Download Adobe PDF - CL Davis Foundation

Document related concepts

Messenger RNA wikipedia , lookup

Eukaryotic transcription wikipedia , lookup

Deoxyribozyme wikipedia , lookup

Evolution of metal ions in biological systems wikipedia , lookup

RNA polymerase II holoenzyme wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Proteolysis wikipedia , lookup

Lipid signaling wikipedia , lookup

RNA-Seq wikipedia , lookup

Gene regulatory network wikipedia , lookup

Endogenous retrovirus wikipedia , lookup

Point mutation wikipedia , lookup

Secreted frizzled-related protein 1 wikipedia , lookup

Two-hybrid screening wikipedia , lookup

Biochemical cascade wikipedia , lookup

Epitranscriptome wikipedia , lookup

Silencer (genetics) wikipedia , lookup

Gene expression wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

Transcriptional regulation wikipedia , lookup

Signal transduction wikipedia , lookup

Paracrine signalling wikipedia , lookup

Transcript
Cellular and Molecular
Pathology: Mechanisms of
Cellular injury
Part 1
Mark R. Ackermann
Department of Veterinary Pathology
College of Veterinary Medicine
Iowa State University
Ames Iowa 50011-1250
[email protected]
515 294 3647
Summer 2008 Copyright ©
Kinases. Kinases are proteins that are enzymes. Kinases also add phosphates to other things, like
fructose. Pyruvate kinase actually transfer a P group for phosphoenolpyruvate to ADP; so it is
adding a P onto ADP to make ATIP (see below). Kinases also activate other enzymes by adding
phosphates groups; in a few cases, addition of the phosphate turns off an enzyme.
kinase
Sugar (no P group) ------Æ Sugar-P
kinase
Enzyme (not active) -------Æ Enzyme-P (now active) (in a few cases, the P actually inactivates the
enzyme).
Phosphatases take off the P group and generally inactivate enzymes
enzymes, etc
etc.
Phosphatase
Enzyme-P (active) -----Æ Enzyme (no P and not active, generally)
Specific examples:
Hexokinase (hexose is a six-carbon sugar)
Glucose ---------------------Æ Glucose 6 P (with P on, the glucose cannot be transported out of the
cell; it also commits the Glucose to GLYCOLYSIS OR GLYCOGEN SYNTHESIS)
Pyruvate kinase
Phosphoenolpyruvate and ADP ----------------Æ pyruvate plus ATP
Tyrosine kinase (a receptor activated by a growth factor binding)
Cell signaling molecule (no P) -----------------------------------------------------------Æ Cell signaling
molecule-P
Injury resulting in ATP loss
•
•
•
•
•
•
•
•
•
Hypoxia/ischemia
Burn, cold
Radiation
Chemicals/drugs/toxins
Infectious agents
Immunologic/autoimmune
Inflammation
Nutrition
Genetics
1
Normal cell volume control
• Na+ is extracellular
• K+ is intracellular
• Membrane permeability
– Selective passage
• Water, carbon dioxide, oxygen, benzene, urea, glycerol
– No passge
• H+, sodium, bicarbonate (HCO3), potassium (K+),
calcium, chloride, magnesium, glucose (some cells),
sucrose
• Many ions and molecules are regulated by:
– Transportors, Channels, and ATPase pumps
Transportors
• Transportors are often
– Slow, 102 to 104 molecules/second
– Do not use ATP
Na+-glucose
• Unitransportors
– Transport
T
t one ion/molecule
i / l
l
Symport
• Coupled transportors
– Transport two ions/molecules
Na+
• Symport (both transported in the same direction)
• Antiport (transported in opposite directions)
Antiport
glucose
Transportors
• Unitransportors
– Glucose
• Can also be coupled with Na+ in the small intestine and
renal tubules
– Fructose
• Intestine and liver
• Coupled transportors
– GABA, Norepinephrine, glutamate
• Symport with Na+
– Peptides and amino acids
• Symport with Na+/H
• Transportors also for:
– Pyruvate, fatty acyl Co A, and ATP
2
Channels
•
Channels are often
–
–
–
–
Fast 107 to 108 ions/second
Do not use ATP
Connect cytosol to cell exterior through narrow passages (ie. pores)
Selective
•
•
•
•
Mainly transport Na+, K+, Ca2+ and Cl-
Transport efficiency is greater than carrier proteins
Transport is always passive
S
Secondary
d
tto membrane
b
d
depolarization
l i ti (voltage-gated)
( lt
t d)
–
Examples
•
Na+/K+ channels for nerve fiber conduction
•
Ca++ channels in sarcoplasmic reticulum of muscle
•
Chloride (Cl-) channels
–
–
–
–
–
–
Nearly 20 disorders known involving defects in skeletal muscle to nervous system
Long QT syndrome
» Loss of K+ channel function or gain of Na+ channel function
Defects in Ryanodine receptor
» malignant hyperthermia
Ligand-gated channels in synapses, CFTR channels, and ClC channels (numerous functions)
Defects in CFTR Cl- channel result in cystic fibrosis
» Although CFTR is a ABC pump that utilizes ATP, it is considered a channel due to the rapid influx
of chloride it can allow
Defects in voltage-gated CLC chloride channels
» Myoclonia congenita, Dent’s disease, Bartter’s disease, osteopetrosis
Figure 1
Inherited mutations alter ion channel function
and structure and cause human disease.
Mutations may alter the permeation pathway
(A) to inhibit the movement of ions through
an open channel pore and may also alter ion
channel gating by changing either the process
by which channels open (activate) (B) or the
process by which they inactivate (C)
(C).
Transitions from the open to the inactivated
state reduce the number of channels that are
available to conduct ions. Mutations that
destabilize the inactivated, nonconducting
state of the channel are gain-of-function
mutations and are common to diverse
diseases, including LQTS, certain forms of
epilepsy, and muscle disorders such as
hyperkalemic paralysis.
Kass RS: J Clin Invest 8:1986-1989, 2005
Na+
K+
Ca+
Cl+
Kass RS: J Clin Invest 8:1986-1989, 2005
3
Inherited loss of channel function:
Malignant hyperthermia
• Dysfunctional ryanodine receptor (RYR
1) activity
– A calcium channel
• Sensitive to halothane/stress
• Muscle contraction
– Myosin ATPase activity, glycogenolysis,
glucolysis, further release of calcium,
excessive heat, lactic acidosis, ATP loss
Inherited loss of channel function: Cystic
fibrosis
• Inherited defect of Chloride channel
– CFTR (cystic fibrosis transmembrane
conductance regulator)
• DeltaF508 mutation in allele
– 90% of CF patients have this mutation
– Results in CFTR remaining in ER and being degraded by
ubiquitin
Cystic fibrosis
• Sweat test is done clinically, but not definitive
• Loss of ions in respiratory secretions
– Mucus layer becomes thick
– Also affects gastrointestinal secretions and pancreatic
secretions
• Increased susceptibility to respiratory infections,
especially Pseudomonas and Burkholderi which are
almost impossible to eliminate
– Loss of ionic concentrations
– Loss of antimicrobial peptide (defensin activity)
– CFTR a receptor for Pseudomonas
4
CLC Chloride Channels
• Nine mammalian genes encode for
channels denoted CLCN1 to CLCN7 and
CLCNKa and CLCNKb
• Present in plasma membrane and
membrane of intracellular organelles
• Function to stabilize membrane potential,
synaptic inhibition, cell fluid volume
regulation, transepithelial transport,
extracellular and vesicular acidification
and endocytotic trafficking
CLC Chloride Channel Structure
• Double-barreled channels
• Each pore is gated and can be opened
and closed individually; gating is
independent
• Common gate exists to close both pores
simultaneously
• Single gate mutations
• Common gate mutations
5
Myotonia Congenita
• Impairment of skeletal muscle relaxation
after contraction
• Mutations in CLC1 which can cause either
dominant ((Thomsen’s)) or recessive
(Becker-type) disease
• Recessive form more common and more
severe
• Dominant mutations act on common gate
affecting both pores
Myotonia Congenita
• Treatment usually not necessary
• Mexiletine (sodium channel blocker)
Myotonia
6
Bartter Syndrome III
• Renal tubular disease characterized by loss of NaCl
reabsorption; PU/PD, bouts of severe dehydration,
failure to thrive
• Mutation in CLC-Kb with autosomal recessive
i h it
inheritance
• Deafness not present because both CLC-Ka and
CLC-Kb are present in inner ear
• Mutation in barttin (beta subunit present in both
CLC-Ka and CLC-Kb) leads to deafness
Bartter Syndrome III
• Treatment based on
clinical presentation,
not genetic mutations
• Na+ and K+
supplements
• ACE inhibitors
• Indomethacin
• Growth hormone
• Response to therapy
variable
Dent’s Disease
• X-linked recessive gene mutation in
CLC5 with proteinuria,
hyperphosphaturia, hypercalciuria and
nephrocalcinosis
h
l i
i
• Endocytosis is impaired
• Luminal parathyroid hormone increases
causing decrease of Na-PO4
cotransporter
• Serum concentrations of active vitD3
variable
7
Dent’s Disease
• Treatment for nephrocalcinosis may slow
progression to end-stage renal failure
• Thiazide diuretics used which enhance Ca2+
reabsorption
Dent’s Disease
Osteopetrosis
• Reduced or absent acidification of
resorption lacuna leading to failed bone
resorption by osteoclasts which results in
brittle bones
• Autosomal
A t
l recessive
i or d
dominant
i
t lilinked
k d
mutation in CLCN7
• CNS neurodegeneration resembling
lysosomal storage diseases produced in
knockout mice
• Retinal degeneration
8
Osteopetrosis
• Treatment success variable
• Bone marrow transplant
• Vitamin D administration (calcitriol) to
enhance bone resorption
• Prednisone given to improve
hematological alterations (neutropenia
and anemia)
Osteopetrosis
Conclusions
Numerous diseases
associated with
CLC defects
attributes to their
wide range of
physiologic
functions and
emphasizes their
medical
importance
9
ATPase pumps
• ATPase pumps are often
– Slow, 10 to 103 ions/molecules/second
– All require ATP (by definition)
• ATP
ATPase
ADP+Pi
– ½ of all ATP is used to maintain transport
gradients
• Types of ATPase pumps
– P, F, V, ABC
ATPase pumps
• P type ATPase pumps
– Na/K, H, Ca transported
– Locations
• Plasma membrane for Na/K
– 3 Na+ out, 2 K+ in
– For cell homeostasis
• Apical plasma membrane of stomach (H+/K+)
• Plasma membrane of all cells (Ca++)
• Sarcoplasmic reticulum (Ca++)
ATPase pumps
• F type ATPase
– H+ ion only
• Inner mitochondrial membrane
– And chloroplast thylakoid membrane (plants)
10
ATPase pumps
• V type ATPase
– H ion only
• Especially for pH regulation
– Locations
• Endosome, lysosome, secretory vesicles
• Plasma membrane of osteoclasts, renal epithelial cells,
neutrophils, macrophages
– Activities
• Endosomal killing/degradation, bone resorption,
breakdown of ligand/receptor complexes (thus controlling
regulation of receptor density), release of enzymes from
Mannose-6-phosphate receptors (thus regulating enzyme
activity)
ATPase pumps
• ABC type ATPase pumps
– Pump ions (many types) and lipophilic substances
– Location and substance
• Endoplasmic reticulum
– Peptide transport for MHC/Ag processing
• Plasma membrane
– MDR (P-glycoprotein)
» Transports lipid-like toxins/drugs
• Macrophages
– ABCA1
C 1
» Transports cholesterol and phospholipids
– defects contribute to atherosclerosis
• Rod photoreceptors
– ABCA4
» Transports retinoyl derivates
» Defects result in macular degeneration
• Liver
– ABCB11 (BSEP)
» Transports bile salts
Cellular energy: required for ATP
Glucose/glycogen
Proteins
Pyruvate
Fatty acid
Fatty Acyl Co A
ACETYL CO A
KREBS (produces NADH, FADH2 for Oxidative phosphorylation)
OXIDATIVE PHOSPHORYLATION
+ NADH
+02
=ATP
Mitochondria
ATP for:
ATPase pumps, signaling, etc.
A transporter is required for:
Fatty Acyl Co A, pyruvate and ATP
Cell plasma membrane
11
Mitochondrial ATP formation
• The outer mitochondrial membrane is
permeable to H+ ions
• The inner mitochondrial membrane is
impermeable
• NADH generated in Kreb’s cycle
– Releases electrons (H+)
(H ) that enters oxidative
phophorylation
• H+ ion is moved from the inner mitochondrial
matrix to the area between the inner and
outer mitochondrial membranes
• Oxygen enters, is converted to water
• H+ enters the F1 ATPase
– Converts ADP to ATP as H+ passes back to the
inner mitochondrial area
Mitochondrial formation of ATP
Cytoplasm
Outer mitochondrial
membrane
H+ H+
H+
FO/F1 ATPase
Impermeable to H+
Inner mitochondrial
membrane
H+
NADH+
½ O2
NAD
H 20
H+
ADP
Pi
ATP
Mitochondrial matrix
Sites of inhibition to ATP synthesis
H+ H+
NADH/CoQ:
ROTENONE
H+
FO/F1 ATPase
H+
NADH+
NAD
Cytochrome oxidase:
CN, AZIDE, HYDROGEN SULFIDE
HERBICIDES, NO
½ O2
ADP
Pi
Loss of membrane
potential:
DNP, SALICYLATE,
VALINOMYCIN,
GRANICIDIN
H 20
H+
ATP
FO/FA ATPase:
DDT, other drugs
Lack of oxygen:
RESPIRATORY PARALYSIS
HYPOXIA, ISCHEMIA (ergot alkaloids,
Infarct, cocaine toxicity), CO, MET Hb
12
Impairment of mitochondrial synthesis of
ATP
• Inhibition of NADH production
• Lack of glucose
• Fatty acid oxidation
• Loss of protein
Effect of ischemia on cells
•
•
•
•
•
•
•
•
•
Decreased oxidative phosphorylation
Decreased ATP synthesis
Decreased ATPase pump activity
Loss of inner mitochondrial membrane potential
Loss of K+ from the cytoplasm
Influx of Na+
Increased intracellular Ca++
Glucose/glycogen breakdown
Lactate formation
– decreased pH
• Decreased transport vesicles
• Decreased synthesis of proteins, lipids, loss of phospholipid
turnover in membranes
Increased intracellular calcium
•
Cytosol levels normally are 10-100nM (extracellular levels are 1-2 mM)
•
•
Inositol triphosphate (IP3) and other factors can induce release
Increased Ca++ levels can pass the external mitochondrial membrane
–
–
–
Intracellular storage in smooth and rough endoplasmic reticulum
Storage kept in check with Ca++ATPase pumps
If persistent, the calcium can then induce the internal mitochondrial membrane to form
pores
•
•
•
•
Pores are formed with adenine nucleotide translocase (ANT), voltage-dependent anion channel
(VDAC), and cyclophilin
Mitochondrial permeability transition, a state of calcium permeability by the inner membrane
Enhances oxidative phosphorylation temporarily and free radical formation
Eventually damages the inner mitochondrial area and calcium aggregates form
•
Simultaneously, calcium activates phospholipases and calpain
•
•
Reduces ATPase activity
Enhances calcium-dependent endonuclease activity and DNA degradation
–
–
Phospholipase A2 degrades plasma membranes
Calpain is a cysteine protease
13
Robbins and Contran Pathologic basis of disease, 7th edition
Cell injury: reversible/irreversible changes
Robbins and Contran Pathologic basis of disease, 7th edition
Brown fat utilization and thermogenesis
• Cold increases
– PGC1 (a powerful transcriptional coactivator) for PPAR
gamma (peroxisome proliferator activator receptor)
– TR (thyroid hormone receptor)
– RAR (retinoic acid receptor)
– ER (estrogen receptor)
• PGC1
PGC1, TR
TR, RAR and
d ER activate
ti t UCP ((uncoupling
li proteins)
t i ) and
d
genes of mitochondrial respiratory chain for ATP synthesis
(ATPase and cytochrome oxidase C)
• The UCP (uncoupling proteins) are in the inner mitochondrial
membrane along with F0/F1 ATPase.
• The UCP allow H+ passage with loss of gradient resulting in heat
with reduced ATP.
– PGC1 also activates NRF 1and 2 (nuclear respiratory
factors) for mitochondrial biosynthesis and increases
conversion of type I to type II muscle fibers
• Increased mitochondria for more heat
14
Radical formation
• Radicals, reactive oxygen species (ROS)
– Form secondary to:
• Ultraviolet light, cell metabolism, inflammation
– Cell metabolism
• Of all oxygen used by cells
– 2% is converted to ROS by mitochondrial electron
transport system (ETS)
– 1/100 oxygen molecules cause protein damage
– 1/200 oxygen molecules cause DNA damage
– Oxidation: loss of an electron
– Reductant: Donates electrons via addition of a
hydrogen ion or removal of an oxygen molecule.
Free radicals
• Free radicals
. :
– An unpaired electron in an outer orbital
– Example: hydroxy radical
: :
• :O:H hydroxy radical (.0H) a radical
• :O:H hydroxy ion (-OH) not a radical
• The dot denotes an unpaired electron, but no inference about
charge; the negative sign denotes charge
• Singlet oxygen (O2)
.
• Superoxide anion (O2 -)
• Hydroxy radical (.OH)
• Peroxyl (ROO.) and alkoxyl radicals (RO.)
An unpaired electron
Nonradicals that are reactive and
contribute to radicals
•
•
•
•
•
•
•
Hydrogen peroxide (H2O2)
Hypochlorous acid (HCLO)
Organic peroxides
Aldehydes
Ozone
Singlet oxygen (1O2)
Peroxynitrite (ONOOH)
15
Terms
• Reactive oxygen species (ROS), oxygenderived species (ODS), oxidants, and
reactive nitrogen species (RNS)
– These are all often used interchangeably.
Free Radical Formation
•
Reactions for Formation
»
Fe++ Fe +++
• Fenton Reaction
H2O2 Æ OH.
• Haber Weiss reaction
H2O2 + O2. Æ OH.
• Electron transport system Æ O2. - = O = O. - (unpaired
electron)
• Cytochrome P450, xanthine oxidase Æ O2. • Ionizing Radiation H20 Æ .OH
• NADPH
oxidase NADPH -2 e’s + 2O2Æ NADP+ + H+ + 2 O2
-
• Lysyl oxidase-crosslinks collagen
• NOS nitric oxide synthtase 1, 2, 3 (neuronal, endothelial,
macrophage) Æ (peroxynitrite) OONO.
• Radical + Nonradical = radical
• Reperfusion injury – xanthine oxidase
Properties of free radicals
•
Hydroxy radical (.OH)
– The most reactive radical known in chemistry
– Diffuses a short distance, 3 nm, short-lived (10-10 sec)
• So active that it doesn’t travel far
• Must be made close to DNA; H2O2 is the latent form that gets close to DNA and then is
converted to hydroxy radical through the fenton reaction
• Damages deoxyribose, all four DNA bases, phosphate backbone, lipids
•
.
Superoxide anion (O2 -)
– Diffuses a short distance, short-lived
– Affects guanine selectively
•
•
Alkoxyl (RO.)(10-6 sec) and peroxyl (ROO.)(17 seconds)
Nitric oxide (NO.)/peroxynitrite (ONOO- or ONO2)
– ONOO- diffuses a longer distance, 9 um, long-lived (minutes)
• Can deaminate or nitrate (add nitrogen) to nucleotides
• Especially damaging to guanine
– Results in formation of 8-oxo-deoxyguanine (easy to detect)
» ONOO- is more damaging to 8-oxodeoxyguanine than guanine
– Also induces formation of 8-nitro-deoxyguanine (difficult to detect)
•
Non radicals, also a short livespan (seconds to hours)
16
Exogenous and endogenous sources
• Exogenous sources:
– Gamma radiation, UV, ultrasound, food,
drugs,
g p
pollutants, xenobiotics, toxins
• Endogenous sources:
– below
Exogenous sources
• Reactive oxygen species (ROS) sources:
–
–
–
–
–
–
–
–
–
–
Ionizing and non-ionizing radiation
Phorbol esters
Bleomycin, paraquat (these induce fibrosis)
Organic peroxides
H
Heavy
metals
l ((next slide)
lid )
Asbestos (fibers)
Smoke
Silica
Nanoparticles
Gases
• Ozone
• di-oxygen (O2) (oxygen itself)
– and many others
Metals and free radical formation
• Metals generate free radicals via the Fenton reaction
• Reduction/oxidation-active metals
– Iron, copper, chromium, and cobalt
• Increase ROS through a Fenton-like reaction
• Reduction/oxidation-inactive metals
– Lead, cadmium, mercury, nickel
• Deplete cells of antioxidants
– Especially thiol-containing antioxidants (glutathione reductase) through binding of
these metals to SH groups
• Activation of transcription factors: NF-kappa B, AP-1 and p53
– Metal-induced formation of ROS activate these transcription factors, all
of which are redox-sensitive
• Metals induce mutagenic lipid products.
– Lipid peroxides, formed from ROS action on phospholipids (more later),
further react with metals to produce mutagenic lipids (malondialdehyde,
4-hydroxynonenal, exocyclic DNA adducts (etheno adducts)).
• Antioxidants may be important for heavy metal poisoning
17
Endogenous location of free radical
formation
• Endoplasmic reticulum (rER, sER)
– sER biometabolism
• Cytochrome P450, conversion of carbon tetrachloride to radical CCl4 to
CCl3
• Mitochondria
• Ubiquinone
• Cytosol
– Fenton reaction, xanthine oxidase, U.V.
• Peroxisome
– oxidases
• Leukocyte granules
– NADPH oxidase
• Extracellular matrix
– Lysyl oxidase (cross links collagen)
Principal locations of radical formation in cells
Cytosol:
Nitric oxide synthetase
UV: hydroxy radical
Fenton reaction
Reperfusion injury
Granules:
NADPH oxidase
sER:
Cytochrome p450
-biotransformation
Mitochondria:
Mit
h di
Oxidative phosphorylation
Peroxisomes:
Catalase, oxidases
Extracellular matrix:
Lysyl oxidase
Robbins and Contran Pathologic basis of disease, 7th edition
18
Reperfusion injury
• Reperfusion injury occurs in sites of
ischemia that once again receive
oxygen
yg
– Examples:
• myocardial infarction
• stroke
• dissolution of a thrombus (clot)
Reperfusion injury
• Exuberant free radical formation occurs
with reperfusion due to:
– Xanthine oxidase pathway
– Mitochondrial electron transport chain
– Conversion of NOS to produce superoxide
(rather than NO)
– NADPH oxidase from infiltrating leukocytes
19
Free radical formation during reperfusion:
xanthine oxidase
Cell ischemia/loss of oxygen leads to ATP degradation. If perfusion is
re-established, oxidative radicals are formed.
ATP
ISCHEMIA
ADP
Xanthine dehydrogenase
AMP
Adenosine
Inosine
Xanthine oxidase
O2
O2
-
Xanthine oxidase
Xanthine
Hypoxanthine
SOD
Uric acid
H2O2
Fe++
REPERFUSION
Tissue injury
.
OH
Proximity of muscle and other cells to
endothelial cells in reperfusion injury
• Direct contact between myofibers and
other cells to endothelial cells allows
passage
p
g of nucleotides via nucleotide
transport proteins (NTP) that can be
used in the xanthine oxidase pathway
Muscle or other cell
ATP-ADP-AMP-Adenosine-Inosine
NTP
NTP
Adenosine-Inosine
X.O. pathway
Endothelial cell
Mitochondrial activity contribution to ROS
in reperfusion injury
• Distal to NADPH dehydrogenase in the
mitochondrial electron transport chain
– Ubiquinone (CoQ) increases ROS
f
formation
ti
• ROS induce mitochondrial permeability
transition (MPT)
• These contribute to ROS formation and
reperfusion injury
20
Increased superoxide generation from
NOS during reperfusion injury
• All three NOS (nNOS, eNOS and iNOS) can
switch from NO to superoxide generation
– This occurs with depletion of NOS substrate
( i i and
(arginine
d BH4)
• Both arginine and BH4 are depleted by ROS (vicious
cycle)
• The increase superoxide production
contributes to ROS damage and reperfusion
injury
Leukocyte infiltration with reperfusion
injury
• Hypoxic tissues, including myocytes
and endothelial cells increase
expression of leukocyte adhesion
molecules
l
l
– Enhanced leukocyte (neutrophil) infiltration
with reperfusion
– Increased activity of NADPH oxidase by
infiltrating leukocytes
• NADPH oxidase produces additional ROS
Reperfusion injury therapy
• Therapies
– Needed to reduce ROS damage but yet
allowing
gp
perfusion and return to normoxia
21
ROS lipoperoxidation
• ROS commonly cause lipoperoxidation
– Linoleic acid, the most common polyunsaturated
fatty acid in cells
– One radical reaction can oxidize 60 molecules of
linoleic acid
– One radical reaction can oxidize 200 arachidonic
acid molecules
• Lipoperoxidated fatty acids are short lived
– Reduced by glutathione peroxidase to a nonradical, or
– Interact with a metal to form:
• Aldehydes: malondialdehyde and 4hydroxynonenal (HNE)
• 4-HNE can interact with a metal to form an epoxide
Free radical damage: targets
• Predilection sites of radical formation/damage
• Mitochondria
– ETS-major site of free radical formation
• Phagosomes, peroxisomes
• Enzymes with divalent cations
– CU++, Zn++, Fe++
– Via Fenton-like reaction, damage occurs at the enzymatic active site
• Endoplasmic reticulum membranes
• Damage to DNA
– O
OH- formed
f
in nucleus when H2O2,
2O2 a latent fform, diffuses
ff
to nucleus
where it can be converted to hydroxy radical by the Fenton reaction
– Peroxynitrite, diffuses longer distances and can cause DNA damage
• Lipid
• Lipoperoxidation, fatty acid loss
• Protein and glycoconjugates
• Oxidizes SH groups to S-S and crosslinks often at cysteine or
methionine residues
• NO
– Nitration: NO binding to an amino acid
– Nitrosation NO binding to SH, amine, or hydroxyl group
• Oxidized proteins:
– Reduced binding of transcription factors, p53, Rb, and enzymes such as
kinases, phosphatases, and DNA repair enzymes.
Oxidative effects on cell signaling
22
Free Radical Scavengers
• Enzymatic
– Superoxide dismutase, SOD
• Converts (O2.-)superoxide anion to H2O2
• SOD1, cytoplasm (zinc/copper)
• SOD2, mitochondria (mangenese)
• SOD3, extracellular
– Glutathione peroxidase
peroxidase, GPX,
GPX glutathione reductase (repairs S
S-S)
S)
• Converts H202 to water (H2O); also reduces glutathione
– Thioredoxin (repair S-S)
– Catalase
• Converts H2O2 to H20
ROS scavengers
• Nonenzymatic (scavenge radicals)
–
–
–
–
–
Alpha tocopherol (vitamin E)
Ascorbic acid (vitamin C)
Retinoic acids (vitamin A)
Lycopenes (Vitamin A-like)
Flavinoids, genistein, resperpins, resveratrol, quercetins—
electron donors
– Ferritin, lactoferrin, ceruloplasm, hemoglobin,
metallothionen, uric acid, histidine residues
– Bind divalent cations or serve as electron donors
– Glutathione (glutamic acid-cysteine-glycine), melatonin (an –
NH group), histidine dipeptides (caronsine, anserine)
Nonenzymatic scavengers
• Nonenzymatic scavengers
– Also termed low molecular weight
antioxidants (LMWA)
– Donate an electron to the radicals but not
reactive themselves
• A wide spectrum of activity--nonspecific
– Better permeability throughout the cells
than enzymes
• Can penetrate cell membranes
• Allows radical scavenging in more areas of the
cell
23
Repair of lipoperoxidation of phospholipids:
Vitamin E and C
PL
Fatty acyl CoA
TOC-OH
Lipoperoxidation
O-ASC-OH
PL-OO
O=ASC=O
HO-ASC-OH
lysophospholipid
PL-OOH
2GR SS
2GR-SS
2GSH
GR-[SH2]
GSSG
TOC-O
GRO-SS
GRO-[SH2]
FA-OOH
GPX
FA-OH
2GSH
GSSG
Phospholipid membrane
2GR-SS
GR-[SH2]
NADPH+ + H+
PL phospholipid
PL-OO
phospholipid that is oxidized (lipoperoxidation)
TOC alpha tocopherol, Vitamin E
ASC ascorbic acid, Vitamin C
NADP+
GRO glutaredoxin
GSH glutathione (GSSG [oxidized])
GR glutathione reductase
GPX glutathione peroxidase
Free radicals and the “penumbra zone”
• Penumbra zone
– A region around an area of necrosis,
infarction that is hypoxic
yp
– Occurs stroke, myocardial infarction or
other inflammatory disease
• The tissue in the penumbra zone may or may
or may not die, depending upon the amount of
hypoxia and level of free radicals
Penumbra zone
24
Free radical theory of aging
• Harman Denhan
– 1956
– Developed the “free
free radical theory of
aging”
• A concept that tissue damage caused by free
radicals results in a “wear and tear” type of cell
injury that is prolonged and over the years,
leads to aging changes in cells
Oxidative stress versus reductive stress
• Redox (reduction/oxidation) potential
– The balance between reductants and oxidants
– The “redox state” is tightly regulated in the cell, like pH
• Free radical theory of aging and suggests that all oxidative
stress is damaging
– But this is not always true, some oxidative stress is needed
for cellular physiology
• Oxidation is needed for thyroglobulin binding to iodine and thus
thyroxine activity
• Oxidation needed for neutrophil/leukocyte killing and lysosomes
– Many studies to prevent cancer and disease by use of antioxidant nutrients do not show great improvement
• Reductive stress, the opposite of oxidative stress can also be
overdone
• A balance in oxidative and reductive stress is likely optimal
– Eat a balanced diet and you’ll be ok
•
“Everything in moderation, including moderation itself”
– Michael Tassler, 1984
Cell signaling and oxidative changes
• GTP-binding protein families
– RAS and RHO
• RHO
– Induces actin and cytoskeletal changes
• RAS
– Active when bound to GTP
– Hras, Nras and Kras, three subtypes
» Kras most ubiquitous expression
– Farnesyltransferase modifies RAS post-translationally
» Allows RAS to be come functional
» Farnesyltransferase is targeted for therapies to reduce RAS
activity
– Epidermal growth factor (EGF) induces RAS activity
» Leads to: RAF/MEK/ERK/C-jun/AP-1 (more following)
» Also leads to: PI3K/AKT and inhibition of BAD (cell survival)
» Also leads to: FALGDS and PLC/protein kinase C (cell
prolifeartion)
» All of the above increase cell survival, cell cycle progression
and calcium signaling
25
RAS
• RAS/RAF/MEK/ERK and RAS/MEK/JNK
activate cJun pathway
– C-Jun a part of AP-1 (activated protein 1)
• AP-1 is a dimeric basic region-leucine zipper
transcription factor
– AP-1 is composed of c-jun and c-fos
» Other
Oth such
h transcription
t
i ti factors
f t
are Maf
M f and
d ATF
• AP-1 and these transcription factors bind
– TPA response elements (TRE) and c-AMP response elements
(CRE)
– TRE and CRE are promoter regions of genes that encode:
» Other transcription factors, matrix degrading proteins,
cyclins, cell adhesion molecules, and cytokines.
Mechanistic basis of free radical activity of
signalling
• Oxidative radicals can increase signaling
pathways
• Any step in these pathways can become
mutated and increase activity
– Acti
Activation
ation of gro
growth
th factor receptor (listed abo
above)
e)
– Activate NFKappa B
» OH- can induce release of NFKappaB inhibitory
subunit
– increased phosphorylation of an enzyme (kinase)
– decreased dephosphorylation (decreased phosphatase
activity)
» ONOO- can nitrate (add N) to tyrosine (on tyrosine
kinases) and block phosphorylation
– Damage to cysteine residue on active enzymatic sites of
regulatory phosphotases such as PTEN (inhibits PI3K)
– Activate oncogenes
» C-jun is activated by H2O2 as is MAPK
Cell proliferation
26
Cell cycle
• Induction from Go to G1
– Growth factors
– Signaling
• Cyclins
• Checkpoints and inhibitors
– G1 to S
– G2 to M
• Checkpoint control
• Transcription and passage through G1/S
– E2F activation
Response of cells to injury
DNA injury
Dividing cells
Non-dividing cells
Inhibition of proliferation
Apoptosis/death
(Failure of repair)
Cancer
Repair
Mutations
Signaling pathways and receptors: some initiate cell
proliferation
•
•
•
•
•
•
PI3 kinase
MAP-kinase
IP3 pathway
cAMP
AMP pathway
th
Steroid pathway
JAK/STAT pathway
• Growth factor receptors
– PI3K
– MAPK
– IP3
• Seven transmembrane
receptor
– cAMP
– IP3
• Steroid receptor
• Cytokine receptor
27
7-transmembrane receptor:
Chemokines, (nor)epinephrine
glucagon, serotonin, vasopressin,
histamine, calcitonin, rhodopsin,
parathyroid hormone
Growth factor receptors:
EGF, KGF, ILGF, PDGF,
FGF, TGF alpha, VEGF,
c-Kit
PLC
IP3-DAG
Ras (GTP/GDP, Raf)
PI3 K
MKKK, MEK, ERK (MAPK)
PLC
Cytokine receptor:
Interleukins, Interferons,
EPO, G-CSF
Hormone receptor:
Thyroid hormone,
Vitamin D, retinoids
JAK JAK
G proteins-ras
Ca++
cAMP
PKC
PK A
STAT
Steroid
receptor
STAT-P
C t k l t l proteins
Cytoskeletal
t i
Ca++/K+ pumps
Calpain
Ca++ BP-calmodulin
PKB/AKT
Ion channels,
vision, olfactory
Steroid TF
cMyc, cJun, cFos, Foxo3 PPAR
NF kappa B
Cyclins
Cell proliferation
Cell metabolism/differentiation
Inflammatory/immune responses
Abbreviations of signaling molecules
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
AKT—protein kinase B (below)
RAF—(MKKK)
MEK—(MKK)
ERK—(MK)
MAPK—mitogen activated protein kinase
IP3 inositol 1,4,5 triphosphate
PI3—phosphoinositol three kinase
DAG--diacylglycerol
PLC phospholipase C
PLC—phospholipase
PKA—protein kinase A (AKT)
PKB—protein kinase B
PKC—protein kinase C
cAMP—adenocyl 3,5, cylic
monophosphate
JAK—janus activating kinases
STAT—signal transducers and activation
of transcription
Ca++ BP—calcium binding protein
Steroid TF—transcription factor
PPAR—peroxisome proliferation activating
receptor
cMyc, c-Jun, c-Fos—transcription factors
for cell proliferation
•
•
•
•
•
•
•
•
•
•
EGF—epidermal growth factor
KGF—keratinocyte growth factor
ILGF—insulin-like growth factor
PDGF—platelet derived growth factor
FGF-fibroblast growth factor
TGF—transforming growth factor
VEGF V
VEGF—Vascular
l endothelial
d th li l growth
th
factor
C-Kit—stem cell factor
EPO—erythropoietin
G-CSF—Granulocyte-colony
stimulating factor
Ras gene induction of cell proliferation
28
Ras/Rho
• Small GTPases
• RAS subfamily
– H-ras, N-ras, E-ras, r-ras, Rap, Ral, rit, rheb
• Rho subfamily
– Rho A, Rho B, Rho C, Rac 1, Rac 2 Cdc42/G25k
• Ras mediates
– PI-3 pathway
– RAF/MEKK pathway
• These induce transcription factors that increase
transcription of Cyclin D
Transcription factor regulation of
proliferation by Myc
• Myc
– c-Myc, n-Myc, and L-myc
– Leucine zipper transcription factors
– Exacerbate or reduce gene expression
• Rather than turning “on
on or off”
off
– If growth conditions are good, Myc promote proliferation
through:
• Enhanced expression of:
– D and E cyclins
– Cyclin dependent kinase 4 (CDK 4)
• Reduced expression of:
– P21 (CIP1) and p15 (INK 4B)
– If growth conditions are poor, Myc also may enhance
apoptosis (see apoptosis)
Cyclins
• Proteins that complex with cyclin dependent
kinases
• When complexed and phosphorylated the
CDK’s become active serine/threonine
kinases
– CDK’s are expressed constitutively
• Cyclins rise and are degraded by the ubiquitin
pathway rapidly
• Cyclins allow cells to pass through the
Cyclinmajor
A
points of the cell cycle:
CDK 1
– Go-G1-S-G2-M
29
Cell cycle checkpoints
• G1 checkpoint
– Major checkpoint after cell signaling activation
• prevent cells from entering cell synthesis
• Best understood checkpoint
• Synthesis checkpoint
– Reduction
R d ti iin DNA synthesis
th i
• Regulated by ATM
• Cyclin A and E remain inactive
• Least understood checkpoint
• G2 checkpoint
– Inhibitory phosphorylations of cdc2
• Prevents cell proliferation right before chromosomal separation
• Cdc25C is complexed with 14-3-3 and unable to activate cdc2
– Cdc25C is a phosphataste inactivated by 14-3-3
• Partially understood checkpoint
– better than S, less than G1
Cell injury
The Cell Cycle
p53
G0- “inactive”
Cyclin B
CIP/KIP
CDK 1
EGF
M
Cdc25c-14-3-3 Cdc2
FGF
1 hr
G2/M checkpoint
+
G1
IGF
8 hr
Nutrients
TGF B
G2
-
2 hr
G1/S checkpoint
S checkpoint
Cell injury
p53
Cyclin D
Cyclin A
ATM
CDK 1
CDK 4
S
Cyclin A
CDK 2
Cyclin D
8 hr
INK4
Cyclin E
CIP/KIP
CIP/KIP
CDK 6
p53
Cell injury
p53
CDK 2
Ras/nutrient induction of cell proliferation: Getting
cells out of Go
Nutrients
Growth factors
TOR
RAS
S6K
PI3K
S6 phosphorylation
40S
60S
TOR
eIF4E
S6 phosphorylation
40S
60S
Cyclin D
ERK
S6K
eIF4E
Cyclin D
Cyclin D
Cell proliferation
30
Cyclin activation of transcription and passage through
G1/S checkpoint
p130 released allowing E2F activity
p130
Transcription
and passage
through G1/S
checkpoint
DP
p130
ATP
E2F
Cyclin D
DP
CDK 4
Cyclin D
CDK 6
CD kinase activity
Cyclin D
phosphorylates RB
E2F
RB
P
P
P
RB-P
CDK 2
P
P
P
Phosphorylated RB releases E2F and
also opens chromatin structure for
transcription.
P
phosphorylation
RB
denotes retinoblastoma protein
Histone acetylase phosphorylate RB,
decrease E2F and close chromatin structure.
E2F a transcription factor that enhances cell proliferation and passage across the G1/S checkpoint
P130 inactivates E2F and prevents Go cells from undergoing proliferation
DP
a DNA binding protein
Cell activities needed for proliferation and activated by cyclin/CDK’s and
E2F transcription
•
•
•
•
Nuclear envelope breakdown
Centrosome function
Spindle assembly
Chromosome condensation
31
With cell injury: Inhibition of proliferation
to allow repair
• If cells have DNA mutations and
continue replication the mutation is fixed
and p
passed to the daughter
g
cell
• If proliferation is ceased, cells can then
attempt repair
P53: The King.
Guardian of the Genome
p53 activation
CK1
CK2
DNA damage induces activation of:
ATM (ataxia telangiectasia)
ATR (ATM and Rad3-related protein kinases)
DNA-PK (DNA protein kinase)
p53
p53 phosphorylated*
P53 phosphorylation
at a.a. site 20 allows binding
to MDM2 but prevents export
to the cytoplasm where
degradation normally occurs.
Activated p53
ARF
MDM2
Ubiquitylation of
p53 by MDM2 (a
Ubiquitin ligase) and
increased cell proliferation
GADD45alpha
(DNA repair)
p21
Inhibition of cell proliferation
Apoptosis
Repair Senescence
*P53 levels are normally
low in cells and bound
to MDM2 (a ubiquitin ligase).
Phosphorylation of p53 at
site 15, 37 greatly enhances
transcriptional activity.
ARF – induced by c Myc, reduces
MDM-2 mediated degradation of p53,
thereby inhibiting
proliferation and promoting repair or
apoptosis
32
P53 modifications
• Mutations in p53 itself
– 18,000
– Especially occur in the DNA binding portion
• Residues 98-292
• Post-translational modifications
– Phosphorylation
p y
of serine and theronine residues
– Acetylation
• Phosphorylation and acetylation “stabilizes” p53 in
the nucleus
– Can also occur on mutated p53 resulting in nuclear
accumulation
• Upon dephosphorylation and deacetylation p53
binds DNA
– Ubiquitylation, sumoylation (small ubiquitin-like
proteins)
p53 inhibition of cell proliferation: INK4 and CIP/KIP
inhibition of cyclins
• p53 enhances
– INK4 inhibitors
•
•
•
•
INK4a (p16)
INK4b
b (p
(p15)
5)
INK 4c (p18)
INK 4d (p19)
– INK inhibitors:
• Reduce activity of:
– Cyclin D by:
– Prevention of cyclin
D binding to CDK4
• p53 enhances
– CIP/KIP inhibitors
• WAF (p21)
• KIP (p27)
( 27)
• KIP2 (p57)
– CIP/KIP inhibitors:
• Reduce activity of:
– Cyclin D, E, A, B
by:
– Forming
heterodimers with
the cyclin/CDK
complex
Therapeutic strategies to inhibit CDK
•
•
•
•
•
•
Direct inhibitors CDK
Prevention of CDK/cyclin binding
Enhancement of CDK-I
Inhibition of CDK-I degradation
Inhibition of cyclin synthesis
Promotion of cyclin ubiquitization
(degradation)
• Inhibition of CDK activating kinases and
cdc25 phosphatases
• Stimulation of CDK-I activation
33
Markers of cell proliferation
Marker
BrdU
Method of detection
Flow, IHC
Sensitivity
S phase
3H thymidine
radioactivity
S phase
Ploidy
Flow
Other
thymidine analoge
Can’t distinguish
G2 from M phase
Ki-67
Flow, IHC
PCNA
Flow, IHC
all proliferative
cells
broad
Cyclins
Flow, IHC
All phases
PHK2
Flow
Sensitive
AgNOR
Light microscopy
Not precise
labile antigen
stabile antigen, DNA
lesions increase
Used in vet med?
easy
BrdU = bromodeoxyuridine; ploidy = number of chromosomes [aneuploidy, diploidy,
Tetraploidy]; PCNR = proliferating cell nuclear antigen; AgNOR = silver stain of
Nucleolar organizing region
Cellular senescence
• Senescent cells, especially in stem cells, reduces
proliferation and therefore, neoplastic transformation
• With DNA damage
– Lymphoid
y p
cells often undergo
g apoptosis
p p
– Epithelial and mesenchymal cells often undergo
senescence
• Molecular mechanisms of senescence:
– Telomere shortening
• p53 activated and reduces cell proliferation
– p16 (INK 4a)/pRB expressed (in response to
oncogenes such as Ras, BRAF)
• inhibition of cell proliferation
• Without senescence, injured cells proliferate and more easily
contribute to cancer development
34
Neosis
• Some senescence may be “leaky”
– A few cells that are senescent and destined to die by
apoptosis may escape the senescent “mitotic crisis”
• Senescent mitotic block by short telomeres, no telomerase, and of
tumor suppressor genes such p53, pRB and p16 (INK 4a)
• Neosis cells have no telomeres, but p53, pRB and p16 (INK4a) are
inhibited (allowing proliferation)
– These cells with no telomerase may get abnormal chromosomal
joining
– Telomerase may then also become active—resulting in additional
lifespan/growth
– Mutations readily increase
– Escaped cells undergo nuclear budding and cytokinesis
– Escaped cells undergo endomitosis of polyploid DNA, DNA
misrepair, chromatin modulation
• Nucleus forms small buds (karyokinesis) that end up in small
regions of cytoplasm (cytokinesis)
» “Raju cells”
Cell Death and Apoptosis
Response of cells to injury
DNA injury
Dividing cells
Non-dividing cells
Inhibition of proliferation
Apoptosis/death
(Failure of repair)
Cancer
Repair
Mutations
35
Apoptosis
• Occurs physiologically in:
– Embryogenesis, hormone-induced
involution (p
(prostate secondary
y to
castration), breast tissue after lactation,
cell deletion (gut crypts, thymic selectin of
lymphocytes), tumor cells, cytotoxic T cells,
secondary to toxins and viruses
Apoptosis: cellular features
Apoptosis
Cells shrinkage, chromatin condensation,
cytoplasmic blebs, phagocytosis, caspase
activity, transglutaminase activity,
DNA degradation dependent on calcium
and magnesium, DNA ladders, annexin V
expression,
i
phosphatidylserine
h
h tid l i expression
i
on outer membrane leaflet, lack of neutrophil
infiltration
Necrosis
Plasma membrane damage,
dilation of cytocavitary network,
peripheralization of chromatin,
infiltration of neutrophils
Apoptosis
Robbins and Contran Pathologic basis of disease, 7th edition
36
Apoptosis
Robbins and Contran Pathologic basis of disease, 7th edition
Apoptosis: 180-200 bp ladder
A. Control
B. Apoptosis
C. Necrosis
Robbins and Contran Pathologic basis of disease, 7th edition
Apoptotic Thymocytes
Susan Elomore: Toxic Pathol 35:495-516, 2007
37
Cytoplasmic Budding
Susan Elomore: Toxic Pathol 35:495-516, 2007
Tingible Body Macrophage
Susan Elomore: Toxic Pathol 35:495-516, 2007
Apoptosis: initiation/prevention
• Initiators of apoptosis:
– TNF, nitric oxide, fas ligand, granzyme, viral
infection, radiation, corticosteroids, DNA damage
• Extrinsic
E t i i pathway
th
• Intrinsic pathway
• Inhibition of apoptosis:
– Growth factors, differentiation factors, adequate
intracellular nutrition, insulin, others
– Activates “survival signaling pathway”
38
p53 activation
DNA damage induces activation of:
ATM (ataxia telangiectasia)
ATR (ATM and Rad3-related protein kinases)
DNA-PK (DNA protein kinase)
CK1
CK2
p53
p53 phosphorylated*
P53 phosphorylation
at a.a. site 20 allows binding
to MDM2 but prevents export
to the cytoplasm where
degradation normally occurs.
Activated p53
ARF
MDM2
Ubiquitylation of
p53 by MDM2 (a
Ubiquitin ligase) and
increased cell proliferation
GADD45alpha
(DNA repair)
p21
Inhibition of cell proliferation
Apoptosis
*P53 levels are normally
low in cells and bound
to MDM2 (a ubiquitin ligase).
Repair Senescence
Phosphorylation of p53 at
site 15, 37 greatly enhances
transcriptional activity.
ARF – induced by c Myc, reduces
MDM-2 mediated degradation of p53,
thereby inhibiting
proliferation and promoting repair or
apoptosis
Susan Elomore: Toxic Pathol 35:495-516, 2007
Transcription factor regulation of apoptosis
• Myc
– c-Myc, n-Myc, and L-myc
– Leucine zipper transcription factors
– Exacerbate or reduce gene expression
• Rather than turning “on or off”
– If growth conditions are good, Myc promotes
proliferation (see cell proliferation)
– If growth conditions are poor, cells do not achieve
the “survival threshold” and Myc enhances
apoptosis
39
Myc enhancement of apoptosis
• Myc can enhance apoptosis by:
– Activation of the intrinsic death pathway
(Cytochrome C release)
• Through:
– R
Reduced
d
d BCL2 expression
i (BCL2 iis antiapoptotic)
ti
t ti )
– Enhanced PUMA expression (PUMA is proapoptotic)
– Synergy with death receptor signalling
– Generation of ROS
– Increased ARF gene expression
• ARF inhibits MDM2 allowing increased p53 activity,
increased p21 and other proteins that inhibit of cell
proliferation
– Allows p53 to induce apoptosis (above)
Extrinsic/intrinsic apoptosis signaling
• Perforin/Granzyme pathway
– Granzyme A and B (serine proteases)
• B activates Caspase 10, ICAD and cleaves Bid
• A DNA nicking
• Extrinsic signaling
– Ligand/receptors
• TNF alpha/TNFR1, Fas/CD95L(Fas Ligand)),
Apo3L/DR3, Apo2L/DR2, Apo2L/DR5
• Death domains of 80 amino acids
– Activates Procaspase 8 to caspase 8 (active)
– Inhibited by cFLIP which binds adaptor protein FADD and
caspase 8 and Toso which blocks the Fas pathway
• Intrinsic signaling
– DNA damage, UV damage, viral infection, toxins,
hyperthermia, free radicals, cell injury
• ATM/ATR-p53 activation
– Direct activation of BAX
– Puma, Noka inhibition of BCL-2
Apoptosis
TNF alpha
APO-2,3L
Trail receptor 1-4
DR 2,3,5
Cytotoxic T cell expressing
FasL (CD95 Ligand)
TNF receptor 1
Fas (CD95)
DNA damage/cell damage
Procaspase 8
ATM/ATR
Granzyme B
p53
PETN
PI3K
Survival Signaling:
EGF, PDGF, IL-2, 3,
Insulin
PDK1,2 AKT
perforin
Cytotoxic T cell:
Perforin,
Granzyme A, B
Caspase 10, BID
Caspase 8
Caspase 12
perforin
BID
Nucleus
Mitochondria
Caspase 12 Cytochrome C
AIF, CAD,
Endo G
Granzyme A
Initiator caspases:
2, 8, 9, 10, 12
SET
BAD
(binds BCL2)
(AKT phosphorylates
BAD, BAD-P binds
14-3-3 releases BCL-2)
BAX
Caspases 3, 6, 7
“Effector caspases”
Transglutaminase X-link proteins
Cytokeratins, PARP, Gelosin (actin cleavage)
Fodrin, NuMA
14-3-3
Puma,
Noxa
Endoplasmic
reticulum
ICAD
CAD mammalian endonuclease
Endonuclease G
AIF
DNA degradation
Effector caspases:
3, 6, 7
BCL 2
released
BAX (forms pore)
SMAC,
Diablo Apaf-1
ATP
Procaspase 9
Apaf-1 Aven
IAP
Survivin
Caspase 9
Components of the “apoptosome”
ATP
Cytochrome C
Denotes inhibition
Denotes activity
or transition
Denotes movement
Apaf-1
Procaspase 9
40
Caspases
•
Caspases
– Precursor-pro-caspases
• Removal of N-terminal domain during activation
– Caspase 1, 4, 5
• Proteolytic activation of IL-1b, not as active with apoptosis
• Involved with inflammation
– Caspases 2, 3, 6-11, 12, 14
• Cysteine proteases involved with apoptosis
– Caspase 12
• Endoplasmic reticulum-released apoptotic factor initiated by amyloid beta
• Released from endoplasmic reticulum and induces activation of effector caspases
– Caspase 13
• Bovine gene
– Caspase 14
• Embryonic tissues
– Initiator caspases
• 2, 8, 9, 10, 12
– Effector caspases
• 3, 6, 7
•
There is no one key enzyme, protein, or signal responsible for the ultimate death
and lethal blow to a cell.
– In other words, apoptosis factors occur simultaneously
BCL-2 families
• Anti-apoptotic
– Contain all BCL-2 homology (BH) domains, BH1-4
• Includes membrane anchoring, channel formation, and
regulation domains
• BCL2 prevents Bax formation of a pore
– Reduces release of Cytochrome C and reduces apoptosome
formation
– Members:
• Bcl-2, Bcl-xl, Bcl-w, Mcl-1, Boo/Diva
• Pro-apoptotic
– Fewer BH domains
– Members:
• Bax, Bak, Bok/Mtd, Bcl-xs (BH3, BH1, BH2 domains)
• Blk, Bad, Bmf, Bid, Puma, Noxa (BH3 only domains)
– BH3 proteins sense cell damage and act only through BAX and Bak
Uptake of apoptotic cells
• Appears to contribute to killing
– Tingle-body macrophages form
• These are macrophages with abudant
cytoplasm and internalized portions of apoptotic
cells
• Apoptotic
A
t ti cells
ll express
– Phosphatidylcholine (PC) and
phosphatidylserine (PS) on the outer
surface
• PC allows detection of apoptotic cells (“find
me”) by macrophages
• PS allows internalizatoin (“eat me”) by
macrophages
41
a. “Find me”
b. “Eat me”
LPC = lipophosphatidylcholine. PtdSer = Phosphatidylserine, ox LDL = oxidized low density lipoprotein; CD36 = scavenger
Receptor; BAI1 = brain angiogenesis inhibitor; ICAM-3 = intercellular adhesion molecule -3; CD14 = Lipopolysaccharide
binding protein receptor; alpha v beta 3 intergrin; MFGE8 = milk fat globule EGF factor 8; MER = receptor tyrosine kinase;
GAS6 = growth arrest-specific 6;
Nature Rev: Immunol 7:964-973, 2007
Non-apoptotic cell death
• Resistance to apoptosis is a hallmark of
cancer cells
• Defects in non-apoptotic
p p
cell death are
associated with cancer
• Non-apoptotic cell death:
– Senescence
– Necrosis
– Autophagy
– Mitotic catastrophe
Senescence
• From Previous slide during cell cycle lecture:
• Senescent cells, especially stem cells, reduces proliferation and
therefore, neoplastic transformation
• Occurs by:
– Telomere shortening
– p53 activated and reduces cell proliferation
– p16 (INK 4a)/pRB expressed (in response to oncogenes such as Ras,
BRAF)
– inhibition of cell proliferation
• Also, new today:
– Senescent cells have:
• Flattened cytoplasm, increased granularity, changes in metabolism
• Induction of senescence-associated beta galactosidase (SA-beta Gal).
42
Necrosis
• Unregulated cell death with release of
intracellular components
– membrane enlargement (stretching) due to
swelling
– cell swelling
• dilation of cytocavitary network
– vacuoles, nuclear membrane, sER, rER, mitochondria
– fragmentation of the nuclear chromatin
ultrastructurally
– Failure of ion transport, ATP production, and pH
balance
– Inflammation (neutrophils, inflammatory
mediators) and damage of surrounding tissue
Autophagy
•
Type I programmed cell death
•
Type II programmed cell death
– Apoptosis
– Autophagosomes and autophagolysosomes accumulate
• Occurs with
– Growth factor withdrawl
– Differentiation and developmental triggers
» I do not know a specific example
– Massive cell elimination
– When phagocytes do not take up dying cells
–
–
–
–
Caspase-independent
Increased lysosomal activity
Protein translocation to autophagosomes
Signalling involves phosphotidylinositol 3-kinase (PI3K) and target of
rapamycin (TOR)
• Defects in this signaling pathway lead to cancer
• Defects in beclin 1 gene, which works with PI3K to induce autophagy lead to
cancer
Autophagy
• A normal cellular process
• Contributes to cellular homeostasis for turnover of
organelles and superfluous proteins
• Maintains an amino acid p
pool for g
gluconeogenesis
g
and protein synthesis during starvation
• Cell death (for when phagocytes cannot keep up)
• Anti-aging mechanism for removing substances
oxidized by free radicals
• Triggered by nutrient depletion (starvation)
43
Autophagy
• Type I programmed cell death
– Apoptosis
• Type II programmed cell death
– Autophagosomes and autophagolysosomes
accumulate
• Occurs when
– massive cell elimination occurs
– When phagocytes do not take up dying cells
– Autophagsomal activity reduces the cellular “mass” making
type I apoptosis more efficient in developmental cell loss
• Type I and type II programmed cell death are
not mutually exclusive
Cellular degradation of macromolecules and organelles
• Microautophagy
– Degradative material is delivered to the lysosomes by
membrane invaginations of cytoplasm into the
lysosome
• Macroautophagy
– Large organelles (especially mitochondria,
endoplasmic reticulum and peroxisomes) within a
double membrane vesicle that are degraded in
autophagosomes that fuse with lysosomes
• Chaperone-mediated autophagy
– Degradation of specific proteins that are bound to
chaperonens and internalized into the lysosome
• Degraded by calcium-dependent cysteine proteases, calpains,
proteasomes
AVi-initial autophagosomal vacuole, contains a mitochondria and rER
rER around the autophagosome
Degraded material
Partially degraded rER
Kelekar: Ann NY Acad Sci 1066:259-271, 2005
44
Mechanisms of autophagosomal assembly
• LC3-microtubule associated light chain arranges, structurally,
the initial endosome
• LAMP1/2 (lysosome-associated membrane protein) adhere to
the phagosomal membrane as it matures and fuses with
lysosome
• Acid phosphatases and cathespins contribute to degradation
degradation.
• Inhibition of autophagosomes
– Growth factors that activate TOR
– TOR inhibits autophagosome formation
• Induction of autophagosomes
– Nutrient and amino acid starvation
• Activate Beclin-1, p150 and Class III PI-3Kinase
• Also activated ERK ½ and GAIP (which initiates GTP degradation to
GDP); GDP induces autophagosome formation
Kelekar: Ann NY Acad Sci 1066:259-271, 2005
Kelekar: Ann NY Acad Sci 1066:259-271, 2005
45
Autophagy and aging
• Cells with limited mitotic activity
• Accumulate more cell degradation products
(garbage)
– Can reduce cell adaptability, viability, and function
• Cells can undergo apoptosis
– Lipofuscin-is a main “waste” substance
• Derived principally from incompletely degraded
mitochondria
– Mitochondria become damaged with time due to ROS
production that injures mitochondrial DNA, mitochondrial
proteins, and mitochondrial processes like fission/turnover
• Lipofuscin and ceroid become difficult to degrade in
lysosomes/autophagosomes and therefore
accumulates
Autophagy in disease
• Especially important in diseases of non-dividing cells
of the nervous system, muscle or when protein
turnover is critical
– Vacuolar myopathies
• X-linked myopathy, inclusion body myositis, Marinesco-Sjorgren
syndrome
– Danon disease (cardiomyopathy and retardation)
• Deficiency in LAMP-2
– Parkinson’s disease, Huntington’s disease, Alzheimer’s
disease, and spongioform enephalopathies
• Protein aggregates occur in these diseases; autophagosomes
degrade the protein aggregates to a point
Mitotic catastrophe
• Death that can, and hopefully does occur when DNA damage
does not induce cell arrest, DNA repair or apoptosis and G2
check point is defective
– Allows cell to enter mitosis with mutations/defects or prematurely
– Cell death
• Can also occur with microtubule damage and disruption of the
mitotic spindle
• Mitotic giant cell formation/multiple micronuclei
• But if cells in mitotic catastrophe escape death
• Neosis
• See above mitotic giant cells and micronuclei
– These sound like karyo and cytokenesis and Raju cell formation
• Survivin
– Promotes cell proliferation and avoid mitotic catastrophe
• Both of these can contribute to cancer
– Loss of survivin promotes apoptosis
• Inhibition of survivan may promote death and be a good therapy
46
Stem cells and cancer
• Long-term residents
– DNA lesion occur over decades
– Especially lesions in pathways regulating stem cell
renewal
• Wnt and beta catenin, Hedgehog, Notch, Myc and
PTEN
– Stem cells (epithelial) grow in niches that have the
right microenvironmental nourishment for replication
• Symmetrical division
– Two progeny cells in the niche
• Asymmetrical division
– One stem cell in the niche
– One stem cell out of the niche
» This one out of the niche losses it “stemness” and become
differentiated or dies
Genetic variation: SNPs.
Epigenetic events: DNA methylation, HDACs,and polyamines
(telomeres covered by Kuipel)
SNP’s and cancer
• Single nucleotide polymorphisms (SNP’s)
– Single-base variations in DNA between individuals tied
to traits and disease
• Code red hair, freckles, pudginess, love of chocolate
g cancer
• Genetic risk for disease, including
• There are 15 million locations in the genome where one base can
differ between individuals
– 3 million SNPs identified by HapMap
– United Kingdom has heavily investigated:
» Rheumatoid arthritis, bipolar disorder, coronary artery
disease, type 1 diabetes, and Crohn’s disease
• Other genetic variations
– Gene copy number account for 20% of differences in
gene activity; SNP’s account for many of the rest
– Chromosomal inversions, insertions, deletions
47
SNPs and cancer
•
SNPedia
– Website of SNPs, including cancer
– www.snpedia.com
– Visit during class
•
Science “Breakthrough of the Year”
– Human Genetic Variation
– Science 318:1842-1843, 2007
– Discuss during lecture
•
Sequenome
•
Projects
– A company in San Diego that does high-throughput SNP identification
through mass spectroscopy
– SeatlleSNPs Variation Discovery Resource
– Cancer Genome Anatomy Project
• SNP500 Cancer project
– NIH’s Pharmacogenetics Research Network
– International HapMap project
SNP’s
• SNP
– A SNP differs by a single base at a given position
in the genome with a frequency of 1% in at least
one population
– Account for 90% of the total variation in the human
genome
• Other types of genetic variation include:
– Nucleotide repeats, microsatellites, gene amplification,
chromosomal amplification
– Nonsynonymous SNP shifts from one amino acid
to another
– Synonymous SNPs have no amino acid change
SNPs and disease (cancer)
•
SNPs associated with cancer susceptibility
–
N-acetyltransferase 2 (NAT2)
–
Glutathione S Transferase (GSTM1)
•
•
•
–
Myeloperoxidase
–
Also SNPs in
•
•
•
•
SNP in the promoter at G-463A is associated with decreased lung cancer risk in Caucasians
cytochrome P4501A associated with lung cancer risk
TNF and IL-1 associated with diffuse B cell lymphoma
SNPs and cancer outcome
–
•
Slow acetylations increase risk for bladder cancer, especially in smokers
Rapid acetylations increase risk for colon cancer
Homozygous individuals at increased risk for bladder and colorectal adenoma
Cytochrome P450 enzyme CYP3A4 associated with increased long term survival
Pharmacogenetics and cancer therapy SNPs
–
Thiopurine S methytransferase
–
Cytochrome P450 CYP2C
•
•
–
Metabolism of alkylating agents
5, 10 methylenetetrahydrofolate reductase (MTHFR)
•
–
Increased thiopurine toxicity
Increased methotrexate toxicity
UDP-glucurosyltransferase (UGT1A1)
•
Increased irinotecan toxicity
48
Epigenetic inheritance
• Definition:
Cellular information, other than the DNA
sequence itself, that is heritable during
cell division.
• Types of epigenetics information:
•
•
DNA methylation.
Histone Modifications.
DNA methylation
• Occurs in cytosines that precede
guanines
• Dinucleotide CpG.
• 60-90% of all CpGs are methylated in
mammals.
• Unmethylated CpGs are grouped in
clusters called “CpG islands” that are
present in the 5' regulatory regions of
many genes.
DNA methylation and gene expression
• DNA methylation
– Covalent binding of a methyl group to the 5-carbon position of
cytosine/guanine dinucleotides (CpG)
• Termed m5CpG
– 60-90% of cytokine/guanine sites are methylated in repetitive
regions of DNA
– Hypomethylated sites are usually in CpG-rich
CpG rich regions (CpG islands)
near promoter regions
• Often near the core promoter and transcription start site
• Transcriptions occurs if:
–
–
–
–
Transcription factors are present
The CpG island is unmethylated
The chromatin state is “open”
Histones are hyperacetylated (opens chromatin structure; more later)
• Many promoters do lack CpG sites
– By evolution, these were lost due to deamination of m5C and loss
– Methylation of CpG islands reduces transcription
• Inhibits binding of transcription factors
• Promotes binding of methylated DNA binding proteins
49
DNA Methylation Reaction Catalyzed by DNA Methyltransferase (DNMT)
DNA Hypomethylation
Some toxic carcinogens act through
methylation alterations:
• Cadmium inhibits DNMT activity .
• Arsenic induces Ras hypomethylation in
mice.
Dietary relation:
• High dietary methionine increases
methylation leading to low cancer incidence.
DNA Hypomethylation and mechanisms of
tumor formation
Generation of chromosomal instability.
• Hypomethylation causes recombination
and chromosomal rearrangements leading
to deletions and translocations.
• Depletion of DNA methyl transferases
leads to aneuploidy.
50
DNA Hypomethylation and mechanisms of
tumor formation
Genomic Imprinting.
• It is the relative silencing of one parental
allele compared to the other parental allele
• Maintained partly by differentially
methylated regions.
• In cancer, hypomethylation disrupts
imprinting called ‘Loss of imprinting (LOI)’.
eg LOI of IGF-2 causes Wilms tumor.
DNA Hypermethylation in tumors
• Hypermethylation of CpG island in the
promoter regions of tumor suppressor genes.
• Results in silencing of gene.
• First reported in Retinoblastoma tumor
suppressor gene.
• Followed by BRCA1, VHL, p16 INK4a.
• DNA repair gene also silenced further
increasing the chances of cancer.
In normal cells (top) DNA methylation is concentrated in repetitive regions of the
genome and most CpG island promoters are unmethylated. In tumor cells, the
compartmentalization breaks down and repetitive DNA loses methylation while CpG
island promoters acquire it, resulting in silencing of the associated gene.
51
DNA methylation and cancer cells
• Tumor cells often have:
– Global hypomethylation of repetitive DNA
– Region-specific hypermethylation (esp CpG regions)
• G
Global
oba hypomethylation
ypo e y a o
– Leads to chromosomal instability/mutation
• Seen with oncogenes, c-myc, ras
– Results in activation of these genes
– Gene-specific hypomethylation also occurs
• MAGE, S100A4
• Regional hypermethylation of CpG islands
– Seen with tumor suppressor genes
• Results in decreased tumor suppressor gene expression
Inactivation of both alleles of a tumor suppressor gene. One allele can be inactivated by methylation and
The second inactivated by point mutation, methylation or deletion.
Gronbaek K, et al: APMIS 115: 1039-1059, 2007
52
DNA methylation pathway: SAM
SAM-universal methyl donor to DNA, RNA, hormones, neurotransmitters,
membrane lipids, proteins and other molecules
DNA
Methylated DNA
(DNA methyltransferase)
S adenosylhomocysteine (SAH) S adenosylmethionine (SAM)
Adenosine
Homocysteine
THF
Cobalamin (B12)
Methionine
pyridoxine (B6))
riboflavin (B2)
Tetrahydrofolate (THF)
Methyl Tetrahydrofolate (THF)
Vitamins B2, B6 and B12 and
folate (THF) involved above
Diet
Methyltransferases
• 75% of methyl groups from SAM
– Go to the formation of phosphatidylcholine
• 25% of methyl groups from SAM
– Go to methylation of DNA
• SAM:
– Is broken down to gluthathione
– Has antioxidative properties in this regard
DNA methylation
•
Can result in point mutations
•
Hypomethylation
•
Promoter hypermethylation
– Loss of m5C and converstion to T
– Can lead to up-regulation of non-desired genes
• Cellular proliferation, decreased apoptosis
– Reduces tumor suppressor gene activity
• Examples:
–
–
–
–
–
–
Autonomous growth (Ras, SOCS)
Enhanced proliferation (p15, p16)
Reduced apoptosis (DAPK)
Tumor invasion (CDH1, TIMP)
Angiogenesis (THBS1)
Genome instability (MGMT, LMNA, MLH1, CHFR)
•
miRNA methylation
•
Methylation can also be a biomarker (hypermethylation of the above
genes)
53
Histone Modifications
• Histones undergo posttranslational
modifications which alter their
interaction with DNA and nuclear
proteins.
t i
• The H3 and H4 histones have long tails
protruding from the nucleosome which
can be covalently modified at several
places.
• Modifications of the tail include
methylation, acetylation,
phosphorylation ubiquitination etc
Histone Modifications
• Acetylation is associated with
transcriptional activation .
• Effect of histone methylation depends
on the amino acid residue and its
position in the histone tail.
Transcription active
HAT adds Ac
H3L4 trimethylation
Transcription inactive
H3L9 trimethylation
MBD is a protein
that binds m5CpG
Attracts HDAC
which removes Ac
Gronbaek K, et al: APMIS 115: 1039-1059, 2007
54
Histone acetylases and histone deacetylases
• Histone acetyltransferases (HAT)
– Contributes to enhanced transcription of genes
• Adds Ac
• Relaxes the chromatin structure
• Increased transcription
• Histone deacetylases (HDAC)
– Contributes to reduced transcription
p
of g
genes byy
• Ac removal
• Tightens the chromatin structure
• Decreased transcription
HDAC, HAT, and HDAC inhibitors
TF = denotes transcription factor
Kim T-Y Bang Y-J,
Robertson KD:
Epigenetics 1:1 14-23,
2006
Histone acetylases and histone deacetylases
•
Histones
– 146 nucleotide wrap around histones = nucleosome
– H1 not involved with acetylation
– H2A, H2B, H3, H4
• Lysine-rich tails are acetylated by HATs which removes the negative charge of lysine and
allows relaxation of interaction between negative histones and the positive DNA
– Histone acetyltransferases (HATs)
• Acetylate lysine residues of histones
– This enhances transcription
– Transcription also enhanced by methylation of lysine 9 of histone 3
• Acetylation also of E2F (key to cell proliferation),
proliferation) p53
p53, GATA
– Regulates transcription of genes for cell proliferation
– Increase cell arrest, apoptosis, and cell differentitation = decreased cancer
– Histone deacetylases (HDACs)
• Deacetylate lysine residues on histones
• Transcription reduced by methylation of lysine 4 of histone 3
– May allow inhibition of tumor suppressor gene
• Repress (decrease) gene transcription by removing the charge-neutral acetyl groups
– Enhances gene transcription of other genes
– Histone deacetylase inhibitors (HDAC-I)
• Allows acetylation and thus gene transcription
• Resultingly acts like HATs to decrease cell cycling (cell arrest), increasing apoptosis, and
differentiation thus decreasing cancer
• Later this semester: Cancer therapies; HDAC-I
55
HDAC inhibitors (HDACi) and transcription
• Inhibition of HDAC (with HDACi) would conceiveably
increase transcription of all genes
– Due to increased acetylation and relaxed chromatin
structure
– For cancer therapy, could up regulate expression of a
tumor suppressor gene
• 20% of all genes are affected by HDACs
• In fact, acetylation increases or decreases transcription
– The ratio of up-regulated to down-regulated genes is 1:1
• Therefore, up and down regulation is equal
• The HDACis may
– Increase tumor suppressor gene expression
– inhibit cell repair of neoplastic cells and also affect
proliferation rates
• http://www.methylgene.com/HDAC_animat.swf
Acetylation effects on the cell
•
Acetylation occurs in:
– Histones, as discussed
• FYI, proteins can be acetylated, methylated, ubiquitinated, phosphorylated, ADPribosylated and deiminated.
– Transcription factors
• The acetylation occurs on lysine which is also the site of ubiquitin
– Therefore, can affect degradation
– Importin
• Import export of proteins across the nuclear envelop
– p53
• Regulating cell proliferation, repair, apoptosis
– HSP90
• Protein stability
– STAT3
• Cytokine signaling
– Microtubules
• Cell structure
– Ku70
• Allows Bax to be free
– Apoptosis
– HMGB1
• A cytokine-like protein involved with inflammation, fever, and nausea
Protein regulation by chaperone proteins
• Chaperones proteins:
– Constitutively expressed in normal cells
•
(1-2% of total cellular protein content)
– Function as complexes with adaptor
molecules and co-chaperone proteins
– Do not covalently modify the substrate or
“client molecules” on which they act
• Interact with client proteins via cyclical binding
pockets
56
HSP 90
HSP 70
HSP 70
HSP 60
HSP 10
Chaperones are required for essential housekeeping functions
• Chaperones mediate
– de novo protein folding during polypeptide-chain
synthesis
– translocation of protein across membranes
– Quality control in the endoplasmic reticulum
– Normal protein turnover
– Post-translational regulation of signaling molecules
– Assembly/disassembly of transcriptional complexes
– Processing of immunogenic peptides by the immune
system
57
a. Prevents aggregation
b. Intracellular trafficking
c. Maintains proteins in stable state for alterations such as ligand binding,
phosphorylation or multi-subunit assembly
d. Target proteins for degradation
Chaperones as Heat Shock Proteins (HSP)
• Upregulated in conditions of cellular stress
–
–
–
–
–
Heat
Hypoxia
Cellular Starvation
Radiation Exposure
Exposure to chemical mutagens
• Upregulated in tumor cells
– General hypoxic environment
– Rapid cell proliferation
– Function as biochemical buffers for extensive genetic
heterogeneity that is characteristic in tumors
Nature Reviews Cancer 5, 761-772 (2005); doi:10.1038/nrc1716
HSP90 AND THE CHAPERONING OF CANCER
58
Chaperone Proteins act on client proteins as complexes
Drug Discovery Today Vol.9, No. 20 October 2004
ATP driven conformational changes is needed for
chaperone/client protein interactions
Stryer, Biochemistry, Fourth Edition
HSP upregulation in tumor cells
• HSP90
– Maintaining cellular signaling protein stability (hormone
receptors and protein kinases)
• HSP70
– Stability of multi-protein complexes
• ER chaperones
– Folding and maturation of immunoglobulins and MHC
class I molecules
– Calreticulin, calnexin, tapaisin, protein disulfide
isomerase
59
Inhibiting HSP to treat cancer
• Several experimental protocols are being used to target
the chaperone proteins that are highly abundant in
tumor cells and use them as therapeutic targets
• HSP70 and HSP90 are the most abundant chaperone
proteins identified in tumor cells,
cells and the focus of most
research
• Many oncogene protein products are “clients” of HSP90
– ErbB2, EGFR, Bcr-Abl tyrosine kinase, Met tyrosine kinase, cRAf, b-Raf, androgen and estrogen receptors, HIF alpha and
telomerase
• Inhibition of HSP90 leads to degradation of the above
oncoproteins
Drug Inhibition of HSP90
RNA
60
• Much of the genome codes for mRNA
– Some of this is spliced out as introns
• 0.5% of the genome codes for rRNA
• 0.2% of the genome codes for tRNA
• 0.0?% codes for miRNA
– Thus, most of the supercoiled genome codes for mRNA
• A lot of the mRNA genes are not heavily transcribed
• Whereas,
Whereas rRNA and tRNA genes are often repeatedly
transcribed
• Of the transcription activity in a cell
• mRNA > rRNA > tRNA
– Of the transcripts that result in actual proteins:
• rRNA > tRNA > mRNA
• Thus much of mRNA is modified, spliced, or unstable and
does not result in protein in comparison to r and t RNA
– r and t RNA are more “routine” RNA’s and proteins
RNA polymerases
miRNA (more later)
DNA
RNA
Polymerase I
39%
DNA polymerases
Pre-RNA:
Pre-ribosomal RNA
RNA
Polymerase III
3%
snRNA Pre-messenger RNA (hn RNA)
1. Modification
(Sno RNA, nucleolus)
2 Processing
2.
(Sno RNA)
3. Self splicing
(group I intron)
RNA:
Gm7
5.8S
RNAase P Pre-transfer RNA
1. Modification
(poly A tail and 5’ cap)
2 Splicing
2.
(sn RNA)
3. Editing
(var.; gRNA)
4. Transport
5. Stability (ds RNA)
1. Modification
(base, ribose)
2 Processing
2.
(ribozyme)
3. Splicing
(enzymatic)
4. Editing
(enzymatic)
mRNA
rRNA
18S
DNA synthesis and
cell replication
RNA
Polymerase II
58%
tRNA
A (200)
28S
1%
17%
79%
Protein
RNA nomenclature
Pre-rRNA: precursor of ribosomal RNAs, 5.8s, 18s, 28s
Pre-mRNA: precursor of mRNAs, contains exons and introns
Pre-tRNA: precursor of tRNA, contains several tRNA
CAP:
Modified guanosine at the 5’ end of all RNA polymerase transcripts
A (200)
Polyadenylated tail of mRNA
RNAase P:
sn RNA:
catalytic (ribozyme) that processes the 5’ end of tRNA
small nuclear RNA, U1-U6, generally U-rich, pre-mRNA splicing
and pre-rRNA processing
sno RNA:
small nucleolar RNA, 200 different types involved with rRNA
modification and processing
Group I intron: self splicing RNA sequence in 28s ribosomal RNA
RNA modifications: RNA stability (cap, poly A tail), formation of tertiary structure,
interaction with RNA proteins (more later)
Transcription of rRNA and tRNA as precursors: ensures equal amounts of RNA
Pre-mRNA splicing: enlarges protein repertoire (more than alternative splicing), it has
evolutionary advantages
61
RNA polymerase II holoenzyme for mRNA generation
DNA arranged in a loop-like structure for
RNA polymerase II transcription
RTF
RTF
Adaptor
80
60
40
RTF-regulatory
transcription factors
250
30a
110
30b
RNA-Pol II
150
TBP
+TFIIB
+TFIIE
+TFIIF
+TFIIH
Holoenzyme
General transcription factors (GTF): TFIID, TFIIB, E, F, H
mRNA stability and transport after synthesis
•
•
•
mRNP (mRNA proteins) are adaptors of mRNA to allow it to
interface intracellular machinery for subcellular location, translation,
decay with miRNA, and signal transduction
– Some mRNP components are activators/others repressors
Most mRNP components (proteins and miRNA for degradation)
bind specific recognition elements in the untranslated 5 prime or 3
primer regions.
A few mRNP components (proteins) target:
– 7 methyguanosine cap, five prime end (CBC20/80)
– Poly A tail at three prime end
• PolyA binding protein nuclear
• PolyA binging protein cytoplasmic (eIF4E, PABP)
– 5’ end for transcription (eIF4G)
– Y box proteins
• Packing proteins along the length of mRNA
– EJC (exon junction complexes)
mRNA stability and transport after synthesis
• mRNA export adaptor proteins
– Target mRNA to nuclear pore for exit
– Exportin 5
62
mRNA stability and transport after synthesis
• mRNA export and fates in the
cytoplasm
– Exported by five prime end and
immediately engaged with ribosome
– Exported by non-five prime end, not
engaged with ribosome
• Transported to specific sites of cytoskeleton
• Co-localized with other mRNA
– Allows close interaction of protein subunits shortly
after translation
– Stored translationally silent
• eIF4E, CPEB, and EJC attached
mRNA moves to a cytoplasmic
site for translation; co-localizes
with other mRNA; promotes
assembly of protein units
NUCLEUS
--mRNA exported non- 5’ first
CYTOPLASM
mRNA circular and
translationally silent
mRNA strand
Nuclear pore
eIF4G
EJC (exons)
mRNA export
adaptor for
nuclear pore
Poly A binding
proteins eIF4E,
PABP; also CPEB,
EJC
--mRNA exported 5’ first and translated
Poly A binding
protein
CBC20/80
ribosome
mRNA degradation
• mRNA degradation
–
–
–
–
Polyadenylases degrade Poly A tail
5 prime cap removed by decapping enzymes
RNA body
y degraded
g
by
y exonucleases
mRNA endonuclease cleavage
• Sequence specific
• occurs by RISC in association with miRNA
– Abberent mRNA’s
• Premature translation stop signal (PTC’s discussed last week)
• Lack a translation signal (nonstop RNA)
– These occur by mutation, missplicing, premature polyadenylation
• More comments on post-transcriptional regulation
later
• Stress cells and mRNA degradation (next slide)
63
mRNA stability and transport after synthesis
• P bodies and Stress Granules
• P bodies (PB):
– Cytoplasmic processing bodies that form around
aggregates of RNP not involved in translation
– These RNP
RNP’s
s are targeted for PB’s
PB s by miRNA/RISC
• Stress granules
– Retirement homes
• Under stress, there is global translation arrest of “housekeeping”
gene transcripts
• Stress granules are composed of:
– Inactive mRNP’s, 40S ribosomal units, mRNA binding proteins
TIA-1 and TIAR
» TIA-1 and TIAR have prion like domains that self oligomerize
and promote assembly
– When stress relieved, stress granules disassemble
More on mRNA post transcriptional processing and it’s effect on
RNA stability
• mRNA structures that affect post
transcriptional processing
–5
5’ cap
– 5’ untranslated region (5’ UTR)
– Open reading frame (ORF)
– 3’ untranslated region (3’ UTR)
– 3’ poly adenine tail
RNA post-transcriptional modifications for stability or degradation
• 5’ Cap
– Methylation of cap regulates expression
– Cap prevents degradation
• Increases stability of mRNA
• 5’ UTR controls mRNA stability by
– It’s ribosomal entry site
– Actions on the 5’ UTR by upstream ORF
– 5’ polypyrimidine sequences
– RNA secondary structure
64
RNA post-transcriptional modifications for stability or degradation
• ORF in mRNA regulate post transcriptional mRNA
stability and activity by
– Unusual codons
• These can slow translation
• 3
3’ UTRs affected by
– RNA binding proteins (RNA BP)
• Alter translation (can increase or decrease translation)
• Tag mRNA for degradation
– Decreased mRNA stabilithy
• Protect UTR from nucleases
– Increased mRNA stability
– RNA BP (CBC20/80, eIF4G, eIF4E, PABP) altered by
• Mutations of the RNA BP gene
• Increased UTR target sequences for the RNA BP to bind
• Decreased UTR target sequences for RNA BP binding
RNA post-transcriptional modifications for stability or degradation
• Poly AAAAA tails
– Prevent degradation
– Long tails have efficient translation
– Short tails inefficient translation
RNA post-transcriptional modifications for stability or degradation
• Signaling affects on stability
– MAPKK
– Increase RNA BP
• PABP-1 and TTP
• These reduce deadenylation of the poly AAAA
tail
• This increases stability and increases
translation
65
RNA silencing
• Small RNA’s from inside or outside the
cells are processed by iRNA machinery
to inhibit g
genes and p
proteins by:
y
– Cleaving mRNA’s
• siRNA fully complementary to mRNA
– Blocking protein synthesis
• miRNA partially matched with mRNA
– Inhibiting transcription (RITS)
• Complex enters nucleus and inhibits chromatin
mi and siRNAs
• iRNA’s
– Term for microRNA (miRNA), small
inhibitor RNA ((siRNA)) and intermediates
– 20-26 nucleotides in length
– Also: repeat-associated small interfering
RNA’s (rasiRNA)
miRNA
– MicroRNAs (miRNAs)
– RNA polymerase II makes miRNA
•
•
•
•
•
•
•
miRNA are double stranded
21-25 nucleotides long
Derived from short hair-pin precursors
1000 miRNA genes (non-protein encoding) currently known
400 different types
30% of human genes regulated by miRNA
Coded in introns from mRNA or snoRNA (small nucleolar RNA [sno RNA,
a.k.a.: ribosomal RNA])
– miRNAs are pieces of mRNA from the non-protein coding regions
• Pri-miRNA’s (folded)
– Come off RNA polymerase II with mRNA and bind Drosha and Pasha
– Double stranded (due to folding)
• Pre-miRNA
–
–
–
–
Formed after Drosha and Pasha cleavage
Bound to exportin, transported to cytoplasm
Cleaved by dicer
Double stranded
66
CYTOPLASM
NUCLEUS
RNA poly II
miRNA gene
Pri-miRNA
Pre-miRNA
(Tails released by Drosha)
Dicer
miRNA
• Dicer protein/enzyme
– Converts pre-miRNA to mi RNA by
cleaving
g off the p
pre-miRNA loop
p end
– Then eliminates one RNA strand
• 5 prime end preserved and enters RISC
complex
– Less stable, unwinds more easily
siRNA
• Short-intefering or silencing RNA
– Long ds RNA and mi RNA
• si RNA sources of formation:
– Endogenous
• Not fully defined
– By synthesis
– By some viruses
• Delivered for therapy by:
– Gene (viral) vectors
– Synthetic delivery
67
Wikipedia.com
si and mi RNA activity on gene silencing
Long ds RNA
micro ds RNA
Dicer
R2D2
ATP
Occurs spontaneously
Occurs with viral infection
Can occur experimentally
Encoded in the genome
Single stranded si RNA
RISC is composed of:
Argonaut protein and ss RNA
Argonaut proteins
Enters nucleus and inhibits
transcription
mRNA degradation if si RNA
is fully complementary
Inhibition of translation/protein synthesis if mRNA
is not fully (only partially) complementary
RISC
• RISC complex
– RNA induced silencing complex (RISC)
• Composed of:
– Argonaut
A
t protein
t i and
d smallll single
i l strand
t d off RNA
• Mediates:
– mRNA cleavage
» si RNA fully complementary
– Protein synthesis inhibition
» mi RNA partially mismatched
– Transcriptional gene silencing
» RITS
» A complex that enters the nucleus and affects
chromatin
68
•
mRNA cleavage by RISC
– RISC (siRNA and argonaut protein) plus mRNA
• siRNA is sequence specific to that mRNA
• mRNA is destroyed
•
Blockage of protein synthesis
– RISC ((mi RNA and argonaut
g
protein plus mRNA))
• mi RNA is only partially matched with mRNA
• mRNA unable to enter ribosome for translation
•
Transcriptional gene silencing
– RITS complex (RNA-induced transcriptional silencing (RITS) complex)
• Composed of:
– RISC (si RNA and argonaut protein)
– Chp1 and Tas 3 proteins
• RITS transported to the nucleus
• siRNA within the RITS
– Matches up with chromatin nucleotides
– Also binds methylated lysine 9 histones
• RITS/chromatin complex enlarges (other proteins bind)
– TRANSCRIPTION (RNA polymerase binding) prevented
How do iRNAs affect cancer?
•
miRNAs can enhance or repress mRNA translation
– miR369-3 stimulates translation during arrested cells (non-proliferating)
– miR369-3 reduces translation in dividing cells
• Science 318:1877, 2007
•
Improper activation of mRNA regulation in cancerous cells
– Oncogenic miRNAs (oncomiRNAs)
• miR-155 enhances cell proliferation
• miR17-92
iR17 92 reduces
d
c-myc apoptosis
t i actions
ti
• miR-21 inhibits TPM and apoptosis
• miRNAs may alter transcription factors, enhance
oncogenes, reduces tumor suppressor genes, and
increase angiogenesis in tumors
• American J Pathol 171:728, 2007
• Some miRNAs inhibit cancer
– miR-15 and miR-16 inhibit BCL-2 allowing apoptosis
– Let-7a inhibits RAS
iRNA as a therapy
•
Therapy
– siRNA delivery
• High pressure tail vein
– Recent work used low volume and normal pressure
– Problem: excretion
» There is rapid excretion
» Degradation
g
is less of an issue
• Vector delivery
– Retrovirus, adenovirus, adeno-associated virus (AAV) with various promotors
» Problems with non-specific delivery to unwanted sites, interferon reactions to the
hairpin RNA
• Benefit
– Knocking down genes with precision
» Other genes unaffected
– siRNAs are more potent and longer-lasting than oligonucleotides (antisense DNA) and
ribozymes
» siRNAs have a greater potency in turning off genes
• Hurdles:
– Delivery to the correct neoplastic cell (“either” deleted)
» This is true for systemic delivery or vector delivery
– Continued activity over time (repeated administration needed possibly)
69
Chronic cell injury caused by intracellular
accumulations of metals, bilirubin
Ackermann
Lipid accumulation
Protein accumulation
Lysosomal storage diseases
Iron, copper, bilirubin, porphyria
accumulation
Robbins and Contran Pathologic basis of disease, 7th edition
Other: hyperglycosylation
Intracellular accumulation of metals
• Iron (Fe++) and Copper (Cu++)
• Accumulation of one or both results in cellular
degeneration
– Commonly hepatic degeneration/cirrhosis
– Mechanism of toxicity:
• Induction of free radical formation
– Fenton reaction
– Occurs in the region of the ion
This region is often an important site for enzymatic
activity, protein structure/conformation, signaling
activity
• Absorption and regulation
– Iron body stores are regulated at the level of
absorption by enterocytes (intestine)
– Copper body stores are regulated at the level of
excretion liver (excretion into bile)
70
Iron absorption
Intestinal lumen
Exfoliation of enterocyte
Site of enterocyte exfoliation
Intestinal wall
Intestinal enterocytes
Intestinal lumen
Intestinal villi
Intestinal crypt
Intestinal villi-higher magnification
Iron absorption
Intestinal lumen containing dietary Iron (10-20 mg in diet/day);
Only 1-2 mg required for absorption
Fe++
Fe++
DMT-1 (divalent metal transporter)
Enterocyte
Labile iron
(Ferritin)
Nucleus
Hepicidin-ferroportin complex degraded
In lysosome-leads to reduced iron absorption
Ferroportin-produced in enterocyte
transferrin protein-carries absorbed iron (1-2 mg/day)
Hepicidin-produced in liver
Key iron proteins
•
Transferrin
•
Ferritin
•
DMT 1
•
Ferroportin
•
Hepcidin
– Carries iron in blood
– Stores iron in cell
– Transfers iron across the cell
– Efflux of iron out of cell into the blood
• The only efflux mechanism for iron
– Binds ferroportin and induces degradation
•
Hemojuvelin
•
Iron regulatory protein 1 and 2
– Increases hepcidin synthesis
– Bind iron regulatory elements (IRE 3’ and 5’)
– IRE 3’ increases transcription of transferrin, ferritin, DMT1 and ferroportin
– IRE5’ decreases transcription
71
Iron sources:
From duodenum
Scenescent rbcs
Liver cells
Syncyiotrophoblasts
y y
p
for passage to fetus
Fig. 4. Major iron flows are controlled by hepcidin–ferroportin interactions. Hepcidin
Blocks outflow of iron from duodenum, liver, macrophages (and placenta)
Hepcidin
• Increased hepcidin
– Decreased ferroportin
– Decreased iron absorption
– Hepcidin
p
increased with inflammation
• Anemia of chronic inflammation
• Keeps iron from microbes
• Decreased hepcidin
– Increased ferroportin
– Increased iron absorption accumulation
– Hemochromatosis
• Hepatocellular damage and cirrhosis
Iron accumulation
• Normally
– 1-2 mg/iron/day absorbed
– Transferrin 30% saturated
• If the body senses enough iron
– Then transferrin does not pick up iron from the
enterocyte
– The enterocyte exfoliates
• Iron stays in feces
• With Hereditary hemochromatosis
– 3-4 mg/iron/day absorbed
– Transferrin 100% saturated
72
Robbins and Contran Pathologic basis of disease, 7th Edition, 2005
Causes of excessive iron accumulation
• Primary
– Hereditary hemochromatosis (adult and juvenile forms)
• Juvenile form most severe
– Man, Minah Birds, Birds of Paradise, Lampreys, Rock
hydraxes
• Secondary to another disease
– Chronic liver disease
• Alcoholic cirrhosis
cirrhosis, chronic viral hepatitis
hepatitis, shunts
shunts,
porphyria
– Congenital atransferrinemia
– Ineffective erythropoiesis (dyserythropoiesis; iron absorbed
but not used)
• Beta thallasemia, aplastic anemia, pyruvate kinase deficiency
(Basenji dogs)
– Idiopathic excessive parental iron injection
• RBC transfusions, iron-dextran injection (sideroblastic
anemia)
– long term dialysis
Treatment for iron storage
• Hemochromatosis
– Blood letting
– Hepcidin
• Costly
• Thalassemias
– Hepcidin?
73
Copper absorption
Intestinal lumen containing dietary copper (1.5-4 mg/Cu++/day)
Cu++
Enterocyte
Liver
Hepatocyte
Hepatoc
te
Cu++ albumin
Cu++ ceruloplasminSystemic delivery
metallothionen
Hepatocyte- copper bound to
metallothionen and
If excess in lysosomes
lysosome
Protein for copper
excretion into bile
Biliary excretion- copper is excreted into bile and eliminated (2-4 mg/day)
Cu passes across the membrane with Ctlr protein, stored with MT or shuttled to the Golgi apparatus by HAH1 and
uses a Cu atpase (ATP7b (WND) which is absent In Wilson’s disease) to pass across the membrane;
Ccs proteins shuttles to SOD; Cox 17 shuttles to the mitochondria for its role with cytochrome C oxidase.
Ceruloplasmin (Cp transfers copper to the plasma)
Copper accumulation
• Primary
– Wilson’s disease
• Defective WND
– Amino acid defect at aa # 1069, commonly
» Impairs biliary excretion of bile by inhibiting
copper transport into the golgi
Ceruloplasmin
Serum copper
Urinary copper
Liver copper
Wilson’s disease
0-200
19-64
100-1000
>250
Normal
200-350
70-152
<40
20-50
74
Copper accumulation
• Primary
– Wilson’s disease (previous slide)
– Bedlingham terriers (BT)
• Primary accumulation of copper
– Metallothionen of BT remains neonatal-like
• Secondary
• In other breeds, copper may accumulate in liver
secondary to bile stasis following liver damage
BT
10000
Cu
In
liver
Dalmation
7500
Doberman
5000
Skye Terrier
Cirrhosis
3000
WHWT
2000
1000
500
250
0
Copper accumulation in sheep
• Sheep on inbalanced diets can store
excessive copper in liver
– Sudden release is often seen
• Copper damages hemoglobin
– Free radical formation?
• RBC’s lyse
– Lost of deformability
– Heinz body formation
Metallothionien: Intracellular regulation of
divalent cations
• Metallothionien
– Has 20 cysteine residues
• SH groups can bind metals
– Capable of binding (or releasing)
• 12 copper ions
• 7 zinc ions
• 20 nitric oxide ions
– MT binds physiological (Zn, Cu, Fe, Se) and xenobiotic (Cd, Hg,
Ag) metals
• Zinc
– Vital for activity of some enzymes
– Vital for inactivation of some enzymes
– Vital for activation of binding of some DNA transcription factors
75
Metallothionein regulation by zinc and anti-oxidative activity
Cytokines
Glucocorticoid
STAT
GRE
Methylation
H2O2
Electrophiles
MRE
ARE
:Up regulation
:Down regulation
Structural gene
MTF-1
Apo MT
Apo-MT
Zn pool
Zinc Finger Protein
Inactive Protein
Zn7-MT
Apo-MT
Oxidative Stress
Degeneration
Overview of metallothionein (MT) gene regulation
The MT promoter has many elements that up-regulate transcription. These include the following: 1)metal
response elements (MRE), which are activated by the metal-responsive transcription factor (MTF-1) after zinc
occupancy; 2) glucocorticoid response elements (GRE); 3) elements activated by STAT (signal transducers
and activators of transcription) proteins through cytokine signaling, and 4) the antioxidant (or electrophile)
response elements (ARE) activated in response to redox status. Davis and Cousins, 2000.
Bilirubin damage to cells
•
Unconjugated bilirubin (UCB)
–
–
Formed by catabolism of heme by splenic macrophages
not water soluble
–
UCB is conjugated with glucuronide by hepatocytes
•
•
•
–
Therefore, transported by albumin
Increases water solubility
Most other cells lack the ability to conjugate bilirubin
Hyperbilirubinemia in infants can occur with:
•
•
Inherited deficiencies of glucuronidation enzymes
Delayed development of glucuronidation enzymes
•
“Bili lights” UV light exposure in infants with elevated bilirubin levels
–
–
Premature birth
Bilirubin levels in serum increase with red blood cell lysis, bile stasis
•
•
UCB readily enters cells
UCB is antioxidant at modest elevations
•
UCB can induce injury at high levels
–
Absorbs electrons
–
Neurons
–
Astrocytes-
•
•
•
Apoptosis (next slide)
UCB diffuses into the cell and alters mitochondrial membranes allowing Cytochrome C release
reduced glutamate uptake
–
Prolonged glutamate at the synaptic cleft
» Increased excitotoxicity (overstimulation of the NMDA receptors)
» Na+, Ca++, Cl-, water influx
» Apoptosis/necrosis of neurons
Bilirubin neurotoxicity
Ostrow JD, et al Trends in Mol Med 10 (2):65-69, 2004
76
Regulation of UCB levels in CNS tissue
• UCB transportor across the blood brain barrier and choroid
plexus
• Protective
– Organic anion transport proteins (OATP)
• Transports UCB two directions
– Into cells and into blood
– Multidrug resistance receptors (p
(p-glycoprotein)
glycoprotein)
• Transports UCB one direction
– Into blood in BBB
– Into blood and into CSF in choriod plexus
» Could increased CSF UCB transport be detrimental?
• These pumps likely explain why UCB accumulates in specific
regions of brain
– E.g. UCB remains in those regions lacking active pumps
– CSF accumulation?
• May allow increased levels around CSF
UCB transporters
Ostrow JD, et al Trends in Mol Med 10 (2):65-69, 2004
Effects of lipids and sugars on cellular
degeneration
Ackermann
77
Factors that mobilize fat
• Intense lactation
• Pregnancy toxemia
• cattle, sheep, guinea pigs
•
•
•
•
•
•
•
•
•
Hyperlipidemia, ponies; hepatic lipidosis, cats
Starvation
Diabetes, hypothyroidism, Hyperadrenalcortisim
Alcoholism
Nephrotic syndrome
Choline deficiency
Anemia
Fatal fasting syndrome, old world primates
Hyperlipidemia diseases (more later); Schnauzers
Robbins and Contran Pathologic basis of disease, 7th Edition, 2005
Cellular energy: required for ATP
Glucose/glycogen
Proteins
Pyruvate
Fatty acid
Fatty Acyl Co A
ACETYL CO A
KREBS (produces NADH, FADH2 for Oxidative phosphorylation)
OXIDATIVE PHOSPHORYLATION
+ NADH
+02
=ATP
Mitochondria
ATP for:
ATPase pumps, signaling, etc.
A transporter is required for:
Fatty Acyl Co A, pyruvate and ATP
Cell plasma membrane
78
Fats and cholesterol
• Are in the blood (as insoluble fatty forms) at
low amounts:
• Triglycerides, free fatty acids, glycerol (draw glycerol,
mono di
mono,
di, and triglyceride)
• Cholesterol, cholesterol esters
• Phospholipids (draw; phosphatidylcholine)
• Vitamins A, D, E, and K
• Also transported as lipoproteins (LDL, VLDL,
etc.) that contain apolipoproteins (Apo E, B,
etc.)
Lipid transport in blood
• Lipoproteins
•
•
•
•
Chylomicron, LDL, VLDL, HDL, IDL
Center is lipid part
Surface is protein part
Classified by molecular size, electrophoretic mobility, and
densities
• Density:
– Chylomicron (least dense), then VLDL, LDL, HDL (most
dense)
– Decreases with triglcyerides
– Increases with protein and phospholipid content
» Chylomicron has 83% triglyceride/2% protein
» HDL has 8% triglyceride/33% protein
Apolipoproteins
• Bind lipids and become lipoproteins. Function in LDL’s,
VLDL’s, chylmicrons, HDL’s, IDL’s.
– If they are free of lipid, then called apoplipproteins
• Serve as ligands for receptors
• Apolipoprotein A
– Major amount on: HDL’s
• Apolipoproteins B
– Major amounts on: chylomicrons, VLDL and LDL’s
• Apolipoprotein C
– Major amounts on: chylomicrons, VLDL, HDL
• Apolipoprotein E
– Major amounts on: VLDL and HDL, some on chylomicron
– Three forms differ by one amino acid
» E3 is the common form
» E2 is associated with high triglycerides/cholesterol
» E4 is associated with high cholesterol
79
B-48
E, C transfer
Apo E, A, C
HDL3
LCAT
HDL2
Lipoprotein
lipase
(Triglyceride release)
Cholesterol
75% of LDL
25%
Other tissues
Macrophage R:
LDL R
SR-A
CD36
•
•
•
•
•
•
•
1%
Robbins and Contran Pathologic basis of disease, 7th edition
Lipoproteins
Chylomicron
– B 48, picks up E and C
VLDL synthesized in hepatocyte
• B 100, E and C
• Dog also has B48
Nascent VLDL receives Apo E and C2 from HDL
• This mature VLDL transports lipids
LDL
• B 100
• B 100 B 48 in dogs
HDL1
• E, A, C
• Dogs only; no HDL1 in man
HDL2
• E, A, C
• Dogs and man
HDL3
• E, A, C
• Dogs and man
Lipoprotein and hepatic lipase
• Lipoprotein lipase
• Tissue capillaries
• Activated by Apo C2
• Hydrolyzes triglycerides from VLDL
• Hepatic lipase
• Removes triglycerides in liver
80
Contents of LDL’s
•
•
•
•
•
Apolipoproteins and cholesterol
Phosphatidylcholine (Phospholipid)
Lysophophatidylcholine
Sphingomyelin
Fatty acids
• Linoleic acid, palmitic acid, stearic acid, free
fatty acids, triglycerides and trans fatty acids
Oxidation/acetylation of LDL’s
– Increases scavenger uptake by foam cells
• 3-10 fold increase
• Non-oxidized/acetylated (native) LDL’s are taken up by
LDLR on hepatocyte and macrophage
» Macrophage uptake of native LDL is slow and
downregulated
– LDL’s “trapped”
pp
byy proteoglycans
p
gy
– Lipoperoxidation of phospholipids, fatty acids, and
Apo B
• Occurs by lipoxygenases, MPO, iNOS, ROS
– Self propagation of the reaction
– Decomposition to of fats to aldehydes, ketones
– Apo B
• Lysine residue oxidized
• This inhibits binding of ApoB to LDLR
Foam cell formation
• Acetylated LDL’s
• Rapidly taken up by macrophage LDLR (SRA-scavenger
receptor A)
– This receptor not down regulated by the cell
• Oxidized LDL’s
• Rapidly taken up by AGE receptors LDLR (SRA), CD36
and
d other
th receptors
t
– These receptors not down regulated by the cell
• AGE receptors take up oxLDL’s and AGE
• AGE (Advanced glycosylation endproducts)
» RAGE (receptor for AGE), 80 K-H, OST-48, galectin-3,
macrophage scavenger receptors type I and II (SR-A)
» CD36 is a class B scavenger receptor, but like SR-A,
takes up oxidized LDL’s and AGE
81
Foam cell
Endothelial cells, smooth muscle
cells, or macrophages, (or Cu++)
Acetyl LDL
Native LDL
Fast
ox LDL
slow
Fast
Native LDL R
(down regulated)
Acetylated LDL R (SR-A)
CD36, SR-A and
(not down regulated )
Other ox LDL receptors
(not down regulated)
Steinberg: Nature medicine 8:1214, 2002
Foam cells fight back: Mechanisms to
eliminate fat
• OxLDL induce oxysterols in the
cytoplasm
• This activates transcription of
»
»
»
»
»
»
»
A E ffor HDLs
ApoE
HDL
ABCA1- for cholesterol channel
Fatty acid synthetase, for forming fats
Fatty acids are made into free cholesterol
Cholesterol bound to ApoA1
Cholesterol/ApoA1 goes to HDL
Increase cholesterol outflow increases removal
of esterified cholesterols also
Cell protection against free cholesterol
Ox LDL
Apo E
-carries FC out
FC
Oxysterols
CE
lysosome
ABCA1
-transports FC
HDL
FC
LXR RXR
Apo E
ABCA1
SREBP1c
FAS
LXR RXR RE’s
FFA
Apo A
-picks up FC
CE
Lipid droplet
LXR = liver X receptor
RXR = Retinoic acid receptor
LIVER
CE = cholesterol ester
FC = free cholesterol, ABCA1 = transporter, FAS = fatty acid synthesis, FFA = free fatty acid
82
Proatherogenic activity of oxidized LDL’s
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
Increase foam cells (increased scavenger uptake)
Products are chemotactic to monocytes, T cells
Cytotoxic, apoptosis
Mitogenic for smooth muscle cells
Increase scavenger receptor expression
Increase PPAR actvity (gene fx, apoptosis)
I
Immunogenic;
i autoantibody
t
tib d and
d T cellll activation
ti ti
Increases LDL aggregation
Substrate of spingomyelinase; aggregates LDL’s
Procoagulant, increases tissue factor, platelet activating factor
Increased monocyte CSF, MCF, IL-1, IL-8, chemokine
receptors,NO
Increased calcium and Nfkappa B
Increased expression of matrix metalloproteinases
Increased adhesion molecule expression
Reduce vasomotor activity and vascular contractility
Trans fatty acids
• A double bond between carbons in a fatty acid that changes the
conformation from cis to trans
– Oleic and elaidic acid are 18 carbon
• Oleic is cis; elaidic is trans
• Trans fatty acids
– IIncrease LDL
LDLs, d
decrease HDL
HDLs, iincrease cholesterol:HDL
h l t l HDL ratio
ti and
d
increase blood triglyceride levels
– Increase B100 size, secretion, lipid content
– Decrease LDLapoB100 catabolism (increasing LDLs)
– Increase cholestrol ester transfer from HDLs to LDLs
• Thus decreasing HDL removal of cholesterol
– Increase ICAM-1, VCAM-1 and E selectin on endothelial cells
• Pro-inflammatory promoting atherosclerosis
Atheroma formation
• Diagram
• Stages:
–
–
–
–
–
–
–
–
–
Fatty streaks in children
Isolated foam
f
cells in adults
Multiple foam cells
Extracellular lipid accumulation
Confluent lipid core
Fibrous cap with smc proliferation
Surface ulcer, thrombus
Calcification
Fibrosis
Can regress
Ab
Above
thi
this liline
83
Robbins and Contran Pathologic basis of disease, 7th edition
Robbins and Contran Pathologic basis of disease, 7th edition
Robbins and Contran Pathologic basis of disease, 7th edition
84
Factors leading to atherosclerosis
• Cigarette smoking
• Two fold increase in atherosclerosis
• Hypertension
• Higher incidence blacks; 3% of Americans,
Americans 75% of those
over 75 years of age
• Diabetes mellitus
• 14 million americans
• Serum cholesterol levels
• Familial hypercholesterolemia
– Increased cholesterol, types on next page
• 50% of Americans (24-70 years) have >200 cholesterol
levels
LDL Receptor
• LDL R family
• Apo E R2, VLDL R, megalin, LDLR, LDL
receptor-related protein
• Genetic mutation can reduce expression
– Multiple types of mutations identified in man
• Results in increased circulating LDL’s,
hypercholesterolemia and foam cell formation
Genetic Causes of Hyperlipoproteinemias
• I increased chylomicron
• IIa increased LDL*
• IIIa increases LDL/VLDL*
• III increased chylomicrons
• IV increased VLDL*
• V increased VLDL/chylo
*10, 40, 45% respectively
• Others
mutations in lp lipase
mutation of LDL or Apo B
mutation of LDL or Apo B
mutation of Apo E
mutation of lp lipase
mut. Lpl or C2
• Decreased HDL; mutation in Apo AI
• Tangier’s dz, mutation in cholesterol transport
• Loss of HMG-CoA reductase
85
Schnauzers, Cats, Cows, Ewes
• Schnauzers develop hyperlipidemia
• Cats, cows and ewes, develop fatty
livers
– Cats, L-Carnitine defect?
– Impaired ability to carry fats into the mitochondria
and out of the hepatocyte
• Cows and ewes
– Often related to
pregnancy/parturition/lactation
Lipid disorders in dogs and cats
Dogs
Primary disorder
Idiopathic hyperlipidemia
hypercholesterolemia
Secondary disorder
high fat diet
pancreatitis
hypothyroidism
yp y
cholestasis
hyperadrenocorticism
nephrotic syndrome
Cats
Primary disorders
inherited
hyerchylomicronemia
primary
hypercholesterolemia
Increased TG, cholesterol pancreatitis
Increased cholesterol
retinal degen
increased cholesterol
increased TG
increased cholesterol
increased cholesterol
increased TG, cholesterol
increased TG, cholesterol
pancreatitis?
pancreatitis
atheroma
insulin resistance
increased TG, cholesterol Decreased LPL
increased cholesterol
xanthomas
TG = triglycerides
Bauer JE: JAVMA 224:668-675, 2004
Appetite regulation
• Arcuate nucleus of the hypothalamus, two parts:
– 1. AgRP/NPY—increases appetite and metabolism
» Activated by: Ghrelin R (GhR) (Ghrelin; stomach),
Agouti-related peptide (AgRP), NPY
» Inhibited by: PYY, YY (gut), leptin (adipose), insulin,
amylin, pancreatic polypeptide (pancreas)
– 2. POMC/CART—decreases appetite and metabolism
» Activated by leptin (adipose), insulin (pancreas), cocaineamphetamine pro-opiomelanocortin
amphetamine,
pro opiomelanocortin POMC
• Nucleus tratus solitarius (NTS), medulla
• Receives second order neurons from the arcuate nucleus
– Also regulated directly by cholecystikinin (liver) and vagus
nerve (for sensing stomach stretching)
– Sends signals to the rest of the body
• The hypothalamus arcuate nucleus and NTS
regulate feed centers and behavior in the
hypothalamus nuclei: ventromedial,
dorsomedial, and lateral
86
Leptin
• Leptin will decrease appetite in those
with mutant leptin, but not normal
individuals
• In other words, excess leptin doesn’t decrease
appetite in an average person
• Leptin protects against weight loss/starvation
– There is decreased leptin with decreased fat,
therefore increased eating
– As above, excessive leptin doesn’t decrease intake
A few obesity targets
•
Inhibiting hunger
–
–
–
Cannibas receptor inhibitor (preventing hunger “munchies”)
PYY peptide (decreasing appetite)
SlimFast (bulky fiber? poorly degraded?)
•
Preventing food absorption
•
Delay gastric emptying
•
Enhance fat burning
–
–
–
–
•
Alizyme, prevents fat absorption in gut
Olestra-WOW! Chips, a fat not absorbed
Amylin
Human growth hormone, amphetamines
Degradation of fats
–
Lipases
•
Altering fat metabolism
•
Nutrisystem, Nurrisystem for men (dan marino), Jenny Craig, Atkins Diet,
Scarsdale diet, Grapefruit diet, Slimfast, Trimspa, Houdia, LA weight loss,
Inches Away, weight watchers,
Exercise
–
Acetyl Co-Enzyme A carboxylase-induces fatty acid synthesis
•
•
TRB3 induces degradation of ACC thus decreasing synthesis
PPAR
• PPAR alpha
– Fatty acid oxidation in liver (heart, muscle, kidney artery)
• Tissues with high fatty acid oxidation
• PPAR gamma
– Lipid storage in liver and adipose tissue
– Activated
A i
db
by TZD (Thi
(Thiazolidinedione),
lidi di
) an anti-diabetic
i di b i d
drug
• Increases skeletal muscle and liver sensitivity to insulin via activation of
PPAR gamma
• Also reduces inflammation
• Weight gain side effect and sodium retention
– Widespread and present in many tissues
• PPAR beta/delta
– Fatty acid oxidation and energy decoupling in adipocytes
• Only weakly activated
87
PPAR
Cholesterol synthesis
• Cholesterol
• 27 carbons, synthesized in liver from acetyl CoA
• Acyl transferase mediates acetyl CoA formation
• Three acetyl CoA’s join to make HMG CoA
– HMG-CoA
» Can make acetoacetate and ketone bodies
» Can make nevalonate to form cholesterol
• High cholesterol levels in hepatocyte
– HMG CoA reductase is inhibited and ketones form
– No LDL receptors are made (to decrease hepatocellular
uptake of LDL and thereby reduce cholesterol intake)
• Low cholesterol levels in hepatocyte
– HMG-CoA reductase makes nevalonate
– Increased LDL receptor to increase uptake of cholesterol
• Cholesterol forms:
– Bile salts
– Steroid hormones
Robbins and Contran Pathologic basis of disease, 7th edition
88
Drugs that lower cholesterol
• Cholestyramia
• Bile acid exchange resin. Binds cholesterol.
• Lovastatin
• Inhibits HMG-CoA reductase. Prevents cholesterol
formation
• Bezafibrate
• Stimulates lipoprotein lipase. Triggers cholesterol
degradation.
• Probucol
• Lowers triglycerides, mechanism uncertain
Utilization of energy
• Brown fat
• Fidgeting
– Levine JA: Nonexercise activity thermogenesis (NEAT): environment
and biology. Am J Physiol Endocrinol Metab. 2004 May;286(5):E67585:
• Nonexercise activity thermogenesis (NEAT) is the energy
expended
d d ffor everything
thi th
thatt iis nott sleeping,
l
i
eating,
ti
or sports-like
t lik
exercise. It includes the energy expended walking to work, typing,
performing yard work, undertaking agricultural tasks, and fidgeting.
• The variability in NEAT might be viewed as random, but human
and animal data contradict this. It appears that changes in NEAT
subtly accompany experimentally induced changes in energy
balance and are important in the physiology of weight change.
Inadequate modulation of NEAT plus a sedentary lifestyle may thus
be important in obesity. It then becomes intriguing to dissect
mechanistic studies that delineate how NEAT is regulated into
neural, peripheral, and humoral factors.
• Exercise
Brown fat utilization and thermogenesis
• Cold increases
– PGC1 (a powerful transcriptional coactivator) for PPAR
gamma (peroxisome proliferator activator receptor)
– TR (thyroid hormone receptor)
– RAR (retinoic acid receptor)
– ER (estrogen receptor)
• PGC1
PGC1, TR
TR, RAR and
d ER activate
ti t UCP ((uncoupling
li proteins)
t i ) and
d
genes of mitochondrial respiratory chain for ATP synthesis
(ATPase and cytochrome oxidase C)
• The UCP (uncoupling proteins) are in the inner mitochondrial
membrane along with F0/F1 ATPase.
• The UCP allow H+ passage with loss of gradient resulting in heat
with reduced ATP.
– PGC1 also activates NRF 1and 2 (nuclear respiratory
factors) for mitochondrial biosynthesis and increases
conversion of type I to type II muscle fibers
• Increased mitochondria for more heat
89
Hyperglycosylation and hyperglycemic
vasculopathy
• Hyperglycemic vasculopathy in diabetes
mellitus
– Persistent levels of blood glucose leads to
glycosylation
l
l ti off vessels
l
– Glucose attaches to amino groups of proteins
• Schiff base is formed and is reversible
• Amadori complex
– A more stable adherence of glucose to the protein
• Advanced glycosylation end products (AGEs)
– Irreversible glycosylation
Mediation of glycosylation damage
• Glycosylation of LDL’s, hemoglobin, albumin,
extracellular matrix proteins, and basement
membrane (thickens)
• AGEs bind RAGE receptors on macrophages
– Induction of IL-1, TNF, inflammation
• Glycosylation of amino groups in DNA
nucleotides
– Altered transcription and increased strand breaks
Intracellular glucose damage
• Occurs in tissues that do not require insulin for
glucose entry (in diabetics)
– Lens, neurons, blood vessels
• Converted to sorbitol by aldose reductase and then
sorbitol to fructose by sorbitol dehydrogenase
• Sorbitol and fructose induce oncotic pressue, influx of
water and swelling
– Sorbitol also reduces ATPase pumps in schwann cells (of
nerve fibers) and retinal capillary endothelial cells
– Also, increased protein kinase C and diacylglycerol can
occur
90