
Definition

if a point mutation occurs within a coding region, it can result in a change of a single amino acid.

 

Term
Silent mutation?
Definition
in many cases, the change in the third base of a codon does not change the amino acid sequence
Term
nonsense mutation?
Definition
point mutation leads to a stop codon, premature termination
Term
Diseases caused by point mutations?
Definition

Hemophilia B- reduces factor IX by 1/3 normal level

Tay-Sachs (some variants)- Hex A gene

Sickle cell- changes composition of b-globin. 1/12 blacks

    is a transversion point mutation

NF1 - stop codon into the NF1 coding region

B-thalassemia - decrease production of b-globin. many different kinds of pt mutations for this one

Term
Diseases caused by small deletions/insertions?
Definition

Tay-Sachs (most common variant)- small insertion of 4bp

Cystic Fibrosis - small deletion

 

Term

diseases caused by large insertions/deletions?

Definition

Red-green blindness - large deletion in the cones (of the eye)

a-thalassemia- large deletion by a aberrent recombination

Charcot-Mari-Tooth : most common inherited neurologic disorder, large insertion caused by aberrent recombination. hammer toe

 

 

Term
Waht is a polymorphism?
Definition
dna variation that is common in the population. two or more alternatives for that position, and all of them equally accepted. for frequent than mutations, at least over 1% of population
Term
What is a SNP?
Definition
A snip, is a single nucleotide polymorphism
Term
What is a STRP?
Definition
microsatellite, short 2-5 bp sequence of dna polymorphism
Term
What is a VNTR?
Definition
minisatellite, 14-500 bp long polymorphism
Term
What is a CNV?
Definition

1000-2000000 bp polymorphism

 

Term
What is a molecular fingerprint?
Definition
the repeat units in VNTR's ranges from person to person and is UNIQUE in every person. less that 1in10 billion chance of having the same
Term
Anemia with normocytic, normochromic and normal CR?
Definition
insufficient erythropoiesis
Term
A mendelian trait is produced by?
Definition
a single gene. Punnet squares etc
Term
An autosomal trait is
Definition
chromosome 1-22
Term

Mendel

Principle of Segregation (1st Law)

Definition

Sexually reproducing organisms possess genes in pairs, and only one of each pair is transmitted to a particular offspring

 

Term

Mendel

Principle of Independent Assortment (2nd Law)

Definition

Genes that reside on different loci are transmitted independently

 

later it would be shown that this law applies to only genes on different chromosomes or very far apart in same chromosome

Term

what disease has ~90% of its cases from new mutations?

 

Definition

Achondroplasia

 

autosomal dominant, FGFR3 gene

gain of function, missense mutation

Term

Homozygotes do not survive infancy (lethal mutation). this is true of all dominant diseases except?

 

Definition
Huntingtons disease
Term
What diseases are from new mutations?
Definition
achondroplasia, marfan, nf1
Term

hereditary hemochromatosis

 

what is its gene?

charcterstics? why is that bad?

Definition

HFE (think high iron(Fe))

 

iron overload. absorb 2-4 times the normal amount of iron. this leads to more of the Fenton rxn

Term

pleitropy?

 

heterogeneity?

Definition

A single gene exerts its effects on multiple aspects of anatomy

 

 

where mutations at different loci produce same phenotype

 

Term
In prader-willi, what is the gene involved?
Definition

SNRPN

 

this is the one coming from the father

Term
in angelman, what is the gene silenced?
Definition

ube3a

 

note, this is on chromosome 15

Term
anicipation is caused by?
Definition

trinucleotide repeat expansion

 

ex for myotonic dystrophy

5-50 repeats =no symptoms

50-100= mild symptoms

100-1000 = full dystrophy

 

number of repeats increases in generations due to slippage of DNA polymerase

Term
genetic load?
Definition

its believed that each person carries 5 recessive genes in heterozygous form, that would kill in homozygous form.

 

this is why consanguinity is bad

Term
Females have two copies of each x-linked gene, while men have one. how is this accommodated for?
Definition
one copy of each gene is inactivated
Term
What is the lyon hypothesis regarding x-chromosomes?
Definition

one of the 2 x chromosomes of females is randomly activated in every somatic cell.

 

x inactivation is random, UNLESS there is a gross structural abnormality of one of the x chromosomes in which the defective one is preferentially inactivated

 

this inactivation is permanent for all descendents of that cell

Term
what is Anhidrotic ectodermal dysplasia?
Definition
x-linked recessive disorder that has abnormal development of structures such as nails, hair teeth or sweat glands. there will be random set of areas with sweat glands and without for example
Term
What is a Barr body?
Definition

inactivated x chromosomes can be detected as a chromatin mass in somatic cells. women will have one less than the number of x chromosomes they have

 

ie. normal women will have 1 barr body. men 0.

 

Turner syndrom, XO, will have no barr bodies,

Quintuple x (XXXXX) will have 4

Term
What is the gene responsible for x inactivation?
Definition

XIST

 

it is in the x inactivation center (XIC)

it is only found in females

 

Term
If Barr bodies represent inactivated X chromosomes, why is having extra inactive chromosomes an issue? aren't they just silenced?
Definition
X inactivation will be incomplete
Term
X-linked recessive disease prevelence in males/females?
Definition

1in10000 males

1in 100,000,000 females

 

no father -->son transmission

Term
In Lesch-Nyhan syndrome, what is the issue with having a HGPRT deficiency?
Definition

in the normal condition, 10% of purine is synthesized while 90% is recycled.

 

in the pathological condition, the decreased activity of HGPRT largely decreases the ability to recycle purine, Huge increase in uric acid production. The brain relies on this parthway so there is CNS damage

Term
What is dystrophin's role in the body?
Definition

accounts for only .002% of striated muscle cell's protein mass. But it has a critical function in maintaining the structural integrity of the cells CYTOSKELETON

 

 

Term
Difference in Beckers MD and Duchennes MD mutation?
Definition

Duchenne's: most mutations are deletions that cause frameshifts leading to absence of dystrophin

 

Beckers: Usually result of in-frame deletion allowing for partial protein function

 

this is why Beckers is a milder form

Term
Duchene Muscular Dystrophy is x-linked recessive, but what is interesting about female heterozygotes?
Definition
They usally exhibit no symptoms as it is x-linked recessive, BUT, 8-10% exhibit some muscle weakness due to the random nature of x-inactivation, usally mild symptoms
Term
What is Gower's sign?
Definition

Children use hands to stand up. this is a sign of muscle weakness and possibly duchennes MD

 

Term
How is fragile-x unlike any other X-linked dominant disease?
Definition
there is reduced penetrance in females so men are twice as likely to get it
Term
What is heteroplasmy and homoplasmy regarding mitochondrial inheritence?
Definition

Heteroplasmy: when a daughter cell receives a mixture of mitochondria, some with a mutation and some without

 

Homoplasmy: when a daughter cell receives only a pure population of either normal mtDNA or mutated mtDNA

 

Mitochondrial diseases can show much variability due to heteroplasmy. the % of mutant mtDNA affects the expression

Term
What % of mitochondrial diseases are inherited from the mother?
Definition

100%

 

eggs have 200000 mitochondria while sperm only has 200.

 

 mitochondrial inheritence is non-mendelian

Term
Which tissues are most affected by mitochondrial diseases?
Definition
Muscle and CNS
Term
Many diseases with congenital malformations (present at birth) are considered what kind of disease?
Definition

Multifactorial inheritence

 

they are becoming a higher % of cases

Term
Are multifactorial diseases mendelian in inheritance?
Definition

No, there may be familial aggregation, but no clear inheritance pattern

 

also, it is not sex linked

Term
A pregnant women neglects to take folate supplements, so will her child have spina bifida?
Definition
not necessarily, multifactorial
Term
The additive polygenic model explains what kind of traits?
Definition

quantitative.

 

bell shape curve

Term

How are multifactorial qualitative traits explained?

 

Definition

Threshold model.

 

Everyone has liability for a disease but different thresholds, once you get to the threshold the trait appears

 

liability is composed of genetic and environmental factors

Term
does the threshold model work for all multifactorial diseases?
Definition
no, example diabetes.
Term
How do you calculate occurence risk for multifactorial traits?
Definition

impossible to do.

recurrence risk can be determined by observing data

Term
what are the two factors that can be used as predictors of recurrence in a multifactorial disease?
Definition

the more individuals affected in a family, the higher the recurrence risk

 

the less the degree of relationship with proband, the less the recurrence risk

 

the risk to relatives also varies with the severity of the condition of the proband

Term
importance of monozygotic twins in twin studies?
Definition

they are identical twins with the same genetics, so any differences are environmental

 

dizygotic or fraternal twins, are only as genetically similar as any other 2 siblings

Term

What is concordance value?

 

what is the equation used to obtain an estimation of heritability of a disease?

Definition

Cmz and Cdz

if both members of a twin pair share a trait, they are said to be concordant, if not:discordant

 

h=2(Cmz-Cdz)

 

concordance itself says nothing about the heritability of a disease, the estimate of a genetic component only comes from comparing concordance of mz and dz twins

Term
What 3 diseases do ALL US states require newborn screening for?
Definition

PKU, galactosemia and hypothyroidism

 

AZ screens 29 genetic disorders

Term
Phenylketonuria and its implications at birth?
Definition

there is a phenylalanine hydroxylase enzyme deficiency, which is required for conversion of phynylalanine to tyrosine.

 

because of the deficiency, both phenylalanine and phenylpyruvic acid build up, accumulate in the blood. they are toxic in high concentrations and produce IRREVERSABLE MENTAL RETARDATION by 1-month of age

Term
why do individuals affected with PKU have lighter hair and skin color?
Definition

One of the products of the rxn inhibited by PAH deficiency, is melanin (the other is adrenaline)

 

The products are decreased so less melanin

Term
Issues regarding Maternal PKU?
Definition

most women with PKU have given up diet therapy by child bearing which has no ill effects on them, but can cause irreperable harm to the fetus, causing brain damage

 

 

Term
How does the Tay-Sachs screening program work?
Definition

preimplantation genetic diagnosis (PGD)

 

couples with high risk for a disease can do this

Take a single cell from the 8-cell blastomere produced by in vitro fertilization and is tested by the PCR (polymerase chain reaction). if healthy, its reimplanted.

Term

What do high levels of fetal AFP correlate to?

 

low levels?

Definition

increased risk of neural tube disorders

 

Increased risk of trisomies

Term
earliest prenatal test that can be done?
Definition

at 10-11 weeks, chrionic villus sampling (CVS)

 

 

Term
What is the prenatal test that you have to wait the longest to do?
Definition

ultrasonography, 16-18 weeks

 

Term
What is the triple screen and what does it test for?
Definition

replacing the standard maternal AFP(a-Fetoprotein)

 

tests anencephaly, spina bifida, down syndrome

 

by measuring AFP, hCG and estriol

Term

What is the leading known cause of mental retardation?

 

as well as leaading cause of pregnancy loss?

Definition

chromosome abnormalities

 

Term
what is a karyotype?
Definition

ordered display of 23 pairs of chromosomes.

 

first classified by size and then position of centromere

Term

What is a p-arm?

q-arm?

Definition

p arm(p stands for petite) is always oriented at the top

q at the bottom and is the long end

Term

metacentric chromosomes?

acrocentric chomosomes?

submetacentric chromosomes?

Definition

centromere occurs near middle of chromosome

 

centromere is near telomere (shorter p arm), and looks different, has a stalk and satelite

 

centromere resides somewhere between telomore and the middle

Term

polyploidy?

tetraploidy?

Monoploidy?

Definition

gain of one or more entire chromosome sets

triploidy is 3 copies, so 69 chromosomes

tetraploidy refers to 4 copies of each. this is a lethal condition, only a few live births ever

 

monoploidy is loss of entire set, NOT seen in humans

Term

what is aneuploidy?

 

cause?

Definition

gain or loss of one chromosome

 

monosomies and trisomies are usually caused by nondisjunction (failure of 2 members of a chromosome to disjoin or or seperate) during meiosis I or II

Term
Occurrence of monosomies of chromosomes 1-22?
Definition
NEVER, these are lethal and not compatible with life!
Term
What are the autosomes in which trisomies are compatible with life?
Definition

Autosomes 13, 18, 21

 

these have the fewest genes of the chromosomes so can probably handle the excess material better

Term

What is trisomy 21?

 

Cause and origin of?

Definition

47, XY +21 or 47 XX +21

Down Syndrome

 

95% of trisomy 21 cases contributed to the mother

 

is the most compatible with live birth

 

only risk factor is advanced maternal age

Term
What is trisomy 18? details..
Definition

Edward syndrome

47 XX/XY +18

 

only 10% babies survive past a year, 50% a month

This is the most common, but more lethal, so not as many born as down syndrome

 

also 95% due to nondisjunction in the mother

Term
What is trisomy 13? details..
Definition

Patau syndrome

47 xx/xy +13

same, 95% nondisjunction caused by mom

 

 

Term
what is 45, X0
Definition

Turner, missing an x-chromosome, an abnormality of sex chromosome tolerated more than on autosomes

 

 

Term
What is 47 XXY?
Definition

Klinefelter

 

the fact that some phenotype is exhibited is due to incomplete x inactivation

Term
how do you get xx males and xy females
Definition

x and y chromosomes pair and undergo recombination. just proximal to the pseudoautosomal region there is a gene called SRY, which initiates male development.

 

In an aberrent recombination, the SRY gets sent to the x chromosome

Term

what are chromosomal translocations?

 

what are the two kinds?

Definition

interchange of genetic material between chromosomes

 

Reciprocal translocation: chromosomes break on 2 diff chromosomes, then they switch places

      generally no phenotype as there is no LOSS of material

 

Robertsonian translocation: involve only acrocentric chromosomes, 13 14 15 21 22, and involves loss of short arms of two acrocentric chromosomes and fusion of long arms at the centromere

      These offspring have one fewer chromosome, ie have a chromosome like t(14q21q). usually exhibit deletions or duplications

Term

What occurs in the G1 phase?

 

Definition

pretein synthesis, preparing for one cell to become 2, so the cell begins replicating non-chromosomal contents

 

Term
What occurs in the S phase
Definition
Dna Replication, each of the 46 chromosomes is duplicated
Term
What occurs in the G2 phase?
Definition
proofreading/repair, the cell double checks duplicated chromosomes for errors making any needed repairs
Term
What is the M phase?
Definition

(mitosis)

cell division, the duplicated DNA condenses into mitotic chromosomes and then daughter cells

Term
what are the "gap phases" of cell cycle?
Definition

G1/ G2

the cell is not actively replicating or dividing DNA

They are more active than G0, which is not preparing for cell division

 

 

interphase is all steps but mitosis

Term
What does the cell need to get to through the G1 stage?
Definition

A signal from a mitogen (growth factor) to enter a new round of cell division, without it, the cell enters G0

 

 

Term
What kind of cells will enter G0 and stay there, only dividing once?
Definition

muscle and nerve cells

 

others will continue to divide as long as they receive growth factor

Term

What are the cyclins and CDKs for the following sections

 

G1 early  

G1 late

G1/S

S/G2

M

Definition

Cyclin D     CDK 4/6

Cyclin E     CDK2

Cyclin A     CDK2

Cyclin A     Cdc2

Cyclin B     Cdc2

 

CDKs must be bound to a cyclin to have activity

Term
How does the cell cycle begin? starting with the mitogen...
Definition

Mitogen (growth factor) bind to receptors on surface of cell and initiate phosphorylation cascade through GTPase Ras. then goes to the MAP kinase which increases synthesis of actiity of transcription factor MYC. MYC increases expression of Cyclin D

 

Mitogen -> Ras ->MAP ->MYC -> Cyclin D

Term
What happens at R-point?
Definition

cell must cross Restriction (R) point, Cyclin D levels need to be high to stimulate CDK 4/6 and must also have growth factor mitogen. After this Cyclin E/CDK2 must kick in to help the cell pass the restriction point.

 

these proteins work together to phosphorylate Rb...

Term
How do Rb and E2F govern the restriction point?
Definition

Rb must be hyperphosphorylated to unbind from E2F which will go on to act as a transciption factor, leading cell to S-phase..

 

Cyclin D HYPOphosphorylates Rb

Ciclin E then HYPERphosphorylates Rb

 

Term
end all be all, what constitutes passage to s-phase?
Definition
hyperphosphorylation of Rb releasing E2F
Term
which protein inhibits all cyclin/CDK except CDK4/6?
Definition

p21

 

p16 gets Cyclin D/CDK4 6

 

these are called CDKI (inhibitors)

 

 

Term
during which phases can a CDKI halt the cell cycle?
Definition

all of them

 

p21 can also block cell at R point by inhibiting cylcin E/CDK2 which will prevent hyperphosphorylation of Rb

Term
what regulates p21?
Definition
p53, which responds to dna damage and promotes p21
Term

what kinds of stress is p16 induced during?

p21?

Definition

p16 is induced in times of environmental stress, i.e starvation

 

p21 is induced following DNA damage

Term
tumors are?
Definition

abnormal proliferation of cells

 

not regulated, replicating constantly

Term

where do carcinomas arise from?

 

sarcomas?

 

lymphomas/leukemias?

Definition

epithelial cells. 90% of tumors are carcinomas

ex: lung prostate breast skin colon

 

supporting tissues: rare 1%

ex blood vessels, adipose, muscle, bone

 

lymphatic and blood origin

lymphoma is a solid mass of white blood cells

leukemia is cancer in which malignant blood cells proliferate to blood stream

 

Term
How does clonal expansion of cancer take place?
Definition

a single progenitor cell gives rise to subpopulation of identical cells

 

takes years, cells acquire many additional mutations to progress to malignancy

 

Cancer is a progressive accumulation, otherwise cancer should be independent of age

Term
What are the properties of cancer cells?
Definition

Sulf sufficiency in growth signals

Loss of differentiation

Tissue invasion and metastasis

Sustained angiogenesis

Limitless replicative potention

Evading apoptosis

Term
how do cancer cells remodel cellular matrix of surrounding connective tissue?
Definition

Cancer cells secrete proteases called Matrix Metalloproteinases (MMPs)

 

the tumor and stroma evolve together, gives tumor cells ability to invade and metastasize (break through basal lamina)

 

Term
what is the process of metastasis?
Definition

invade local tisue and vessels

move through circulation

leave vesse

establish new colonies at distant site

Term
where do tumar cells from GI tract metastasize to?
Definition

Liver (Portal system)

 

so stomach and colon cancer often ends up in liver

 

this is because cancer cells are larger than RBCs so get caught in first capillary bed they end up in

Term
where do most tumor cells (not from stomach/colon) metastasize to?
Definition
lungs via the heart
Term
Where do tumor cells from breast and prostate metastasize to?
Definition

bone

 

 

Term

how do tumor cells use angiogenesis?

 

Definition
The cancer cells become hypoxic and need O2, Vascular endothelial growth factor VEGF responds and induced vessels grow to the area
Term
in its basic element, how do you get cancer?
Definition

either increased cell divisionand normal apoptosis

 

or

 

normal cell division and decreased apoptosis

 

both lead to a tumor

Term

what is a tumore initiator?

tumor promotor?

Definition

initiator: single application does not cause cancer but does cause latent genetic damage

ie. chemical or environmental

 

promotor: nonmutagenic, but repeated exposure to areas treated with tumor initiator will cause cancer

Term
what is the three-stage model a normal cell procedes through becoming a tumor?
Definition

initiation phase:   cell receives a mutation that gives it the potential for abnormal growth

 

promotion phase, the initiated cell is stimulated by external factors

 

progression phase:   cell has sustained enough mutations that it no longer needs external factors (such as growth factors) to proliferate

Term

example of a virus promoting cancer?

 

how does it do this?

Definition

HPV, human papilloma virus

 

HPV has two proteins, E6 and E7

E6 destroys p53, E7 liberate E2F

Term
how does gardasil work?
Definition
like a vaccine, exposure to HPV proteins so if a real virus comes body can mount an immediate response
Term
What two genes can mutations affect leading to cancer?
Definition

oncogenes (found in most cancers)

they are a mutated version of a proto-oncogene(NOT mutated), can bypass restriction checkpoints

 

tumor suppressors

when inactivated, tumor cells arise

Term

Oncogen mutations:

 

RTK mutation?

Definition

Receptor tyrosine kinases RTK, are normally only activated when a growth factor binds.

 

1: truncated ones do not need a ligand, they continually send messages

 

2: other cases, amplification results in too many receptors, so even small amounts of ligand signal cell growth

Term

what happens to Ras so it cannot stop cell division in tumors?

 

Definition
in 1/3 tumors, Ras becomes mutated so that it cannot hydrolyze ATP to turn off the signal response
Term
what is considered the "guardian of the genome?"
Definition

p53

50% of tumors have mutation or deletion of p53

Term

what are the 2 forms of Retinoblastoma?

 

 

Definition

Familial- occurs in family clusters, affected children frequently develop tumors in both eyes, because already have one mutation

 

sporadic - no family history, usually only develop in one eye due to its random mutation

Term
what is Li-Fraumeni syndrome?
Definition

hereditary defect in p53 gene

 

these individuals have a 90% chance of developing cancer, but no specific cancer dominates

 

some get multiple cancers in succession

Term
What is hereditary non-polyposis colon cancer? HNPCC?
Definition

also called lynch syndrom

 

inability to correct mistakes made during DNA replication. 90% of individuals with an HNPCC mutation will develop cancer

Term
What is xeroderma pigmentosum XP?
Definition

inability to repair DNA damage from uv radiation

 

skin cancer develops

Term
Breast cancer mutations?
Definition
BRCA 1 and BRCA 2 are essential for accurate repair of double-stranded DNA breaks. they mutate and lead to cancer
Term
Obesities link to cancer?
Definition
Strong correlation, no direct cause
Term
HPV infection put women at risk for cancer why?
Definition
HPV 16 and 18 deactivate cell apoptosis and cell cycle checkpoints become inactivated
Supporting users have an ad free experience!