Anda di halaman 1dari 33

MOL 345: Final Study Guide

Chapter 27: Integration and Cytoplasm: glycolysis, FA synthesis, and cholesterol synthesis
regulation, Pathways that take place in Inner mito membrane: oxidative phosphorylation
the cytoplasm, IMM, and mitochondrial Mitochondrial matrix: CAC, FA oxidation, ketone bodies synthesis
matrix, interplay between 2 Matrix and cytoplasm: gluconeogenesis; urea cycle
Glucose transporters Work by facilitated diffusion
Glut 1 + 3: in all cells, take up glucose, small Km for constant glucose transport
Work by ____ Glut 2: liver and pancreatic β cells, high Km – sense glucose level and increase
secretion of insulin
Glut 4: transport glucose  muscle and fat (↑ insulin causes ↑ Glut 4 transporters in
plasma membrane, ↑ exercise and ↑ Glut 4 in muscle membrane)
Glut 5: small intestine, fructose transporter
Acetyl CoA A hub for all metabolic pathways
Hub for ___ metabolic pathways Definition: An activated 2-carbon unit
What is it? Derived from: pyruvate (oxidative decarboxylation) and ultimately glucose
Derived from? fatty acids (β -oxidation), catabolism of ketogenic amino acids (those that give rise
Fates? to ketone bodies or fatty acids: Ile, Leu, Trp, Phe, Tyr, Lys)
Fates: Complete oxidation to CO2
Conversion to HMG CoA for synthesis of cholesterol and ketone bodies (in liver)
Conversion to citrate for export from mitochondria to cytosol for synthesis of FAs
Pyruvate A major junction for carbohydrate and amino acid metabolism
A junction for ___ and ___ metabolism Derived from: G6p (glycolysis)
Derived from? lactate (fermentation)
Fates? catabolism of amino acids Gly, Ala, Ser, Cys, Thr, and Trp
Fates: reduction to lactate under anaerobic conditions (to regenerate NAD+ and
allow glycolysis to continue)
transamination to alanine (a major link b/t carbohydrate and aa metabolism)
carboxylation to OAA (in mitochondria) for gluconeogenesis and replenishment of
CAC intermediates
oxidative decarboxylation to acetyl CoA (in mitochondria) for oxidation via CAC,
synthesis of fatty acids
Glucose 6-phosphate A major hub of carbohydrate metabolism
Hub of ___ metabolic pathways Derived from: phosphorylation of glucose, gluconeogenesis (from pyruvate and
Derived from? gluconeogenic aa), glycogen breakdown
Fates? Fates: oxidation to pyruvate when ATP or carbon skeletons are required
Entry into pentose phosphate pathway to produce NADPH for reductive
biosynthesis, Synthesis of glycogen, Dephosphorylation and export from cell (liver)
Brain Neither stores nor produces fuel molecules
Store/production of fuel molecules? In resting state, uses 60% of body’s glucose
Uses what as fuel in resting state? During starvation, uses ketone bodies (fatty aid bound to plasma proteins, cannot
Starvation? cross the blood-brain barrier)
Glucose transporter Glucose transported into brain by GLUT3 (low KM) 0 usually saturated.
Skeletal Muscle Uses glucose during activity, fatty acids while resting
Use ___ as fuel during activity, ___ 1. Synthesizes and stores ¾ body’s glycogen, mobilized during activity
during rest G6P produced by glycogen breakdown, cannot be exported because muscle
Does it synthesize/store fuels? When does not have glucose-6-phosphatase, is charged
does it produce lactate? Where is 2. Production of lactate from pyruvate during active contraction and rate of
lactate transported to? glycolysis >> rate of CAC, NAD+ must be regenerated from NADH for glycolysis to
Where does alanine come into play? continue, Pyruvate + NADH + H+ ↔ lactate + NAD+
Lactate: transported to the liver (Cori cycle): shift metabolic burden to liver
Alanine: Muscle absorbs and transaminates branched aa to use C-skeleton as fuel,
can’t form urea. Nitrogen released into blood as alanine, which the liver converts
into urea and glucose
Adipose Tissue A major storage depot (TAGs stored in adipose tissue – enormous fuel reservoir)
A major _____ (Why?) Specialized for esterification of fatty acids and their release from TAGs
Specialized for ___ Esterification requires glycerol-3-phosphate (glycolytic intermediate)
Esterification requires ____ Intracellular [glucose] dictates whether fatty acids are re-esterified or released
__ dictates whether FAs are ___ or ___ When released, FAs and glycerol are exported to the liver
When released, FAs and glycerol are _ Endocrine organ: adipose synthesizes peptide hormone leptin, signals fed state in
Function as an endocrine organ? hypothalamus
Synthesis and turnover of Glucose (from liver)  GAP
triacylglycerols by adipose tissue VLDL (from liver) and Chylomicron (from intestine  fatty acyl CoA
Role of glucose in tissue GAP + Fatty Acyl CoA  TAGs
TAGs are hydrolyzed to FAs and glycerol by lipases. (lipase is reversibly
phosphorylated)
Glucose determines whether FA are released in blood (no GAP) or reesterified
Kidney Uses fuel to power reabsorption of glucose and water during filtration of blood
Uses fuel to? Reabsorption of glucose plasma (waste products excreted in urine)
via ___, powered by ___ Reabsorption of glucose is via glucose symporter, powered by NA+K+ ATPase
During Starvation, a site of ___ During starvation, a site of gluconeogenesis
Liver Most metabolites absorbed by the intestine pass first through the liver
What passes through liver? Major site Major site of amino acid deamination and synthesis of urea
of? Uses ___ as fuel. Uses α-keto acids (from amino acids degradation) as fuel to yield acetyl CoA
Other metabolic functions provided by Lactate, alanine (muscle), glycerol (adipose), glucogenic amino acids converted to
liver G6P, produces glycogen + acetyl CoA (which produces FAs, cholesterol, bile salts)
Role in regulating lipid metabolism Fumarate produced by urea cycle yields the OAA necessary for acetyl CoA to be
processed in TCA cycle
When fuels are abundant, FAs are esterified and secreted into blood as low density
Why can’t it use branched chain amino lipoproteins. (When Malonyl CoA is abundant, FA can’t enter mito matrix for β
acids, acetoacetate as fuel? oxidation, instead are exported to adipose tissue to be incorporated into TAGs)
Can’t use branched chain amino acids as fuel because it cannot complete
transamination of amino acid as can muscles.
Can’t use acetoacetate as fuel because it doesn’t has transferase needed to active
acetoacetate to acetyl CoA (used in brain)
Control sites of Glycolysis PFK (committed site) F6P  F-1,6-BP: (-) ATP, Citrate, H (+) AMP, F26BP
*in liver: (+) F-2,6-BP (produced by PFK2 when blood glucose is high), allosteric
activator of PFK by ↑ velocity at low [substrate] and ↓ responsiveness of
PFK1 to allosteric inhibitor ATP.
Hexokinase (-) G6P
PK (controls outflow of glycolysis)
(+) F1,6BP, (-) ATP, Alanine, phosphorylation by glucagon-stimulated cAMP
cascade (in liver)
Control sites of Pyruvate Pyruvate  acetyl CoA (pyruvate dehydrogenase) = irreversible link between
Dehydrogenase Complex carbon atoms of carbohydrates/amino acids to oxidation by CAC/synthesis of lipids
(-) acetyl CoA inhibits transacetylase (E2), NADH inhibits E3 – energy needs of cell
are met or FA are being degraded, save glucose, ATP, phosphorylation by kinase
(+) pyruvate, dephosphorylation by phosphatase (simulated by Ca2+)
Hormones: epinephrine  α-adrenergic receptor  ↑ Ca2+  activates phosphatase
 ↑ PDH
Insulin  ↑ phosphatase  ↑ pyruvate  acetyl CoA  ↑ FA in tissues with FA
synthesis (liver, adipose)
Control sites of CAC 1. isocitrate  α-ketoglutarate (isocitrate dehydrogenase)
(-) ATP, NADH (cooperative inhibition of NAD+)
(+) ADP, NAD+, Mg2+
2. α-ketoglutarate  succinyl CoA (α-ketoglutarate dehydrogenase)
(-) ATP, succinyl CoA, and NADH
3. Acetyl CoA + OAA  Citrate (citrate synthase)
(-) ATP, ↑ Km for acetyl CoA
Complete oxidation of an acetyl unit by CAC = 1 GTP, 3 NADH, 1 FADH2
Control sites of oxidative Respiratory control – tight coupling of electron transport and ADP
Phosphorylation phosphorylation, ensures rate of CAC matches need for ATP
(+) ADP, NADH, FADH2, O2, Pi
Control sites of gluconeogenesis F-1,6 BP  F6P (F16BPase)
(-) AMP, F2,6BP
(+) Citrate
Control sites of glycogen synthesis Breakdown: Glycogen + orthophosphate  G1P (glycogen phosphorylase)
and degradation Synthesis: UTP + G1P  UDP-glucose (UDP-glucose pyrophosphorylase)
1. Protein Kinase A (phosphorylation)
↑ phosphorylase kinase  ↑ glycogen phosphorylase  [↑ glycogen breakdown]
↓ glycogen synthase (ab)  [↓ glycogen synthesis]
2. Protein Phosphatase 1 (dephosphorylation)
↓ phosphorylase kinase  ↓ phosphorylase a  [↓ glycogen breakdown]
↑ glycogen synthase (ba)  [↑ glycogen synthesis]
PKA regulates PP1:
Epinephrine/glucagon  activates PKA:
a. phosphorylate RG1, releases catalytic unit (assoc. needed for PP1 activity)
b. phosphorylated inhibitor binds to catalytic unit of PP1
3. Insulin (activates glycogen synthase kinase, PP1)
Insulin  activates tyrosine kinase receptor in plasma membrane  phosphorylates
IRS  signal cascade  glycogen synthase kinase inactive  active
dephosphorylated glycogen synthase (PP1 also dephosphorylates glycogen synthase)
Insulin phosphorylates RG1 subunit on PP1 (associates with glycogen)
Control sites of FA synthesis Acetyl CoA + ATP + HCO3-  malonyl CoA + ADP + Pi + H+ (Acetyl CoA
carboxylase) = committed step
- global regulation: hormones change phosphorylation
Active dephosphorylated carboxylase  inactive phosphorylated carboxylase
(AMP-activated protein kinase AMPK)
(+) Glucagon, epinephrine
Inactive phosphorylated carboxylase  active dephosphorylated carboxylase
(protein phosphatase 2A)
(+) insulin (stimulate accumulation of TAGs by muscle and adipose tissue)
- local regulation:
(+) Citrate  polymerizes inactive carboxylase dimers into active filaments,
counteracts phosphorylation (high ATP/acetyl CoA = raw material sand
energy available for FA synthesis)
(-) AMP  turn on AMPK  phosphorylates and inactivates carboxylase (low
energy charge, fats only synthesized when energy is abundant)
(-) Palmitoyl CoA (abundant when FA abundant)  carboxylase filaments
disassemble, inhibits translocase that transports citrate from mit cyto,
inhibits G6Phosphate dehydrogenase, generates NADPH
Control sites of FA Degradation Malonyl CoA (from carboxylase) --] Carnitine acyltransferase
(block entry of FA CoAs into mito for degradation when synthesizing FA)
NADH --] HMG-CoA dehydrogenase
Acetyl CoA --] thiolase (β -oxidation enzyme inhibited when energy charge is high)
Well Fed state reactions - glucose and aa transported from intestine to blood
- dietary lipids are packaged into chylomicrons and transported to the blood by
lymphatic system, secretion of insulin by β cells in pancreas (stimulated by glucose
and PNS)
- Insulin initiates protein kinase cascades, ↑ glycogen synthesis in muscle and liver,
↓ gluconeogenesis by liver, ↑ glycolysis and synthesis of FAs
- Liver has glucokinase (high Km, only active when blood-glucose levels are high),
liver forms glucose-6-phosphate more rapidly as blood-glucose levels rises. Increase
in G6P + insulin = ↑ glycogen.
-Phosphorylase a = glucose sensor + cleaves glycogen
- high glucose  ↑ phosphatase activity  inactivates glycogen phosphorylase (a
b): allosteric shift from glycogen degradation to synthesis mode
- high insulin = entry of glucose into muscle and adipose tissue. (glucose  GAP for
TAGs), Insulin promotes uptake of branched-chain aa (val, leu, ile) by muscle,
stimulates protein synthesis, inhibits intracellular protein degradation
Early Fasting Low insulin, high glucagon
Hormones? Processes: Mobilization of TAGs in adipose tissue, gluconeogenesis in liver
Dominant metabolic processes? Changes: liver oxidizes FAs from adipose, muscle shifts to use fatty acids because
Changes in fuel Use? low [insulin] leads to reduced uptake of glucose, the acetyl CoA product inhibits
conversion of pyruvate to acetyl CoA.
Pyruvate, alanine, and lactate are exported to liver for gluconeogenesis
Prolonged Starvation After several weeks, synthesis of large quantities of ketone bodies in liver
After several weeks, synthesis of ___ Acetyl CoA from β -oxidation of fatty acids in liver cannot enter CAC because
Why can’t Acetyl CoA enter CAC? gluconeogenesis limits [OAA], ketone bodies are made and exported
Production of ___ reduces need for __, Production of ketone bodies decreases the need for glucose, less protein is degraded
less ___ is degraded. Death results than during the initial period of starvation
from? Protein degradation accelerates after reserves exhausted, death from loss of heart,
liver, or kidney function
Where does Acetoacetate come from? Acetoacetate is activated by transfer of CoA from succinyl CoA to give acetoacetyl
CoA. Thiolase cleavage yields to molecules of acetyl CoA, which then enters the
Effects of excess Acetyl CoA CAC. Ketone bodies = fatty acids that are an accessible fuel source of the brain.
Glucose  Pyruvate  OAA or Acetyl CoA
Acetyl CoA blocks PDH from making more acetyl CoA, stimulates gluconeogenesis
and production of OAA from pyruvate (pyruvate carboxylase).
Obesity: What is it? Symptoms? Metabolic disorder, BMI > 30, may develop hypertension, cardiovascular disease,
2 neural signals that regulate appetite? type II diabetes
In _____ In hypothalamus a. hormones (leptin – produced from fat in adipose, signals well-
fed state), 2. Ghrelin, peptide YY (made in intestinal cell), released upon eating and
signals hunger and satiation
b. metabolites: 1. glucose (signal for insulin release, low concentration promotes
hunger), 2. free fatty acids and branched aa’s – signal to suppress feeding
Diabetes Type I : Cause: Insulin-dependent
___ dependent auto-immune destruction of β cells in islets of Langerhans in pancreas (early in
Cause life)
Symptom Symptoms: no insulin produced, glucagon overproduced  high glucose,
Treatment permanent “starvation” state
inhibits glycolysis, increases gluconeogenesis in liver, because of PFK2 and
F26BPase = glycogen breakdown
Glucose released into blood, concentration already high
Excreted by kidney = water loss and dehydration
Uncontrolled lipid and protein breakdown
- ↑ acetyl CoA high (lipid)  ketone bodies
- ↑ ketone bodies exceeds acid-base balance in kidney  ↓ pH 
dehydration and coma
1. Without insulin, liver can’t take up glucose, OAA, CAC + β -ox of fatty acids ↓
2. Free FA are released from adipose tissue into bloodstream
4. Liver picks up FA, converts into ketone bodies
5. Ketone bodies are moderately strong acids, blood pH drops
6. Acidosis, Coma + Death
Treatment: administer insulin, control diet and exercise
Diabetes Type II : Cause: Although insulin is made normally, cells can’t respond to hormone (messed
Cause up insulin receptor/production)
Symptom Symptom: patients are hyperglycemic (excess blood glucose) because cells do not
Treatment take up and utilize glucose normally
Treatment: drugs that stimulate insulin production or responsiveness to hormone
(metfomin), diet and exercise to use up glucose
What controls development of Type II WT adipocyte (releases Leptin – signals fed state, adiponectin – promotes insulin
diabetes? action, removes glucose from blood) converted to ‘Fat’ adipocyte
*excess carbohydrates, proteins, or fat converted to FA, stored as TAGs
‘Fat’ adipocyte, stores more TAG, increase number and size (releases retinol
binding protein 4 RPB4, block insulin axn, contributes to insulin resistance, TNF α,
IL6 – inflammatory cytokines, induce chronic, low level inflammation in adipose,
linked to development of insulin resistance)
Compare allosteric interactions and Allosteric interactions – enable enzymes to rapidly detect diverse signals and adjust
covalent modifications activity accordingly
Covalent modifications – a final step in an amplifying cascade that allows
metabolic pathways to be rapidly switched on or off by low concentrations of
triggering signals. Insulin, glucagon, and epinephrine stimulate protein kinases.
Covalent modifications (s-min) last longer than reversible allosteric interactions (ms
– sec)
CAC provides what intermediates for Succinyl CoA forms porphyrins and citrate forms fatty acids.
biosynthesis?
Reciprocal regulation of AMP inhibits F16BPase (gluconeogenesis), activates PFK (glycolysis)
gluconeogenesis and glycolysis Citrate activates F16BPase (gluconeogenesis), inhibits PFK (glycolysis)
F26BP inhibits F16BPase (gluconeogenesis), activates PFK (glycolysis)
How is compartmentation involved in Fatty acids are transported into mitochondria for degradation only when energy is
FA synthesis/degradation required, whereas fatty acids in cytoplasm are esterified or exported.
Chapter 15: Principles of Catabolism + Anabolism
Metabolism
Define metabolism, catabolism,
anabolism
Three major purposes of living
organisms that require constant input of
free energy
Significance of free-energy change of
reactions and relation to the
equilibrium constant, concentrations of
reactants and products of reaction
Structure of ATP
Role
How can coupling a reaction with the 108
hydrolysis of ATP change the
equilibrium concentration of the
products to the concentrations of the
reactants by a factor of ___

Compounds that have high group- Compounds that release large amounts of free energy on hydrolysis or oxidation
transfer potential (define)
Creatine phosphate serves as ___ “high-energy” buffer in vertebrate muscle
ATP-ADP cycle of energy exchange in
biological systems
How does oxidation of carbon
compounds drive the formation of
ATP?
Define proton gradient
How does proton gradient couple
unfavorable reactions to favorable
ones?
Major stages in extraction of energy
from foodstuffs
Why are most high-energy compound
and reduced electron carriers
kinetically stable, despite high-energy
status?
Require __ for reactions enzymes
Structure of CoA, role as ___ Carrier of acetyl/acyl groups

List major activated carriers (electrons


and activated groups) in metabolic
reactions
Six basic types of chemical reactions
encountered in cellular metabolism
Three major mechanisms for the
regulation of metabolism
Energy charge vs. phosphorylation
potential
Common structural features of NAD,
FAD, and ATP
Chapter 16: Glycolysis Glycolysis = 1 glucose  2 pyruvate + 2 ATP
Define glycolysis
Three stages of glycolysis Stage 1: energy input, trap glucose in cell to be cleaved into 3 C units
Stage 2: cleave F1,6BP into 3C units
Stage 3: ATP output, oxidation to pyruvate
Why phosphorylate glucose? 1. G6P can’t pass through membrane, not a substrate for transporters
2. destabilizes glucose, facilitate further metabolism
Hexokinase mechanism upon glucose Induced-fit rearrangement
binding, two important consequences 1. environment around glucose – nonpolar, favors phosphorylation
of this step in glycolysis 2. keep out water
The net reactions for the transformation 1. Glucose + ATP  G6P + ADP + H+ (hexokinase) phosphoryl transfer
of glucose into pyruvate and enumerate 2. G6P ↔ F6P (phosphoglucose isomerase) isomerization
the ATP and NADH molecules formed 3. F6P + ATP  F1,6BP + ADP + H+ (PFK) phosphoryl transfer
4. F1,6BP  GAP + DHAP (aldolase) aldol cleavage
5. DHAP ↔ GAP (TIM) isomerization
6. GAP + NAD+ + ATP  1,3 BPG + NADH + H+ (GAP DH) ox. phosphorylation
7. 1,3 BPG + ADP  3 PG + ATP (Phosphoglycerate kinase), sub level phosphor.
8. 3PG  2 PG (phosphoglycerate mutase), phosphoryl shift
9. 2PG  PEP + H2O (enolase) dehydration
10 PEP  Pyruvate + ATP (pyruvate kinase), substrate level phosphorylation
Rxns for the conversion of pyruvate 1. Ethanol: pyruvate  acetaldehyde + CO2 (pyruvate decarboxylase)
into ethanol, lactate, or acetyl CoA. Acetyladehyde + NADH ↔ ethanol + NAD+ (alcohol dehydrogenase)
Role of alcoholic fermentation and 2. Lactate: pyruvate + NADH ↔ lactate + NAD+ (lactate dehydrogenase)
lactate formation No oxidation-reduction, regenerate NAD+ for glycolysis to continue anaerobically
Structure of the NAD+-binding region Rossman fold
common in dehydrogenases
Why does PFK use AMP and not ADP Some ATP can be salvaged from ADP, AMP is signal for low-energy state, provides
as positive regulator? an especially sensitive control
Regulation of glycolysis in skeletal Liver PFK: (+) F-2,6BP, (produced when abundant F6P) feed forward stimulation
muscle cells to that in liver cells Glucokinase provides G6P for glycogen + FAs synthesis (lower affinity for glucose)
L and M isozymes of P, L form controlled by reversible phosphorylation:
When BG low, glucagon-triggered cAMP cascade leads to phosphorylation,
decrease activity.
Physiologic significance of Maintain blood glucose levels
gluconeogenesis. Primary precursors of AA (OAA, pyruvate), lactate (pyruvate), glycerol (DHAP)
gluconeogenesis Glycerol  Glycerol phosphate (glycerol kinase)  DHAP (glycerol phosphate DH)
How do Cancer and Exercise training Cancer cells grow blood vessels, use lactic acid as primary source of ATP, increase
affect Glycolysis in a similar fashion? expression of GLUT1 + 3 to uptake glucose.
Anaerobic exercise: enhance blood-vessel growth and generate ATP anerobically
Different steps in gluconeogenesis. Glycolysis Gluconeogenesis
Enzymes, intermediates, and cofactors Glucose + ATP  G6p + ADP G6P + H2O  Glucose + Pi
involved in reactions (hexokinase) (glucose 6 phosphatase)
F6P + ATP  F16BP + ADP (PFK) F16BP + H2O  F6P + Pi (F 1,6
biphosphatase) – hydrolysis
PEP + ADP  Pyruvate + ADP Pyruvate + CO2 + ATP + H2O  OAA
(pyruvate kinase) + ADP + Pi + 2H+ (pyruvate
carboxylase)

OAA + GTP  PEP + GDP + CO2


(PEP carboxykinase)
Major organs that carry out Liver, kidney
gluconeogenesis. Location of enzymes Matrix (PEP carboxykinase, F16Biphosphatase, G6phosphatase) and cytoplasm
in cell compartments. (pyruvate carboxylase).
Transfer of carboxylase reaction OAA (product of carboxylase reaction) transported to cytoplasm in the form of
products malate, which is reoxidized to OAA in cytoplasm.
Pyruvate carboxylase: Role of biotin, Biotin = covalently attached biotin group, carrier of activated CO2 in the pyruvate
Effect of acetyl CoA, maintaining the carboxylase. Biotin is not carboxylated unless acetyl CoA is bound to enzyme.
level of CAC intermediates
Calculate the number of high-energy Six triphosphate molecules are hydrolyzed to synthesize glucose  pyruvate in
phosphate bonds consumed during gluconeogenesis (G6P, 2 Pyruvate + 4 ATP)
gluconeogenesis, number formed 2 molecules of ATP are generated in glycolysis  pyruvate
during glycolysis Extra cost of gluconeogenesis = 4
Coordinated control of the enzymes in * energy needed = glycolysis, energy surplus =gluconeogenesis
glycolysis and gluconeogenesis 1. F6P  F1,6BP
Gluconeogenesis: (-) AMP, F-2,6-BP (+) Citrate
Glycolysis: (+) AMP, F-2,6-BP (-) ATP, Citrate, H
2. PEP ↔ Pyruvate
Gluconeogenesis: (-) ADP (+) Acetyl CoA (pyruvate  OAA)
Glycolysis: (+)F-2,6-BP (-) ATP, Alanine
Fused-domain structure of PFK2 that Low BG: F2,6BP loses a phosphoryl group  f6p, doesn’t bind PFK
forms and degrades F26BP. Phosphorylates bifunctional enzyme – lower level of F26BP, ↓ glycolysis
Reciprocal regulation of enzyme High BG: F6P accelerates formation of F-2,6-BP , facilitate the dephosphorylation
Substrate cycle can of bifunctional enzyme
Amplify metabolic signals
Produce heat
Outline Cori cycle. Muscles produce lactate, move to liver, gluconeogenesis to produce glucose
Biological significance transported back to muscles.
Relieve metabolic burden of muscles
Chapter 17: The Citric Acid Cycle Common pathway for oxidation of carbohydrates, fatty acids, and amino acids. Most
Role of CAC in aerobic metabolism enter as Acetyl CoA
Pyruvate dehydrogenase as a multi- Produces CO2 and NADH
enzyme complex E1: TPP prosthetic group –decarboxylation and oxidation of pyruvate
E2: Lipoamide – transfer of acetyl group to CoA
E3: FAD – regeneration of the oxidized form of lipoamide
Cofactors that participate in pyruvate (catalytic) TPP, lipoic acid, FAD, (stoichiometric) CoA and NAD+
dehydrogenase complex reactions 1. decarboxylation of pyruvate by E1-TPP
2. transfer of acetate to lipoamide
3. transfer of acetyl group to CoA
4. regenerate lipoamide and extract electrons
Enzymatic mechanism of citrate Condensation reaction, binds OAA first, then acetyl CoA, forms an enol
synthase intermediate  citryl CoA complex. Enzyme only cleaves citryl CoA, not acetyl
CoA. *induced fit* prevents undesirable side reactions
Role of iron in the enzyme aconitase Citrate  aconitate  isocitrate (dehydration, hydration), Iron binds to citrate
through COO- group and Oh group. Helps dehydrate, rehydrate substrate
Compare the rxn catalyzed by the α- - substrate: Pyruvate, α-ketoglutarate decarboxylation of α-ketoacid)
ketoglutarate DH to the rxn catalyzed - formation of a thioester linkage with CoA (high transfer potential
by pyruvate dehydrogenase complex - product: acetyl CoA, succinyl CoA
Enzymatic reactions of the CAC in 1. acetyl CoA + OAA + H2O  citrate + CoA + H+ (citrate synthase), condensation
sequence. Name enzymes 2. citrate ↔ isocitrate (aconitase) hydration, dehydration
3. isocitrate + NAD+  α-ketoglutarate + CO2 + NADH (isocitrate DH), ox. decarb.
-or- Examples of condensation, 4. α-ketoglutarate + NAD+ + CoA  succinyl CoA + CO2 + NADH (α-keto DH),
dehydration, hydration, oxidative decarboxylation
decarboxylation, oxidation, and 5. succinyl CoA + ADP  succinate + CoA + ATP (succinyl CoA synthase),
substrate-level phosphorylation substrate level phosphorylation
reactions 6. succinate + E-FAD  fumarate + E-FADH2 (succinase dehydrogenase), oxid.
7. fumarate + H2O  malate (fumerase), hydration
8. malate + NAD+  OAA + NADH (malate dehydrogenase), ox.
Yield of ATP form the complete 3 NADH x 2.5 + 1 FADH2 x 1.5 = 9 ATP + 1 ATP = 10 ATP
oxidation of pyruvate or of acetyl CoA
Summarize the regulation of the (-) acetyl CoA (bind E2), NADH (inhibits E3), ATP – energy needs of cell are met,
pyruvate dehydrogenase complex fatty acids are being degraded, no need for pyruvate to acetyl CoA
through reversible phosphorylation Phosphorylation (kinase, associated with E2) of E1 switches off activity
Major activators and inhibitors of the Dephosphorylation (phosphatase) of E1 switches on activity
kinase and phosphatase (+) ADP, pyruvate, Ca2+, epinephrine
Control points of the CAC, activators Isocitrate dehydrogenase: (+) ADP, (-) ATP, NADH
and inhibitors α –ketoglutarate dehydrogenase: (-) ATP, NADH, succinyl CoA
Build up? Inhibited isocitrate DH = buildup of citrate, transported to cytoplasm to stop
glycolysis, source of Acetyl CoA for FAs (ATP-citrate lysase)
CAC intermediates that can be used as OAA: Glucose, aspartate, amino acids, purines, pyrimidines
biosynthetic precursors Citrate: Fatty acids, sterols
α-ketoglutarate: glutamate, other amino acids, purines
Succinyl CoA: porphyrins, hem, chlorophyll
Roles of anaplerotic reaction Replenishes pathway components
Pyruvate carboxylase reaction Pyruvate carboxylase produces OAA from pyruvate for CAC
Consequences and biochemical basis of Beriberi – no thiamine (vitamin B), precursor of TPP (cofactor of prosthetic group of
thiamine deficiency. pyruvate, α-ketoglutarate DH, transfer activated aldehyde unit), neurons use only
glucose, so enzyme deactivation = no fuel, other tissues can use fats in CAC
Compare the effects of heavy-metal Mercury + arsenite have high affinity for neighboring sulfhydryls (those in
poisoning with mercury or arsenite dihydrolipoyl groups of E3, inhibits complex = CNS disease.
Chapter 18: Oxidative Oxidative Phosphorylation: process in which ATP is formed as a result of transfer of
Phosphorylation electrons from NADH or FADH2 to O2 by a series of electron carriers in IMM
Define oxidative phosphorylation and Respiration: the release of energy from glucose inside living cells
respiration
Energy yield from glycolysis and 4 ATP, and 10 NADH + 2 FADH2
CAC?
The coupling of ____ by a ___ (____) The coupling of oxidation of fuels to phosphorylation of ADP by a protein
forms ATP gradient (proton-motive force) forms ATP
Energy transfers of CAC, oxidative CAC: produce electrons with high transfer potential
phosphorylation Ox phos: electron-motive force converted to proton-motive force, then to a
phosphoryl-transfer potential.
Respiratory chain 3 electron-driven proton pumps (NADH-Q oxidoreductase, Q-cytochrome c
oxidoreductase, and cytochrome c oxidase) convert electron-motive force into
proton-motive force. Contain quinones, flavins, iron-sulfur clusters hemes, and Cu2+.
Compartments and membranes of OMM: has mitochondrial porin, permeable to small molecules
mitochondria, respiratory assemblies IMM: impermeable to ions/polar molecules
and the N and P sides of the inner Transporter shuttles: ATP, pyruvate, citrate
membranes Matrix side (-), cytoplasmic side 9P)
Quantitatively relate redox potential ∆ G0’ = -nF∆ E0’ (n = # electrons, F = 96.48 kJ/mol V)
(∆ E0’) and free energy change (∆ G0’) ∆ G0’ unit is kJ or kJ/mol, ∆ E0’ unit is volts
Redox potential (∆ E0’) of a redox Measure electromotive force generated by a sample half-cell connected to a standard
couple relative to the standard reference cell. If electrons flow to standard reference, reduction potential is negative.
reference half-cell Reducing agent (NADH) = negative, Oxidizing agent (O2) = positive reduction pot.
Free energy stored in concentration ∆ G = 2.303RT log10(C2/C/1) + ZF∆ V
gradients C2 = [destination], C1 = [origin], R = 8.315E-3 kJ/mol, Z = charge of species, ∆ V =
membrane potential
Calculate energy of proton gradient ∆ G = 2.303RT log10(C2/C/1) + ZF∆ V
If pH is 1.4 units lower outside than inside, log10(C2/C/1) = 1.4 (more H+ outside)
Components of the respiratory chain I. NADH-Q oxidoreductase
and electron-carrying molecules II. Succinate-Q reductase (contains succinate DH, generates FADH2)
III. Q-cytochrome c oxidoreductase
IV. cytochrome c reductase
I, III, IV: e- flow through transmembrane complex  transport of H+ across IMM
Describe the entry of electrons from Electrons carried by reduced form of coenzyme Q (ubiquinone, hydrophobic,
NADH into NADH-Q oxidoreductase diffuses rapidly in IMM, five isoprene unit tail)
(Complex I) [Cytochrome c (small soluble protein) shuttles electrons from Q-cytochrome c
Trace electron path through proton oxidoreductase  cytochrome c reductase (reduces oxygen)]
pump NADH binds complex I, transfers 2 high potential electrons to FMN prosthetic
What groups transfer electrons? groups  Fe-S clusters (undergo oxidation-reduction w/o proton exchange)  Q
*4 H+ pumped out of matrix of mitochondria, Q accepts 2 electrons = QH2, leaves in
hydrophobic interior of the membrane
Role of coenzyme Q as a mobile Quinone can exist in 3 oxidation states
electron carrier between NADH-Q Q = fully oxidized, two ketones
oxidoreductase and cytochrome QH· = 1 e- and 1 H+, semiquinone, Q·- = lose hydrogen, semiquinone radical anion
reductase (Complex III) QH2 = 2 e- and 2 H+, ubiquinol (fully reduced)
Q pool: a pool of Q and QH2 in IMM
Describe the entry of electrons into the CAC: succinate  fumarate (succinate DH), part of succinate-Q reductase complex
respiratory chain at the succinate-Q FADH2 electrons transferred to Fe-S centers  Q  ETC
reductase complex (Complex II) from *does not transport protons, less ATP is made from FADH2 than NADH
Is Complex II a proton pump? No.
The prosthetic group and functions of Cytochrome water-soluble, electron-transferring protein w/ a heme prosthetic grp.
cytochromes. (Iron ion reduced ferrous +2 state  ferric +3 state)
Contrast features of cytochromes b, c1, cyt b, c1, cyt c: iron-protoporphyrin IX (like Mg and HB), different electron affinities
c, a, and a3 (cytoplasmic side = lower affinity for electron than matrix)
Components of the Q-cytochrome c 3 heme groups = b and c + iron-sulfur protein (Rieske center), stabilizes reduced
oxidoreductase complex. center so it can accept electrons from QH2
How does ubiquinol transfer its Cycle a: 1st QH2 molecule binds complex, gives up 2 electrons (one to cyt c1  cyt
electrons to cyt c1 and b and ultimately c, one to bound Q  Q·-) and releases two protons New Q enters Q pool.
to cyt c? Cycle b: 2nd QH2 gives up electrons (1 to cyt c, another to reduce Q·- to QH2). And
Role of heme? takes up two protons from matrix.
1 cycle = 2 QH2 molecules oxidized, 1 Q molecule reduced to funnel electrons from
two=electron carrier to 1 electron carrier (cyt c). Cyt b = a recycling device in
reductase that enables both electrons of QH2 to be used effectively.
How does a two-electron carrier QH2 electron flows to Rieske 2 Fe-2S cluster  cyt c1  cyt c.
(ubiquinol) interact with a one-electron
carrier (Fe-S cluster)
Describe the cytochrome oxidase Transports electrons from cyt c to molecular oxygen
proton pump (Complex IV) and its 4 electrons funneled to O2 to produce H2O, 4 protons pumped to cytoplasmic side of
electron carrying groups IMM, ∆ G°’ = -231.8 kJ, energy captured as proton gradient, used in ATP synthesis
Cyc c oxidase has 2 heme A groups, 3 copper ions: CuA/CuA (accepts electrons from
reduced cyt c), CuB coordinated by 3 histidine residues (alternates between cuprous
Cu+ and Cu2+ cupric form when accepting electrons.)
**Heme a3 (Fe) and CuB is active center where O2 is reduced to H2O.
Heme A in cyt c oxidase different from cyt c because heme is not covalently
attached to the protein.
Outline the mechanism for the 1. Electrons from 2 cyt c  CuA/CuA  Heme a  CuB and heme a3.
reduction of oxygen to water on 2. O2 binds and abstracts an electron from each nearby ion = peroxide bridge.
cytochrome oxidase 3. Two more cyt c bind and release electrons that travel to active center. Two
Describe the path of the electrons protons cleave the peroxide bridge.
4. Addition of two more protons leads to release of water.
Chemiosmotic model of oxidative Chemiosmotic hypothesis: electron transport and ATP synthesis are coupled by a
phosphorylation and relate proton gradient across the IMM, protons flow back into matrix and equalize
experimental evidence that only the concentration gradient. Proton motive force = pH and charge gradient. pH is 1.4
proton-motive force links the units lower inside, membrane potential is .14 V (outside positive).
respiratory chain ad ATP synthesis. Test: Synthetic vesicle with bacteriorhodopsin (light-driven proton pump) +
mitochondrial ATP synthase. Exposed to light, ATP was formed, showed respiratory
chain and ATP synthase are biochemically separate systems linked only by a proton-
motive force.
Mitochondrial location, subunit ATP synthase = Complex V.
structure, and function of eukaryotic Fo subunit: in IMM “stick”: proton channels of complex, 10-14 c subunits
ATP synthase embedded in membrane. A subunit binds outside of the ring.
F1 subunit: “ball” = catalytic ATPase activity: α3β 3, central stalk: ε + γ (breaks
symmetry), extends into α3β 3 hexamer, δ
2 ways Fo and F1 are connected: by central γ ε stalk + exterior column
Exterior column = a subunit, 2 b subunits and δ subunit (prevent hexamer
rotation)
Moving = c ring + γ ε stalk, stationary: hexamer
Mechanism of ATP synthesis by ATP ATP forms in absence of proton-motive force, but doesn’t leave catalytic site unless
synthase during proton flow. proton flows through enzyme. Proton gradient helps release ATP from synthase.
Catalytic cooperativity’s relation to Binding-change mechanism: (Boyer) β subunit performs 3 sequential steps in
binding-change mechanism? ATP synthesis by changing conformation:
1. ADP + Pi binding: loose conformation
2. ATP synthesis: tight conformation, bind ATP so tightly ADP and Pi are converted
3. ATP release: open conformation (LTO)
Evidence for rotational catalysis System with only α3β 3γ subunits, β subunit has amino-terminal polyhistidine tag
(high affinity for nickel ion), immobilized on glass surface coated with nickel. γ-
subunit linked to fluorescently labeled actin filament (long stem that can be observed
under fluorescence microscope). Add ATP causes actin filament to rotate
unidirectionally in a CCW direction. γ subunit rotation driven by hydrolysis of ATP.
Catalytic activity of an individual molecule observed. Lower [ATP] showed γ
subunit rotates in 120 ° increments = hydrolysis of 1 ATP molecule.
How does proton flow from Fo drive a subunit = 2 hydrophilic half-channels that do not span membrane, next to
the rotation of the γ subunit? membrane spanning ring of c subunits
c subunit = a pair of α helices that span the membrane, aspartic residue found in
middle of one of the helices.
1. proton rich environment: proton enters a channel and binds the aspartate residue.
2. Subunit w/ bound proton (neutralized) rotates through membrane, occupies the
hydrophobic environment of the membrane.
3. Proton-poor environment: aspartic acid releases proton.
**c ring is tightly linked to γ and ε subunits. As c ring turns, stalk turns inside
hexamer of F1, promotes binding-change mechanism.
ATP yields for NADH, FADH2 NADH = 2.5 mol of ATP, FADH2 = 1.5 mol of ATP

Is IMM permeable to NADH? IMM impermeable to NADH and NAD+. Electrons from NADH enter the matrix via
Implications? shuttles (Malate-aspartate shuttle, Glycerol 3-phosphate shuttle)
ATP-ADP translocase mechanism and ATP-ADP translocase = transport protein (antiporter) that couples ATP and ADP
energy cost flow. Positive membrane potential increases rate eversion from matrix to cystolic
In an actively respiring mitochondrion, side when ATP is bound. ¼ of energy yield from electron transfer by respiratory
chain is consumed to regenerate membrane potential tapped by this process.
Net yield of ATP from the complete Glycolysis:
oxidation of glucose, different shuttles Phosphorylation of glucose -1
for the cytoplasmic reducing Phosphorylation of F6P -1
equivalents of NADH Dephosphorylation of 2 molecules of 1,3 BPG +2
Dephosphorylation of 2 molecules of PEP +2
Oxidation of 2 molecules of glyceraldehyde 3-phosphate 2 NADH
Pyruvate  Acetyl CoA 2 NADH
CAC:
2 molecules of Succinyl CoA  Succinate +2
Oxidation of 2 molecules of isocitrate, α-ketoglutarate, malate 6 NADH
Oxidation of 2 molecules of succinate 2 FADH2
Oxidative Respiration
2 NADH from glycolysis x1.5 +3
2 NADH from pyruvate  acetyl CoA x 2.5 +5
2 FADH2 from CAC +3
6 NADH from CAC x 2.5 +15
Net ATP yield/molecule of glucose 30 ATP
Source of uncertainty in estimation Stoichiometries of proton pumping, ATP synthesis and transport processes (glycerol
3-phosphate shuttle vs. malate –aspartate shuttle) do not have fixed values.
Respiratory control Electrons only flow through ETC to O2 if ADP is simultaneously phosphorylated to
Relation to energy charge ATP. ↑ ADP (active muscle), ox phos ↑
Respiratory control: regulation of rate of ox phos by ADP level
Compounds that block electron 1. Block Electron transport:
transport and locate sites of action Rotenone, amytal block electron transfer in NADH-Q oxidoreductase
Antimycin A – interferes with electron flow from Q-cyt c oxidoreductase  cyt c
CN-, azide, CO can block electron flow from cyt c  O2
2. Inhibit ATP synthase: Oligomycin, DCCD
3. Uncouple electron transport from ATP synthesis: DNP (carry proton across IMM
down concentration gradient, dissipates proton-motive force = heat, no energy)
4. Inhibit ATP export: atractyloside, bongkrekic acid inhibit ATP-ADP translocase
Other uses of proton gradients Active transport of Calcium ions by mitochondria, entry of aa and sugars into
bacteria, rotation of bacterial flagella, transfer of electrons from NADP+ to NADPH,
generate heat in hiberantion
Chapter 21: Glycogen Metabolism Glycogen: branched polymer of glucose, mostly α-1,4 glycosidic linkages and α-1,6
Structure of glycogen, location? glycosidic linkages (branch point) α-linkages = open helical polymers
β linkages: straight strands (fibrils/cellulose), Liver (10%), skeletal muscle (2%)
Three steps of glycogen catabolism, 1. Glycogen  G1P (glycogen phosphorylase)
fate of its product, glucose-6-phosphate 2. remodel glycogen to permit further degradation
3. G1P  G6P (phosphoglucomutase)
Glucose (Glucose 6-phosphatase in liver), glycolysis, pentose phosphate pathway
Precursors of glycogen anabolism UTP + G1P, UDP-glucose is added to nonreducing ends of glycogen
Phosphoglucomutase mechanism phosphate on enzyme’s Ser transferred to G1PG1,6,BPG6P(~PG mutase)
Role of hormones in regulating Epinephrine, glucagon ↑ breakdown, ↓ synthesis
glycogen metabolism Insulin ↓ breakdown, ↑ synthesis
Reaction catalyzed by glycogen Cleaves α-1,4-linkage using orthophosphate  (phosphorolysis), removes glucosyl
phosphorylase residues from nonreducing end (b/t C-1 and C4), stops 4 residues from branch point
Rxn driven by? Phosphorolysis is favored by high [Pi]:[G1P] ratio > 100
Advantage of Phosphorolytic cleavage Phosphorolytic cleavage of glycogen gives a phosphorylated sugar that can go down
of glycogen over hydrolytic cleavage glycolytic pathway. No transporters exist for G1P in muscle cells, stays in cells.
Steps in the degradation of glycogen Transferase shifts block of three glucosyl residues from one outer branch to other.
Α-1,6-glucosidase hydrolyzes the α-1,6-glycosidic bond.
**α-1,6-glucosidase and transferase are on one polypeptide chain.
Importance of glucose 6-phosphatase in Liver’s job is to keep a constant level of glucose in blood, glucose 6-phosphatase
the release of glucose by the liver enables glucose to leave liver. Absent in brain and muscle because retain glucose for
Absent in __ and ___. the generation of ATP (main source of fuel) In contrast, liver uses α-ketoacids as
Why this tissue distribution main source of fuel.
Roles of pyridoxal 5’-phosphate, ____ 5’ phosphate group of PLP acts in tandem with orthophosphate by serving as a
catalysis proton donor and then a proton acceptor (general acid-base catalyst)
Processivity? How does it relate to Processivity = enzyme that can catalyze many reactions without having to
glycogen phosphorylase activity disassociate and re-associate after each catalytic step.
Glycogen binding site is 30 Å away from catalytic site, connected by a narrow
crevice that accommodates 4-5 glucose units. Enzyme can phosphorolyze many
residues without having to dissociate/re-associate.
Two primary regulatory mechanisms Intxns w/ allosteric effectors (signal energy state)/reversible phosphorylation
for glycogen phosphorylase (response to hormones), isozyme: differ in catalytic properties/response to regulator
Liver enzyme (a/R form) is allosterically inactivated by glucose (T form) –
Difference in liver and muscle produces glucose for use by other organs/tissues, conserves glycogen when glucose
isozymes of glycogen phosphorylase is plentiful, a-form is phosphorylated by phosphorylase kinase, hormone: glucagon
Muscle enzyme (b/T form is predominant) ATP, G6P are negative allosteric factors
that stabilizes T state, dephosphorylated by PPI, hormone: epinephrine
Phosphorylase kinase regulation Activated by Ca2+ in δ calmodulin and phosphorylation of β subunit by PKA
Hormone 7TM receptor  G protein activates adenylate cyclase ATP to cAMP
Role of G-proteins in covalent  activate PKA  activates phosphorylase kinase  activates phosphorylase a
modifications α –adrenergic receptor initiates phosphoinositide cascade (induces release of Ca2+
from ER)

Phosphorylation of phosphorylase by phosphorylase kinase


___ and dephosphorylation by ____ protein phosphatase 1 (PP1)
Molecular bases for the relative T state is less active because catalytic site is partly blocked. R state – catalytic site is
inactivity of the T state more accessible.
Steps in the synthesis of glycogen, 1. Priming (Glycogenin = dimer of α-1,4-glucose residues, catalyzes addition of 8
pertinent enzymes glucose units from UDP-glucose to other subunit in dimer = autoglycosylation,)
2. UDP-glucose + Glycogenn  UDP + Glycogenn+1 (glycogen synthase)
3. branching enzyme moves blocks of 7 residues to interior site, α-1,6 linkages
Functional importance of branching in Increases the solubility of glycogen, creates a large number of terminal residues (site
the glycogen molecule of action of glycogen phosphorylase and synthase = ↑ rate of synthesis/degradation)
Efficiency of glycogen as a storage 1 ATP hydrolyzed to incorporate G6P into glycogen (activate G1P = synthesis)
form of glucose 10% : 1 ATP used to phosphorylate glucose molecules to G6P. (breakdown)
Complete oxidation of G6P = 31 molecules of ATP
Overall efficiency = 97%
Regulation of glycogen synthase. Active Glycogen synthase a: dephosphorylated (active w/ or w/o G6P)
Effects of phosphorylation on glycogen Inactive Glycogen synthase b: phosphorylated (active only with high G6P)
synthase and glycogen phosphorylase Phosphorylation has the opposite effect on glycogen phosphorylase
Role of protein phosphatase I in control PPI dephosphorylates, inactivates phosphorylase a + phosphorylase kinase, ↓
of the activities of glycogen glycogen breakdown + activates glycogen synthase b  a, ↑ glycogen synthesis
phosphorylase and glycogen synthase. Catalytic unit of PPI bound to GM/GL regulatory unit (scaffolds that bring
How is it regulated? phosphatase + glycogen). To ↓ PPI activity during glycogen degradation,
Epinephrine/glucagon  cAMP  PKA activated 
1. GM phosphorylated and releases catalytic subunit
2. inhibitor phosphorylated and inactivates PPI catalytic subunit.
To ↑ PPI activity during glycogen synthesis, Insulin  activated Tyr Kinase 
Protein Kinase phosphorylates RG1 of PPI, which associates with glycogen.
Effects of insulin on glycogen. Why Insulin stimulates the synthesis of glycogen
does a distinct pathway exist for the insulin binds receptor tyrosine kinase  phosphorylates IRSs  protein kinases
biosynthesis and degradation of phosphorylate and inactivate glycogen synthase kinase  glycogen synthase no
glycogen? longer phosphorylated (also dephosphorylated by PPI = restore glycogen reserves)
Enables both pathways to be thermodynamically favorable at all times. Biosynthetic
pathway is exergonic by coupling to hydrolysis of a sufficient # of ATP molecules.
After meal, what happens to Phosphorylase a is the glucose sensor in liver cells, when glucose binds, shifts from
phosphorylase a and glycogen synthase R  T form. And the participation of phosphorylase a and PP1 in glucose sensing.
b/c of the presence of glucose in the Ab results in release of PPI, free to activate glycogen synthase and
liver? dephosphorylate glycogen phosphorylase. **binding of phosphatase to
phosphorylase a prevents premature activation of glycogen synthase
Provide examples of glycogen storage Pt with hypoglycemia (glycogen storage disease, low blood-glucose between meals),
diseases; relate the biochemical defects due to lack of glucose 6-phosphatase from liver, glucose cannot be formed from
with the clinical observations. G6P. Excess G6P in liver causes glycolysis = high level of lactate and pyruvate in
blood. Pts depend on fat metabolism. –OR – Mutation in glucose 6-phosphate
transporter – transport G6P to lumen of ER to be hydrolyzed by phosphatase
Lack α-1,6-glucosidase can only use outermost branches of glycogen. Only fraction
of glycogen is active as a store of glucose
Lack muscle phosphorylase – limit pt’s ability to perform strenuous exercise, pH
usually drops in muscles because of lactate. McArdle disease = alkaline because of
breakdown of Creatine phosphate. Low glycolytic rate
Chapter 22: Fatty Acid Metabolism Fatty acid = long hydrocarbon chain + terminal carboxylate group
Define Fatty acid 1. fuel molecules, stored as TAGs, and mobilized to meet energy needs, insulation
Four major physiological functions 2. Fas are building blocks of phospholipids and glycolipids (biological membranes)
they serve 3. Proteins modified by FA are targeted to a membrane location
4. FA derivatives = hormones and intracellular messengers

Structure of a triacylglycerol Uncharged esters of Fas + glycerol


How are lipids transported? Free fatty acids and monoacylglycerols are absorbed by intestinal epithelial cells,
resynthesized into TAGs, and packaged into chylomicrons. Chylomicrons are
released into lymph system, bind lipases, degraded into FA and monoacylglycerols
and transported into tissue. TAG resynthesized in cell and stored or oxidized to
provide energy in muscle.
Overview of degradation of fatty acids, 1. FA activated
list chemical reactions used 2. Oxidation to introduce a double bond
3. Double bond is hydrated to introduce a hydroxyl group
4. Alcohol is oxidized to a ketone
5. FA thiolysis by CoA  Acetyl CoA + FAn-2 [OHOT]
Overview of synthesis of fatty acids, 1. Condensation of activated acyl group + malonyl unit  4C
list chemical reactions used 2. Caronyl group reduced
3. Dehydration
4. Reduction  butyryl CoA [CRDR]
Why are TAGs highly concentrated They are reduced and anhydrous (nonpolar)
forms of stored metabolic energy
Adipocyte Adipocyte: fat cell that store triacylglycerols, hormones
Chylomicron Chylomicrons: carry fatty acids from intestine to other tissue
Bile Salts Bile salts: amphipathic molecules synthesized from cholesterol I the liver and
Lipases secreted from gall bladder. Incorporate TAG into micelles
Lipase: intestinal enzymes secreted by pancreas that degrade TAGs to free Fas and
monoacylglycerol.
Role of cAMP in regulation of lipase in Lipases activated by glucagon, epinephrine, and adrenocorticotropic hormone via
adipose cells 7TM signal transduction cascade, inhibited by insulin.
Serum album carries ___ Glucagon/epinephrine  7TM receptor in adipose tissue  activate adenylate
cyclase  increase cAMP  stimulate PKA  activate lipase by phosphorylation
Serum album carries fatty acids from adipocytes to other tissues.
Reactions of Glycerol Liver absorbs glycerol  glycerol 3-phosphate  DHAP (glycolysis/gluconeogenesis)
Physiological importance of these Glycerol can be converted into pyruvate or glucose in liver (interconvertible
reactions glycerol and glycolytic intermediates)
Reaction that links CoA to a fatty acid FA + ATP ↔ Acyl Adenylate (enzyme bound) + Ppi
Acyl Adenylate + HS-CoA ↔ Acyl CoA + AMP
Where located? Enzyme? Location: outer mitochondrial membrane, catalyzed by Acyl CoA synthetase
Involvement of carnitine in transport of FA must be oxidized in mitochondrial matrix.
fatty acids from cytoplasm into Acyl CoA + Carnitine  Acyl carnitine (carnitine acyltransferase I – bound to
mitochondria outer mitochondrial membrane)
Acyl carnitine shuttled across inner mitochondrial membrane by translocase
Acyl Carnitine  Carnitine + Acyl CoA (carnitine acyltransferase II )
Four reactions of the β -oxidation 1. ox: acyl CoA + FAD  trans ∆ ²-Enoyl CoA + FADH2 (acyl CoA DH)
pathway of fatty acid catabolism, 2. hyd: trans ∆ ²-Enoyl CoA + H2O  L-3-hydroxyacyl CoA (enoyl CoA hydratase)
substrates and products 3. ox: L-3-hydroxyacyl CoA + NAD+ 3-ketoacyl CoA + NADH (L-3-H CoA DH)
Function of NAD+ and FAD in 4. thiolysis: 3-ketoacyl CoA + HSCoA  Acyl CoAn-2 + Acetyl CoA (β -
reactions ketothiolase)
Different Acyl CoA dehydrogenases C12 – C18 FA oxidized by long chain Acyl CoA dehydrogenase
C4 – C14 FA oxidized by the medium chain Acyl CoA dehydrogenase
Energy yield in ATP molecules for β palmitoyl CoA (C16)
oxidation of palmitoyl CoA 108 – 2 (ATP to activate palmitate) = 106 ATP/1 molecule palmitate
Two reactions used to oxidize naturally *db bond is not a substrate for Acyl CoA dehydrogenase – prevent formation of a
occurring unsaturated fatty acids C2-C3 db bond.
1. isomerase
2. reductase
Oxidation reactions of odd-numbered Yields Propionyl CoA in final thiolysis step
fatty acid Propionyl CoA  Succinyl CoA (Propionyl CoA carboxylase) to enter CAC
When can acetyl CoA enter CAC? If Can oxidize Acetyl CoAs in CAC only when balanced with carbohydrate
not? degradation. Otherwise, form Ketone Bodies.
Name and structure of ketone bodies, When fasting, starving, diabetes, OAA  glucose and is unavailable for
Where produced? Why available to condensation with acetyl CoA. Acetyl CoA diverted to form D-3-hydoxybutyrate
other organs? and acetoacetate (ketone bodies) Ketone bodies produced in liver. Liver lacks CoA
transferase
Describe the synthesis of the ketone 2 Acetyl CoA  CoA + Acetoacetyl CoA (thiolase)
bodies. Acetoacetyl CoA + H2O + acetyl CoA  HMG-CoA + CoA (HMG synthase)
HMG-CoA  Acetoacetate (HMG CoA cleavage enzyme) + acetyl CoA
The role of ketone bodies in normal Acetoacetate  D-3-Hydroxybutyrate (D-3 DH) + Acetone (decarboxylation)
human metabolism Ketone bodies = water-soluble, transportable form of acetyl units
Favorite source of fuel for heart muscle + renal cortex, brain during starvation
Describe the catabolism of the ketone Acetoacetate + Succinyl CoA  Acetoacetyl CoA + Succinate (CoA Transferase)
bodies. Acetoacetyl CoA + CoA  2 Acetyl CoA (Thiolase)
The effect of high levels of acetoacetate High levels of acetoacetate in blood signify an abundance of acetyl units, leads to a
on fat metabolism in adipose tissue decrease in the rate of Lipolysis in adipose tissue.
Biochemical basis for the inability of Pyruvate + CoA + NAD+  acetyl CoA + CO2 + NADH + H+ = Irreversible!
animals to convert fatty acids into 2 C’s of acetyl group enter CAC, but 2 C leave CAC in decarboxylations, OAA is
glucose regenerated, not formed de novo when acetyl unit is oxidized by CAC.
Plants have enzymes that can convert acetyl CoA  OAA
Contrast fatty acid oxidation and fatty FA Synthesis FA Degradation
acid synthesis Cytoplasm Mitochondrial matrix
Intermediates linked to sulfhydryl Covalently attached to sulfhydryl
groups of ACP (acyl carrier protein) groups of CoA
Enzymes joined in 1 ppt chain: fatty Enzymes not associated
acid synthase
Activated donor = malonyl ACP, Activated donor: Acyl CoA
driven by release of CO2
Reductant is NADPH Oxidants are NAD+ and FADH
Elongation stops at palmitate (C16) Degradation stops at Acetyl CoAs
Substrates and products of the 1. Committed step: Acetyl CoA + HCO3 + ATP  Malonyl ACP + ADP + Pi + H+
-

committed step in fatty acid synthesis (Acetyl CoA carboxylase – contains biotin covalently attached to ε amino of Lys)
and catalytic mechanism. 2. Acetyl CoA ↔ Acetyl ACP (Acetyl Transacylase – AT)
Role of biotin in the acetyl CoA 3. Malonyl CoA + ACP ↔ malonyl ACP + CoA (Malonyl Transacylase)
carboxylase reaction 4. Condensation: Acetyl ACP + malonyl ACP  acetoacetyl ACP + ACP + CO2
(Acyl-malonyl ACP condensing enzyme)
The four reactions of the elongation 5. Reduction: Acetoacetyl ACP + NADPH + H+ ↔  D-3-hydroxybutryl ACP +
cycle of fatty acid synthesis NADP+ (β -ketoacyl ACP reductase)
6. Dehydration: D-3-hydroxybutryl ACP ↔ Crotonyl ACP + H2O (3-hydroxyacyl
ACP dehydratase)
7. Reduction: Crotonyl ACP + NADPH + H+  butyryl ACP + NADP+ (enoyl ACP
reductase)
Round Two of synthesis Butyryl ACP + malonyl ACP  C6 ACP
End of FA synthesis?  C16 ACP (good substrate for thioesterase, hydrolyzes C16 ACP 
palmitate + ACP)
Domains of Animal Fatty Acid Domain 1: substrate-entry and condensation unit:
Synthase multifunctional enzyme Acetyl transferase, malonyl transferase, β -ketoacyl synthase/condensing enzyme
complex. Domain 2: reduction unit: acyl carrier protein, β -ketoacyl reductase, dehydratase,
and enoyl reductase
Advantages Domain 3: palmitate release unit: thioesterase

(+) coordinate different reactions, efficient intermediate transfer from sites without
leaving assembly, enzymes more stable when covalently joined
Compare ACP and CoA Both have a phosphopanthetheine as reactive units
Malonyl CoA provides ___ Malonyl CoA provides the driving force for the condensation of acetyl units with the
growing acyl chain by releasing CO2
The energy cost of the synthesis of a 8 molecules of Acetyl CoA, 15 molecules of NADPH, and 7 molecules of ATP
Palmitate
Enzymatic machinery for FA synthesis Eukaryotic synthases are linked in 1 polypeptide chain, forms a dimer
in bacteria vs. eukaryotes
Describe the transport of acetyl groups Fas made in cytoplasm, acetyl CoA made from pyruvate in mitochondria.
across the inner mitochondrial To transfer acetyl CoA from to cytoplasm, citrate carries acetyl groups across IMM.
membrane, why not use carnitine? Acetyl CoA + OAA  citrate, in cytoplasm, citrate is cleaved by ATP-citrate lyase
(cleave C-C, C-O, or C-N bonds by elimination, forms double bond), cost 1 ATP
Carnitine only carries long-chain FA in degradation.
When is NADPH synthesized? 1. OAA +NADH + H+ ↔ malate + NAD+
2. Malate + NADP+  pyruvate + CO2 + NADPH (pyruvate can enter mitochondria,
carboxylated to OAA by pyruvate carboxylase)
*One molecule of NADPH is generated for each molecule of acetyl CoA that is
transferred from mitochondria to the cytoplasm.
Sources of NADPH used in fatty acid Pentose phosphate pathway
synthesis, pathways of FA synthesis CAC, transport of OAA from mitochondria, pentose phosphate pathway = carbon
atoms + reducing power
Glycolysis + Oxidative phosphorylation provide ATP
Different modes of regulation of acetyl Synthesis: acetyl CoA + ATP + HCO3  malonyl CoA + ADP + Pi + H+ =
CoA carboxylase committed step must be regulated (acetyl CoA carboxylase)
Reciprocal control of fatty acid (+) citrate – acetyl CoA and ATP are abundant
synthesis and degradation through local (+) insulin
(intracellular), hormonal, and adaptive (-) palmitoyl CoA – excess FA
regulation (-) glucagon
Degradation: Malonyl CoA inhibits carnitine acyltransferase I (prevent acyl CoAs
from entering mitochondrial matrix)
Chapter 23: Amino Acid Metabolism Cannot be stored (like glucose/FA) nor excreted. Must be used as metabolic fuel. α-
Fate of exogenously supplied amino amino group removed, carbon skeleton converted into major metabolic intermediate.
acids that are not use for biosynthesis Amino group can go through urea cycle
Where do amino acids come from? Amino acids come from 1. digestion (diet) – low pH + enzymes:
What are they used for? Stomach – pepsin, Pancreas: chymotrypsin, rypsin, carboxypeptidase, elastase
2. degradation (turnover) of cellular proteins (damaged by oxidation, errors in
translation, misfolded, changing metabolic needs, unneeded and ubiquitinated)
Used to make proteins or nitrogenous compounds (nt bases)
Half-lives of proteins in mammals Ornithine decarboxylases – 11 min, Hb – red blood cell, Crystalline – life of
organism
Define Ubiquitin Ubiquitin: protein attached to the ε-amino group of lysine residues of proteins to be
Function of E1, E2, E3 that participate degraded (isopeptide bond)—energy from ATP hydrolysis
in the attachment of ubiquitin to E1: Ub-activating enzyme (adenylates Ub, transfers Ub to its Cys by thioester bond)
proteins targeted for degradation E2:Ub-conjugating enzyme (transfer Ub to its own Cys residue)
E3: Ub-protein ligase (read N-terminal, transfer Ub to Lys residue on target protein)
N-terminal rule Half life of a cytoplasmic protein is determined by its amino-terminal residue
Stabilizing residues (t½ > 20 mins) Met, Pro
Destabilizing residues (t½ < 2 mins) Arg, Leu
Describe the fates of the peptides Broken into amino acids that are then broken into:
derived from a ubiquinated protein 1. amino group  urea cycle
2. carbon group  glucose/glycogen synthesis, cellular respiration, FA synthesis
3. stay intact for biosynthesis
Markers for destruction Ubiquitination
Cyclin destruction boxes – aa sequences that marks cell-cycle proteins for
destruction
PEST sequence – proline, glutamic acid, serine, threonine
N terminal
What digests the Ub-tagged proteins? The 26S proteasome = 20S catalytic unit (α2β 2) + 19S regulatory unit (binds poly-
Ub chains, ATP hydrolysis to unfold substrate, induce conformational changes in
20S to pass substrate through center)
recycles Ub
Two types of intracellular proteases 1. Sequestered proteases: lysosomal cathepsin (active at low pH), ER proteases
(degrade proteins that fold incorrectly), 2. Regulated proteases: calpain
Describe proteolysis within 20S core Contains an N-terminal Thr whose –OH group is nucleophilic, attacks carbonyl
particle bonds in protein to form acyl-enzyme intermediate (~ chymotrypsin), MHC I
Major organ responsible for amino acid Liver, muscle also degrades branched aa (Leu, Ile, Val)
degradation in mammals
Describe the two steps to remove In Liver
nitrogen from AA in ____ 1. α-aa + α-ketoglutarate  α ketoacid + glutamate (aminotransferase/transaminase)
2. glutamate + NAD+ + H2O α-ketoglutarate + NADH + NH4+ (glutamate dh)
How is glutamate dehydrogenase Direction determined by concentration of R and P, reaction usually driven toward α-
regulated? ketoglutarate because of rapid removal of NH4+ into urea cycle
Essential cofactor for many enzymes of Pyridoxal phosphate (PLP) = prosthetic group of aminotransferases, derivative of
aa metabolism (Dietary precursor?) vitamin B2, has ability to tautomerize
Structure of pyridoxal phosphate (PLP) Functional group: aldehyde (forms Schiff base with amino group of amino acids)
and pyridoxamine phosphate (PMP) allows α-amino groups to be shuttled between amino acids and keto acids.
Reactive functional group Internal  external aldimine, resonance stabilized carbanion, hydrolysis to PMP.
Transport of nitrogen from muscle to Muscles transport ammonium ions as alanine or glutamine.
liver
Fate of NH4+ Used to synthesize purine and pyrimidine bases
Excess excreted as NH+ in aqueous animals
Excreted as uric acid (remove 4N) in birds/reptiles, excrete more N + use less water
Excreted as urea in terrestrial vertebrates (ureotolic), secreted in solution in urine
Where is urea synthesized? Urea is synthesized in Liver (also major site of deamination)
How are ammonium ions transported? Ammonium ions are transported to liver as alanine or glutamine
Molecules that bring nitrogen and 1 Nitrogen from aspartate, 1 Nitrogen from free ammonium
carbon into the urea cycle Carbon atom from HCO3-
Carbamoyl phosphate synthetase Enzyme: carbamoyl phosphate synthase (CPS), hydrolyze 2 ATP, irreversible rxn
mechanism 1. Bicarbonate +ATP  ADP + carboxyphosphate
ATP requirement of the urea cycle 2. carboxyphosphate + NH2  carbamic acid + Pi
3. carbamic acid + ATP  carbamoyl phosphate + ADP
Enzymes of the urea cycle, intracellular 1. Ornithine + carbamoyl phosphate 
locations citrulline (ornithine transcarbamoylase – mito
Molecular connection between urea matrix)
cycle and citric acid cycle 2. citrulline (transported to cytoplasm) +
Reaction catalyzed by each enzyme aspartate + ATP  argininosuccinate +AMP
+ Ppi (argininosuccinate synthetase)
3. argininosuccinase  arginine + fumarate
(argininosuccinase)
4. arginine + H2O  ornithine + urea
(arginase)

Structural/functional properties of two CPS I CPS II


isozymes of carbamoyl phosphate In mitochondrial matrix In cytoplasm
synthetase in mammals Function in urea cycle only Functions in pyrimidine biosynthesis,
first reaction in multi-subunit complex
Uses NH3 as substrate Uses glutamine as substrate
Allosterically activated by binding of Hydrolyzes side chain amide of
N-acetyl glutamate (made when aa glutamine to produce HN3 needed for
breakdown ↑s, signal to activate urea the first reaction, NH3 is not a direct
cycle substrate
* catalytic site in one isozyme = allosteric site in other*
Deficiencies in different enzymes of Urea = major route of ammonium removal.
the urea cycle give rise to ___, outline Block carbamoyl phosphate synthesis, or urea cycle  hyperammonemia (elevated
strategies for coping with deficiencies ammonium levels in the blood), may produce osmotic effects/brain swelling
in urea synthesis - argininosuccinase deficiency: add arginine in diet, restrict total protein intake
(arginine split into urea and ornithine, reacts with carbamoyl phosphate to form
citrulline), nitrogen secreted as argininosuccinate
-carbamoyl phosphate synthetase deficiency/ornithine transcarbamoylase
deficiency (can’t use citrulline/argininosuccinate to dispose nitrogen because
formation is impaired), excess N accumulates in glycine/glutamine.
- supplement diet with benzoate + phenylacetate
How do humans catabolize carbon Pyruvate (A, C, G, S, T, W), acetyl CoA (I, L, W), acetoacetyl CoA(L, K, P, W, Y),
atom skeletons of amino acids, name 7 α-ketoglutarate (R, Q, E, H, P), succinyl CoA (I, M, T, V), fumarate (D, P, Y),
major metabolic products formed oxaloacetate (N, D)
Basis for the glycogenic-ketogenic Ketogenic : aa degraded to acetyl CoA/acetoacetyl CoA (can be converted to ketone
designations of the amino acids and bodies/fatty acids)
classify each amino acid. Limitations of Glucogenic: aa degraded to pyruvate, α-ketoglutarate, succinyl CoA, fumarate, or
classification OAA (can be converted to PEP, then to glucose)
Chapter 26: Fatty acid and Phospholipids, spingolipids, cholesterol.
Cholesterol Metabolism Cholesterol is a membrane component and precursor of signal molecules (steroids
Three lipid based components of progesterone, testosterone, estrogen, and cortisol)
biological membranes. Biological Cholesterol is transported in blood by LDL, taken up by cells with LDL receptor on
Significance of cholesterol surface
What is apo B? Protein that transports triacylglycerols and cholesterol by forming an amphipathic
Two forms spherical shell around the lipids carried in lipoprotein particles
Apo B-48: synthesized by small intestine, carries dietary fat in the form of
chylomicrons
Apo B-100: synthesized by liver, participates in the transport of lipids synthesized in
the cell
Primary site of TAG synthesis Liver, where TAGS are transported to muscle for energy conversion, adipose cells
Transported where? Function? for storage
Major sites in cholesterol biosynthesis, 1. In cytoplasm: acetoacetyl CoA + acetyl CoA +H2O  HMG CoA + CoA:
key intermediates - mevalonate (HMG CoA reductase – integrated membrane protein in ER)
Synthetic paths leading __ or ___ *committed step*
The major regulatory enzyme in - acetoacetate (ketone body) + acetyl CoA
cholesterol biosynthesis? Mevalonate    isopentenyl pyrophosphate (C5) = activated isoprene
Conversion of mevalonate into ___ 2. In ER: isomerize isopentenyl pyrophosphate ↔ dimethyl pyrophosphate
(activated ___) dimethyl pyrophosphate + isopentenyl pyrophosphate ↔ geranyl pyrophosphate (C10)
Outline condensation reactions leading geranyl pyrophosphate + isopentenyl pyrophosphate ↔ farnesyl pyrophosphate (C15)
from isopentenyl pyrophosphate to 2 farnesyl pyrophosphate ↔ squalene (C30)
squalene 3. squalene activated by conversion into squalene epoxide
Then? Squalene epoxide cyclized to lanosterol by protonation (oxidosqualene cyclase)
Lanosterol -3 methyl groups, (19 steps) = cholesterol
Sources of cholesterol Diet or de novo synthesis in liver, intestine
Mechanism of regulation of cholesterol 1. control HMG-CoA reductase activity: phosphorylation by AMP-dependent
biosynthesis protein kinase ↓ activity [no cholesterol/FA synthesis when ATP is low]
2. control HMG-CoA reductase concentration:
- monitor blood cholesterol: scap senses cholesterol, escorts SREBP when no
sterols are in cell from ER to Golgi, S1P and S2P releases NH2 end of
SREBP = TF, SREBP dimers bind sterol response element SRE in nucleus,
enhance transcription of HMG-CoA reductase gene
- regulate translation of HMG-CoA reductase mRNA, inhibited by cholesterol and
nonsterol compounds derived from mevalonate
- regulate HMG-CoA reductase degradation (stimulated by cholesterol, sterols, and
farnesol), has an intrinsically short half life
3 fates of liver cholesterol 1. used for membrane synthesis, organ needs
2. converted to bile salts (only way to excrete cholesterol)!
3. excess cholesterol and TAG packaged into LDL and exported to blood.
How are Cholesterol and TAG Transported in the form of lipoprotein particles. Each particle has a core of
transported in body fluids? hydrophobic lipids surrounded by a shell of polar lipids/proteins. Apoproteins =
Macromolecular aggregates? 2 roles? macromolecular aggregates. Two roles: solubilize hydrophobic lipids, contain cell-
Apolipoproteins targeting signals, Apolipoproteins (protein constituents of lipoprotein particles)
synthesized and secreted by liver and intestine
Classes of lipoproteins, their lipid and Chylomicrons – carry TAG, cholesterol, and lipids from intestines (~90% TAG)
protein components *lipoprotein lipases (on blood vessel linings in muscles that use FA as fuels and
Lipid transport functions in synthesis of lipids) hydrolyze and release TAG
Chylomicron remnants – liver takes up these cholesterol-rich residues
vLDLs – mostly lipids + apo-B100 and apo-E, as they travel through blood stream,
TAG are hydrolyzed by lipoprotein lipases on capillary surfaces (muscle, adipose)
IDLs – ½ taken up by liver, ½ converted by removal of more TAG
LDLs – major carrier of blood cholesterol, core contains ~1500 esterified
cholesterol molecules surrounded by a shell of phospholipid and 1 copy of apo B-
100 (recognized by target cells)
LDL transports cholesterol to peripheral tissues to provide this compound to cells
that do not make cholesterol de novo (all except liver and intestine)
HDLs picks up cholesterol released into plasma from dying cells and membranes
undergoing turnover, acyl transferase esterifies cholesterol, returned to liver
Steps in delivery of cholesterol to cells LDL taken up by receptor-mediated endocytosis
via LDL receptor. 1. LDL binds LDL receptor
2. complex invaginated to form an internal vesicle
3. LDL-containing vesicle separates form receptor, fuses with lysosomes
4. LDL degraded, cholesterol released
Fates of released unesterified released unesterified cholesterol is used for membrane biosynthesis, esterified by
cholesterol? Danger of high acyl CoA-cholesterol acyltransferase for storage in cell
concentration? Regulation of cellular disrupts membrane integrity
functions by the LDL pathway SREBP regulates txn of LDL receptor gene regulated, blocked by high [cholesterol]
Biochemical defects of LDL receptor - Familial hypercholesterolemia: genetically defective LDL receptor synthesis, cells
that result in familial can’t take up LDL, high LDL and cholesterol in blood, cholesterol deposited in
hypercholesterolemia various tissues (skin and tendon), oxidized LDL taken up by macrophages, form
foam cells, form atherosclerotic plaques, narrow arteries.
Approaches to reduce serum - block reabsorption of bile alts
cholesterol - ↑ production of LDL receptors (remove cholesterol form circulation)
Key: ↓ [cholesterol ], ↑ txn of LDL receptor gene.
1. oral administration of positive polymers that bind negative bile salts to prevent
reabsorption of bile salts
2. inhibit cholesterol synthesis by inhibiting HMG CoA reductase (statins)
3. manage lipid intake in diet (egg, shrimp, etc)
Physiologic roles and general structures polar derivatives of cholesterol made in liver, stored in gall bladder, released in
of the bile salts small intestine, solubilize dietary lipids by forming micelles containing TAGs
Derivatives of Cholesterol Progesterone (made in corpus luteum) implant embryo, maintain pregnancy, ovarian cycle
Estrogen (made in ovaries) – develop 2° sexual chrs, maintain ovarian cycle
Androgens (made in testes) – develop 2° sexual chrs
Glucocorticoids (cortisol – made in adrenal cortex) – promote gluconeogenesis, ↑
degradation of fat and protein, inhibit inflammation
Farnesyl pyrophosphate – precursor for chlorophyll, Vitamin K, Coenzyme Q
Chapter 28: DNA 1. two polynucleotide chains coil to form a right-handed double helix
Features of Watson and Crick’s double 2. purine and pyrimidine bases are on inside of the helix, phosphate and deoxyribose
helical structure of DNA units are on the outside
3. A-T, G-C base pairs
Functions of free nucleotides 1. energy currency
2. Signaling molecules and regulators (cAMP, GTP/GDP)
3. activation of other molecules (UDP-glucose)
4. electron carriers (FAD/NAD+)
5. constituents of co-factors (CoA
Data Watson and Crick deduced 1. structure of bases and nucleotides
2. base pairing from ratios
3. x-ray diffraction data was diagnostic of a right handed helical structure, space
between nucleotide (3.4 Å).
4. fiber density indicated 2 molecules DNA per structural unit (double helix)
The role of deoxyribose puckering in A-DNA, C-3’ lies out of plane (C-3’ endo), bp tilt 11 degrees away from normal to
determining the structural differences the helix, phosphate bind fewer water molecules
between A-DNA and B-DNA B-DNA: C-2’ lies out of plane (C-2’ endo)
Compare structures of dsRNA and dsRNA and RNA-DNA hybrids have similar double-helical form as A-DNA
RNA-DNA hybrids with that of the A- RNA’s ribose 2’OH can’t form B helix because of steric hindrance (2’-oxygen
DNA helix would be too close to three atoms of adjacent 2’ phosphate group.) A type: 2’oxygen
projects outward, away from atoms
Major groove vs. Minor groove of B- Minor groove: pyrimidine O-2 and purine N-3
DNA Major groove: opposite side of the pair
Thymine’s methyl lies in major groove
B DNA: major groove is wider (12 vs. 6 Å) and deeper (8.5 vs 7.5 Å)
Larger groove makes it more accessible for interactions with proteins that recognize
specific DNA sequences
Structural information provided by x- X-ray diffraction studies (less-hydrated DNA fibers) = A-DNA
ray diffraction analysis of DNA fibers - not uniform, local deviations from average structure
and DNA crystals - rotation angles vary
- two bases are not perfectly coplanar, show propeller twist to enhance base stacking
Compare A, B, and Z type DNA Characteristic A B Z
Width Broadest Intermediate Narrowest
Rise/bp 2.3 Å 3.4 Å 3.8 Å
Helix diameter 25.5 Å 23.7 Å 18.4 Å
Screw sense Right Right Left
Glycosidic bond Anti Anti Alternating anti
and syn
Bp/turn of helix 11 10.4 12
Pitch/turn of helix 25.3 Å 25.4 Å 45.6 Å
Tilt of base pairs 19° 1° 9°
from normal to
helix axis
Major groove Narrow and very Wide and deep Flat
deep
Minor groove Broad and Narrow and deep Narrow and deep
shallow

Z-DNA’s helix is what handedness? Left-handed


Shape of path traced by phosphodiester Zigzags, form adopted by short oligonucleotides that have sequences of alternating
backbone pyrimidines and purines.
Linking Number Lk = number of times that a strand of DNA winds in right handed direction around
Twisting number the helix axis
Writhing number (+) = right handed winding, (-) left handed winding
DNA topoisomer? Tw = measure of helical winding of DNA strands around each other
(+) = winding, (-) = unwinding
Wr = measure of coiling of the axis of the double helix (supercoiling)
(+) = left handed supercoiling, (-) = right handed supercoiling

Topoisomer = DNA that differ only in linking number


How does supercoiling affect the Supercoiling makes DNA molecule more compact and it moves faster in
electrophoretic and centrifugal mobility centrifugation or electrophoresis.
of the molecule?
How can a negative super-helix density Negative supercoiling introduces a torsional stress that favors unwinding of the
facilitate the unwinding of the helix? right-handed B-DNA double helix.
How is free energy of negative Free energy is minimized when 70% of change in Lk is expressed in Wr (everything
supercoiling partitioned into Tw and is bp’d so more energetically favorable) and 30% is expressed as Tw. A lowering of
Wr? Lk causes both negative supercoiling of DNA axis and unwinding of the duplex
Topoisomerase Topoisomerase – introduce or eliminate supercoils
Type I Type I: relax supercoiled DNA, thermodynamically favored, tyrosine cleaves one
Type II strand of DNA, controlled rotation
Substrate, product, mechanisms Type II: utilize free energy from ATP hydrolysis to add negative supercoils to
DNA, cleave both strands of DNA, G “gate” ds segment binds enzyme, T
“transported” segment binds loosely, enzyme domain binds ATP, conformational
change cause domains to come together, cleave G segment and trap T segment,
strand joined by tyrosine-phosphodiester linkage , DNA cannot rotate, decreases
linking number by two.
Both use tyrosine to form covalent links to the polynucleotide backbone when it is
transiently broken
Linking number effect of Topoisomerase I unwinds 1 negative supercoil per cycle:
Topoisomerase I (↑ Lk +1, Wr +1, Tw = 0)
Topoisomerase II Topoisomerase II breaks both DNA strands,
(↓ Lk -2, Wr -2, Tw = 0)
How can you tell the difference ssDNA has a higher absorbance at UV λ 260nm
between ss and dsDNA?
How do you detect specific DNA/RNA Hybridization of labeled DNA to nucleic acid bound to membrane = southern
sequences in vitro? blotting (DNA), northern-blotting (RNA)
In vivo? - electrophoresis on agarose gel, 2. transfer of DNA by blotting onto
nitrocellulose sheet, 3. add labeled DNA probe, 4. autoradiography shows
DNA probe
PCR amplification of DNA
In vivo: fluorescent in situ hybridization (FISH)
Compare structure of DNA, RNA, and DNA RNA Protein
proteins Secondary Double helical, B- Diverse – stems, α-helices, β -
structure conformation loops, bulges sheets
Tertiary structure No, supercoil Considerable, H- H bond, ionic
bonding of bonds, van der
unpaired bases Waals
Structural Limited Considerable Enormous
plasticity deviations from B
Physical stability High Moderate Low
Chemical stability High (bases in Moderate Variable
center) (susceptible to alkaline
hydrolysis)
Chemical variety Low – 4 bases Low – 4 bases, High—20 aa
modified
Chemical Very low – bping, Moderate: RNAs High: catalyze lots
reactivity bases and backbone can catalyze limited of rxns, bind ions,
can be bound by rxns like metabolites, and
proteins transesterifications macromolecules
Key features and reactions catalyzed by DNA polymerase catalyzes the formation of polynucleotide chains, adds nt to 3’ end
DNA polymerases of polynucleotide chain, catalyzes the attack by 3’-OH to the α phosphoryl group of
the nucleoside triphosphate. Needs a primer to initiate
What are the common structures and Finger and thumb – wrap around DNA and hold it across enzyme’s active site
evolutionary relationships among DNA Palm: active site
polymerases? Two metal ions – catalyze the polymerase reaction
What are the two roles of Mg2+ in the 1. activate the 3’-OH of the primer, facilitates nucleophilic attack of α-phosphate
reaction catalyzed by DNA group of dNTP substrate. Two ions help stabilize the negative charge that
polymerases? accumulates on the pentacoordinate transition state.
2. stabilize the negative charge on the pyrophosphate product.
What orients the metal ions? Conserved Asp

How does a DNA polymerase select the Shape complementarily – nucleotide with similar shape, but different hydrogen
correct incoming dNTP substrate? bonds, can still be incorporated
1. DNA polymerase forms hydrogen-bonds with the minor groove side of the bp in
the active site to check for proper spacing
2. DNA polymerase close down around incoming dNTP, finger domain forms a tight
pocket that ensures proper shape of base pair.
All replicative DNA polymerases are 3’5’ exonuclease activity
associated with _____
Klenow Fragment Contains polymerase unit and 3’5’ exonuclease activity

Enzymes that form and remove RNA Primase forms RNA for initiation
primers from the genome. DNA pol I erases primer with 5’  3’ exonuclease and fills in gaps on lagging
strand with polymerase activities
Substrates and reaction mechanisms of Seals the nicks after replacement of RNA primers
DNA ligase Seals breaks in double-stranded DNA molecules, cannot link two molecules of
ssDNA or circularize ssDNA
Similarities and Differences between Similarity: origin of replication, synthesis the same
bidirectional replication of linear vs. Difference: nature of products, circular DNA becomes intertwined and must be
circular DNA? separated by topoisomerase II, end problem of linear DNA
Reactions and movements of DNA DNA pol III synthesizes the leading and lagging strand simultaneously at the
strands that occur during replication/ replication fork. Primase synthesizes RNA primer, duplex DNA ahead is unwound
define replication fork by DnaB (helicase), SSB bind unwound strand, Topoisomerase II introduces
negative supercoils to prevent topological crisis
How do helicases separate the strands A1 (P-loop NTPase fold) and B1 bind ssDNA, ATP binds and the cleft closes and
of duplex DNA? domain A1 slides along the DNA. On ATP hydrolysis, the cleft opens up, pulling
DNA from B1 toward domain A1.
Define Processivity Processivity – the ability of an enzyme to catalyze many consecutive reactions
Role of β2 subunit of DNA pol III without releasing its substrate
Role of γ β2 unit keeps the polymerase associated with the DNA double helix = sliding clamp
γ binds ATP, induces conformational change, binds and opens β ring
ATP hydrolyzed and β ring closes and clamp loading complex released
Distinctive features of DNA pol III Polymerase core: αεθ
holoenzyme Sliding clamp: β2
How does an asymmetric dimer of the clamp loader: γτ2δδ’
enzyme coordinate the synthesis of DnaA: binds oriC, forms hexamer
leading and lagging strands of the DnaB: helicase
daughter duplexes? DnaC: helicase loader protein
DnaG: primase
___ model Lagging strand looped out so it passes through polymerase site in the same direction
as the leading strand template. Pol I lets go of the lagging-strand template after
adding 1000 nts by releasing the sliding clamp. A new loop is formed, a sliding
clamp is added, and primase is synthesized in short stretches. Trombone model –
loop lengthens and shortens.
Role of DNA pol I and DNA ligase in Gap between lagging strands filled in by pol I. (has 5’  3’ exonuclease activity)
replication DNA ligase connects the fragments.
Function and features of the nucleotide Origin of replication has 5 copies of a sequence that are binding sites for DnaA +
sequence of oriC tandem 13-b AT sequence repeats.
Bidirectional replication initiation in E. To start replication:
coli? 1. DnaA binds to 5 sites in oriC, come together to form a cyclic hexamer
Proteins that interact with DNA in (dissociates at beginning of initiation to prevent additional rounds of replication
oriC, reactions catalyzed and functions from beginning), DNA wraps around
served 2. ssDNA are exposed in the pre-priming complex. DnaB (helicase) loaded by
DnaC, unwinds DNA, SSBP traps ssDNA. DnaG (primase) inserts RNA primer
3. Polymerase holoenzyme: DNA pol III holoenzyme assembles, initiated by
interactions between DnaB and sliding clamp subunit. ATP hydrolysis within
DNA subunits breaks up assembly, signal the initiation of DNA replication
Problem arising from DNA length and 4.6 million bp, humans have 6 billion bps. 1 vs 46 chromosomes, circular vs. linear
cell cycle in eukaryotes. Rate of chromosome, rate – 60-90 nt/sec. 30,000 replication origins (each origin is the
replication? Solutions? starting site for a replication unit, replicons)
Telomeres: prevent loss of information because polymerase can only act in 5’ 3’
direction
In Eukaryotes, DNA synthesis is ___ DNA synthesis is only takes place during S phase of cell cycle
Replication licensing ensures ____ Replication licensing ensures that the genome is replicated once, and only once, per
cycle
Replication Initiation in Eukaryotes 1. assemble ORC (6 proteins ~DnaA)
2. Cdc6 + cdt1 recruit Mcm2-7 (helicase) = Licensing factors + Replication protein
A (SSBPs)
3. polymerase switching: Pol I ~ α initiates replication, Pol III ~ δ = processive pol
4. after polymerase α adds 20 dNTPs to primer, replication factor C (RFC)
displaces pol α. RFC attracts sliding clamp (PCNA – proliferating cell nuclear
antigen ~ β2)
PCNA + DNA pol δ makes the enzyme highly processive, continue to replicate until
adjacent replicons fuse
Licensing factors accumulate only G1
during ___ After phosphorylation, Mcm – nuclear export
Ways to get rid of licensing factors? Phosphorylated cdc6 – degraded
Why important to eliminate factors? Cdt1 – sequestered by binding Genin (present from s M phases, then degraded)
To prevent setting up new origin of replication
Define Telomeres Telomeres: specialized structures at the ends of linear chromosomes.
How does telomerase make DNA of (Tandem repeats of six-nt sequence at 3’ end of DNA, the single-stranded segment
defined sequences in the absence of a of G-rich strand extends from the end of the telomere. Ss region invades the duplex
DNA template? and forms a large duplex loop to protect genomic ends.)
Telomerase contains an RNA molecule that serves as a template for elongation for
the G-rich strand, reverse transcriptase
Common types of mutations in DNA 1. formation of pyrimidine dimers induced by UV irradiation
2. depurination and deamination
3. spontaneous alterations
4. exposure to chemical mutagens – nonstandard base pairing
Base analog mutagens (5-BU, 2-AP incorporated into DNA, form transient
tautomers)
Nitrous acid deaminates bases
Involvement and limitations of error- DNA polymerases that can replicate DNA across many lesions, allow the
prone DNA polymerases. completion of a draft sequence of the genome that can be partially repaired by DNA-
repair processes, DNA recombination
Trinucleotide-repeat expansions An autosomal dominant neurological disorder, mutated gene creates glutamine
How do they relate to Huntington residues, increased number of trinucleotide sequence. Polyglutamine stretches
disease? aggregate
General strategies for repairing 1. Proofreading – growing polynucleotide chain leaves the polymerase site,
damaged DNA migrates to nuclease active site, remove incorrect bases
How are mismatches, pyrimidine 2. mismatch repair – 2 proteins (one detects mismatch, other recruits endonuclease
dimers, alkylated bases repaired? to cleave DNA) MutS recognizes mismatch, MutL calls MutH to cleave backbone.
(newly synthesized DNA is unmethylated)
Double strand breaks induced by Ionizing radiation, oxidizing agents, errors in replication
Why are they potentially dangerous? Potentially dangerous because there is no intact template strand to guide repair (as I
Repaired by NER & BER)
They are repaired by nonhomologous end joining, homologous end joining.
Why are low rates of mutation In germline to maintain thee species
essential? In somatic cells to prevent individual from developing cancer, harmful mutations
How are low mutation rates 1. accuracy during DNA synthesis
maintained? 2. post-replicative mechanisms that repair most damage
High fidelity in DNA replication is the 1. Double checking of base-pairing geometry before covalent addition of a
result of nucleotide (higher affinity for correct nt because of bping with template)
2. remove first new nts synthesized that are most likely to be mispaired
3. exonucleolytic proofreading – mispairing induces change from active site to
exonuclease site (kinetic proofreading – delay after mistake allows spontaneous
melting and released 3’ end to contact exonuclease site, excises mismatch)
Difference between base-excision Base excision repair – mediated by DNA glycosylases (enzyme flips out nucleotide
repair and nucleotide-excision repair to evaluate each bp, removal of sugar phosphate residues by cleaving glycosidic
bond) DNA backbone is intact, base is missing AP site (ex: AlkA enzyme)
Nucleotide-excision repair – excise pyrimidine dimer, entire nucleotide including
backbone. The distortion of the B-helix is recognized by the multienzyme repair
complex.
Chapter 29: RNA metabolism Holoenzyme: α2β β ’σ Core enzyme: α2β β ’
Subunit composition of RNA α – binds regulatory proteins and sequences
polymerase of E. coli for holoenzyme + β - binds NTPs and catalyzes bond formation
core enzyme β ’ – binds the DNA template
σ70 – recognizes the promoter (-35 and -10 region) by scanning sequence in 1
Describe function of each subunit direction and initiates synthesis, only binds DNA when associated with core
enzyme, otherwise has an autoinhibitory domain to block DNA-binding surfaces
Comparison of DNA and RNA Similarities:
synthesis 1. initiate at specific site in genome (origin – replication, promoter – transcription)
2. produce new nt chains by synthesis in the 53’ direction
3. driven by hydrolysis of pyrophosphate after incorporation of dNMP or NMP from
dNTP or NTP precursors
Differences:
1. only specific segments of genome are copied into RNA
2. only one of two strands serves as template
3. RNA polymerases initiate synthesis de novo
4. transcription has higher degree of regulation, genes only transcribed under
specific conditions
How do RNA polymerases catalyze Nucleophilic attack by terminal 3’ OH in nascent RNA chain on the α-phosphate
nucleophilic attack? group of the incoming NTP
Strong vs. Weak promoters strong promoter: have close match in -10 and 35 to consensus sequence, can be
transcribed every 2s (promoters encoding rRNAs)
weak promoters: differ at multiple sites within -10 and -35 sequences, or space
between them (17 bp is optimal)
Three stages of RNA synthesis and 1. promoter binding and initiation
functions of RNA polymerase in 2. elongation of nascent RNA transcript
processes 3. termination at end of gene
Eukaryotic differences in transcription - regulated by TF binding (control promoter activity)
- enhancer sequences imitate transcription
- modified transcripts: 5’ cap and 3’ poly A tail
How does σ factor enable RNA RNA polymerase and σ factor slides along DNA, specifically binds to promoter site
polymerase to recognize promoters? on template DNA. Released after RNA chain 9-10 nts long (acts catalytically)
Difference between σ70 and σ32? Σ70 recognizes consensus sequence
When temperatures are raised suddenly, E. coli synthesizes σ32, which recognizes
promoters of heat-shock genes (promoters exhibit -10 sequences that are different
from standard promoters, leads to synthesis of protective proteins). Σ plays a role in
determining where RNA polymerase initiates transcription (starved, heat, etc)
How was topoisomerase I used to Experiment: analyze the supercoiling of circular duplex DNA exposed to varying
determine the number of promoter amounts of RNA polymerase.
DNA base pairs that are unwound upon Topo I added to relax (Wr +1) part of circular DNA not in contact with polymerase
binding of RNA polymerase molecules. Remove bound protein and analyze DNA samples by gel
 how does negative supercoiling electrophoresis.
relate to promoter efficiency? ↑ # of RNA polymerase bound/template DNA led to ↑ in degree of negative
supercoiling.
∴ RNA polymerase unwinds DNA by (-Tw or –Wr: supercoil)
Each bound polymerase molecules unwinds a 17 bp segment of DNA, which
corresponds to 1.6 turns of B-DNA helix.
Two strands of DNA twist around the 10.4 bps
helica xis once very ___ bps
Closed promoter vs. Open promoter Closed – DNA is double helical
complexes Open – DNA segment is unwound
How does topoisomerase II (Aka: ___) Aka DNA gyrase facilitates transcription by introducing negative supercoils.
facilitate transcription? Negative supercoiling in circular DNA favors transcription because it favors
unwinding. (Exception: negative supercoiling of promoter site for topoII gives
negative feedback, decreases efficiency, prevent excessive supercoiling).
Detail the de novo initiation of chain Unwind the template to form an open promoter complex
growth by RNA polymerase, name Very tight association of enzyme with template
unusual initiating ribonucleosides First base: pppG or pppA
2 steps to transcriptional Elongation 1. promoter clearance : after 5-10 nt are added to nascent RNA, σ dissociates from
the complex (lose contact with promoter, affinity of the core enzyme for template
increases by a factor of 104 – nonspecific to sequence)
2. Processive elongation without accessory proteins. RNA polymerase acts a
molecular motor on the DNA – produces greater forces than myosin/kinesin
What is the transcription bubble? Region containing RNA polymerase, RNA-DNA hybrid
Number of base pairs in RNA-DNA 8 bp, 1 bp of DNA unwound in elongation phase (constant length of unwound region
hybrid. means DNA is rewound at the same rate as unwound)
Rate of RNA chain elongation (nt’s RNA synthesis: 50 nt/sec
added? Distance on the template Distance traveled: 170 Å/sec
traversed by RNA polymerase?) vs DNA synthesis: 800 nt/sec
DNA rate

Termination signals in RNA Transcribed regions of DNA templates contain stop signals (GC palindrome hairpin
loop followed by 4+ uracils. RNA polymerase pauses after synthesizing hairpin,
weakly bound nascent RNA dissociates from DNA template, DNA template
renatures to close transcription bubble.
ρ -dependent vs. ρ -independent ρ protein helps to terminate transcription of some genes (ex: 23S RNA  10S, 13S
transcription termination. Outline and 17S RNA)
mechanism of ρ protein and role of ρ hydrolyzes ATP n the presence of ssRNA, but not in the presence of DNA or
ATP hydrolysis in function duplex RNA. Hexameric ρ binds ssRNA (72 nt) so that RNA passes through center
of structure, ATPase activity enables protein to pull the nascent RNA while pursuing
RNA polymerase. When it catches up to RNA polymerase at transcription bubble,
ρ acts as a helicase and breaks RNA-DNA hybrid helix
ρ structure similar to ____ ATP synthase, hexameric
Compare specific subunits ssRNA plays role of γ subunit
Mechanisms of inhibition of Rifampicin is an antibiotic that interferes with the formation of the first few
transcription by rifampicin and phosphodiester bonds in the RNA chain by blocking the channel into which the
actinomycin D RNA-DNA hybrid generated by enzyme must pass. After initiation, RNA-DNA
hybrid in the enzyme prevents antibiotic blocking. Bacterial RNA polymerases
conserved, rifampicin useful against tuberculosis.
Actinomycin D inhibits transcription by binding tightly and specifically to double-
helical DNA, prevent effective template for RNA synthesis. Intercalcation –
actinomycin slips in between neighboring bps in DNA, inhibit rapid cancer growth
Processing and modification of the Prokaryotic mRNA undergoes little modification
precursors of mRNA, rRNA, and tRNA In E. Coli, three rRNAs and a tRNA are excised from one primary RNA transcript
with spacer regions.
Ribonuclease P (Rnase P) generates the correct 5’ terminus of all tRNA molecules.
Addition of CCA sequence at the 3’ end of all tRNAs
Rnase III excises 5S, 15S, 23S rRNA precursors by cleaving double-helical hairpin
regions at specific sites
Modification of bases and ribose units of rRNAs (methylation = greater structural
and functional versatility)
3 Regulatory differences? 3 characteristics of gene expression: nuclear membrane compartmentalization,
transcriptional regulation, RNA processing

Spatial/temporal differences in
transcription and translation between Prokaryotes Eukaryotes
prokaryotes and eukaryotes. Translation begins as transcription is Transcription and translation take place
till taking place in different cellular compartments,
Difference in mRNA processing in transcription takes place in nucleus,
prokaryotes and eukaryotes translation outside in cytoplasm
3 promoter elements: -10, -35, UP Elements that regulate transcription can
elements be bound at a variety of locations in
DNA up or down stream, long
distances
Little mRNA modification 5’ cap, 3’ poly(A), splicing
3 RNA polymerases of eukaryotes. Type Location Cellular Effects of α-
Location in cell. RNA they synthesize, Transcript amanitin
effect of α amanitin I Nucleolus 18S, 5.8S, and Insensitive
28S rRNA
II Nucleoplasm mRNA precursors Strongly inhibited
and snRNA
III Nucleoplasm tRNA and 5S Inhibited by high
rRNA concentrations

What unique feature does RNA Carboxyl-terminal domain (CTD) - contains YSPTSPS consensus sequence, activity
polymerase II contain? is regulated by phosphorylation on Serine residues of CTD
Why is α-amanitin toxic? Can does it Α-amanitin binds tightly to RNA pol II (small Kd = 10 nM), blocks the elongation
differentiate the three eukaryotic RNA phase of RNA synthesis.
polymerases?
Sequence elements of eukaryotic RNA pol I: promoter sequences are located in stretches of DNA separating the 3
promoters. Contrast nucleotide rRNA genes. Ribosomal initiator element – rInr (TATA-like sequences at start
sequences and locations of the TAT site), and upstream promoter element – UPE (150 to 200 bp from start site)
box of eukaryotes and the -10 sequence RNA pol II: enhancer elements that are more than 1 kb from start site
of prokaryotes RNA pol III: promoters are within the transcribed sequence, downstream of start
site
TATA-box binding protein role? - active in assembling TFII transcription complexes, between -30 and -100 position,
- Initiator Element (Inr): sequences found at transcriptional start site (-3 to 5)
- Downstream core promoter element (DPE): (+28 to +32)
Transcription factors, other DNA- TFIID (includes TBP that recognizes TATA box) begins initiation by binding to
binding regulatory proteins that direct TATA box -- binds with 105 higher affinity
RNA polymerase to specific genes TBP provides docking site for TFIIA, TFIIB, then TFIIF, RNA po II, TFIIE, and
TFIIH = basal transcription apparatus
TBP mechanism TATA box of DNA binds to concave surface of TBP (Saddle-shaped protein),
binding induces conformational changes in DNA, double helix unwound by
widening of minor groove, enables extensive contacts with antiparallel β strands on
the concave side of TBP 1. DNA is bent, 2. unwound
Nucleosome Repeating units of chromatin (the complex of DNA and protein that makes up
chromosomes.) – DNA + 4 pairs of histones
What marks transition from initiation to TFIIH phosphorylates CTD, stabilizes transcription elongation by RNA pol II and
elongation in RNA synthesis? recruits RNA-processing enzymes that act during elongation
Intragenic regulators of RNApII Sp1 (stimulatory protein 1) – sequence-specific transcriptional activator
HSF – activated in response to elevated temperature
Properties of enhancer sequences Enhancers – cis-acting sequences that stimulate transcription several thousand base
pairs away, have no promoter activity of their own, can be up, down, or in the
middle of a transcribed gene.
tRNA modifications in eukaryotes 5’ leader cleaved by Rnase P, 3’ trailer is removed, CCA is added, base and ribose
moieties modified, pre-tRNAs are spliced by an endonuclease and a ligase to remove
an introns
Structure of 5’ end of eukaryotic Immediate, a phosphoryl group is released by hydrolysis. The diphosphate 5’ end
mRNA, purpose? attacks the α-phosphorus atom of GTP to form a 5’-5’ triphosphate linkage “cap.”
Protect 5’ end from phosphatases and nucleases, enhance translation
How does mRNA produce poly(A) Endonuclease cleaves AAUAAA sequence in primary transcripts (with important
tail? context), poly (A) polymerase adds 250 adenylate residues
RNA editing Change in nucleotide sequence of RNA after transcription by processes other than
Examples of effects on gene expression RNA splicing. Two forms of Apo B (apo B-100, Apo B-48). Cytidine deaminated to
uridine, deaminase only in small intestine
Splicing of eukaryotic mRNA Excising of introns and linking of exons, two transesterification reactions
Consensus sequence Introns begins with GU and ends with AG
Splice site junctions 3’ end of introns: 10 pyrimidines, C, then AG
Other nt sequence elements in process Branch site – 20-50 nt upstream of 3’ splice site, contains nucleophilic 2’-OH
Chemistry of splicing process 1. cleavage of phosphodiester bond between the upstream exon and the 5’ end of the
introns. Attacking group = 2’0OH group of adenylate residue in branch site to form
2’-5’ phosphodiester bond between A residue and 5’ terminal of introns
(transesterification) lariat formed
2. 3’-OH terminus of exon 1 attacks phosphodiester bond between introns and exon
2. Exons join and introns is released as a lariat. (transesterification)
Compare number of phosphodiester Same before and after, no energy source needed
bonds broken and formed during Phosphodiester bond between exon 1 and 5’ introns broken
mRNA splicing Phosphodiester bond between 2’OH and 5’ introns formed
Phosphodiester bond between exon 2 and 3’ introns broken
Phosphodiester bond between exon 1 and exon 2 formed
Spliceosome, detail structure and Small nuclear RNAs (snRNAs) – have less than 300 nucleotides, located in nucleus
catalytic involvement of small nuclear Ex: U1, U2, U4, U5, U6 – essential for splicing mRNA precursors
ribonucleoprotein particles in mRNA Small nuclear ribonucleoproteins (snRNPs)
splicing Spliceosome = assemblies of snRNPs, splicing factors, and mRNA precursor
Role of snRNPs U1: binds the 5’ splice site and then the 3’ splice site
U2: binds the branch site and forms part of the catalytic center (binding requires
ATP hydrolysis)
U5: binds the 5’ splice site
U4: masks the catalytic activity of U6
U6: disengages from U4 and base pairs with U2, displaces U1 @ 5’ end of introns,
catalyzes splicing with U2 (catalytic center)

2nd transesterification: U5 helps 3’-OH grp of exon 1 positioned to nucleophilically


attack 3’ splice site. U2, U4, U5 bound to excised lariat introns released to complete
splicing reaction
Ways in which transcription and One of the serines must be phosphorylated in CTD, which is controlled by kinases
translation are coupled in eukaryotes, and phosphatases. Phosphorylation state helps CTD recruit:
describe role of carboxyl-terminal 1. capping enzyme, 2. components of the splicing machinery, 3. an endonuclease
domain (CTD) of RNA polymerase II that cleaves the transcript at the poly(A) addition site, creating a free 3’OH group for
3’ adenylation
Ribonuclease P Catalyzes the maturation of tRNA by endonucleolytic cleavage of nucleotides from
5’ end of precursor
Reactions during self-splicing 1. add guanosine nucleotide can serve as energy source or an attacking group that
conversion of rRNA precursor from attacks 5’ end of introns
Tetrahymena into mature rRNA 2. exon 1’s free 3’-OH attacks 3’ splice site = second transesterification to join the
two exons, release intron
Describe the role of proteins in the 1. Package pre-mRNA or snRNAs (hnRNP, snRNP proteins)
spliceosome 2. Facilitate snRNA-mRNA interactions
3. Catalyze RNA rearrangements (RNA helicases)
4. Regulate splicing
2 pieces of evidence that U6 snRNA 1. coordination of Mg2+ by U6 snRNA is essential for splicing
catalyzes splicing 2. U2/U6 snRNA complex synthesized in vitro can bind to a synthetic RNA with a
branch point sequence and catalyze attack of the 2’OH of the branch point A on a
specific phosphodiester bond of U6 snRNA
Chapter 30: Protein Synthesis Adapter molecules that bind to a specific codon, brings with it an aa for
Describe tRNAs incorporation into the polypeptide chain
Contains certain modified nucleosides, half nucleosides are base-paired helical
regions, contain fewer than 100 ribonucleosides
Ribosomes Complexes with three large RNA molecules and 50 proteins. Ribozyme = RNA
component plays the most fundamental role
Links AA to each tRNA Aminoacyl-tRNA synthase, driven by ATP cleavage
Driven by _____
Common features of all tRNAs 1. 73-93 ribonucleotides
2. 7-15 unusual bases (methylated/demethylated, methylation = hydrophobic
interactions with syntheses and ribosomal proteins)
3. ½ nts bp’d to form a double helices
5 groups not bp’d (help with unique distinguishing), 3’ CCA terminal region
(acceptor stem), TψC loop, DHU loop (dihydrouracil), anticodon loop
4. 5’ end of tRNA is phosphorylated, 4’ terminal = pG
5. activated aa attached to –OH group of adenosine residue, located at 3’-CCA
component of the acceptor stem
6. anticodon near center of sequences
Why tRNA molecules have common Common reactions: ribosome, Efs
structural features? Unique? Different: activating enzyme
Bonds formed by aminoacyl-tRNA - a mixed anhydride bond is formed to AMP
synthetase reaction - acyl ester link to 3’ or 2’ –OH of tRNA
Transpeptidation is ___, driven by __ Spontaneous, driven by formation of peptide bond that is stronger than the ester
bond

∆ G°‘ of reaction catalyzed by the ~0 kcal/mol because free energy of hydrolysis of ester bond of aminoacyl-tRNA =
aminoacyl-tRNA synthetases is free energy of hydrolysis of phosphate bond ATP  AMP + Ppi
tRNA structure (det by ___) Determined by x-ray crystallography
L-shaped
A form DNA (4 helical regions form 2 db helix)
Nonhelical region = H-bond interactions
CCA terminus has aa-attachment site, SS region: change conformations during aa
activation and protein synthesis
Two-step reaction sequence of 1. AA + ATP ↔ aminoacyl adenylate + Ppi (mixed anhydride, COOH group
aminoacyl-tRNA synthetases of aa linked to π group of AMP)
2. aminoacyl-AMP + tRNA ↔ aminoacyl-tRNA + AMP
Where are the high-energy phosphate  2 ATP consumed to synthesize aminoacyl0tRNA
bonds that are consumed in the overall
reaction?
Describe the mechanisms of amino acid 1. acylation site eliminates Aas that are too big (won’t fit in)
selection and proofreading that 2. hydrolytic site eliminates Aas that are too small (editing site removes AA by
contribute to overall accuracy of hydrolysis if AA fits)
peptide
Difference in class I and class II I: acylates the 2’-OH group of terminal adenosine of tRNA
aminoacyl-tRNA synthetases CCA arm = hairpin conformation, monomeric
II: acylates the 3’OH group of terminal adenosine of tRNA
CCA arm = helical conformation, dimeric
3D structure of a prokaryotic ribosome Ribonucleoprotein made of small (30S) and large (50S) subunits
RNA percentage, RNA and protein RNA = 2/3 mass of ribosome
subunits, binding sites, mRNA bound 3rRNAs 5S, 16S, 23S, folded into structure with short duplex regions
within ___ subunit Have 3 tRNA binding sites that bridge the 30S and 50S subunits
mRNA bound within 30S subunit
Shine-Dalgarno sequence Initiator region of purine-rich sequence with 10 nt on 5’ side of initiator codon in
mRNA of prokaryotes (bp’s with 3’ end of 16S rRNA of 30S subunit contains a
sequence that bp’s with purine rich initiator site)
2 interactions hat determine where 1. pairing of mRNA bases with 3’ end of 16S rRNA
protein synthesis starts 2. pairing of initiator codon on mRNA with anticodon of an initiator tRNA molecule
Protein synthesis in bacteria starts with N-formyl methionine

How does polypeptide chain escape Through tunnel in ribosome


ribosome?
Describe initiation of Protein synthesis 1. 30S subunit and mRNA bind through Shine0Dalgarno sequence
2. initiator tRNA formylmethionine binds initiator AUG
3. 50S subunit binds 30S subunit – complete 0S subunit
4. Initiator tRNA in P site on ribosome
5. charged tRNA with anticodon complementary to codon in A site binds
6. peptide bond formation: Metf transferred to Aaa site, thermodynamically
spontaneous reaction, catalyzed by site on 23S rRNA (peptidyl transferase center),
amino group of aminoacyl-tRNA in A site attacks ester linkage between initiator
tRNA and Metf
7. P site tRNA = uncharged, protein enzyme EF-G translocates mRNA so next
codon shifted to A site, hydrolyze GTP
How does peptidyl transferase center 1. forming a nucleophilic NH2 group on A site aminoacyl-tRNA
help formation of peptide? 2. Stabilize Th intermediate

Chrs of peptidyl transferase center Only rRNA groups in the peptidyl transferase center: nearest protein group is 18Å distant
Initiation factors IF1: complex with 30S ribosome to prevent 50S from joining without mRNA,
IF3: complex with 30S ribosome, recognizes fmet-tRNAf bt not other tRNAs
IF2: G protein family binds GTP, conformation change associates with met-tRNAf
binds mRNA, initiator tRNA, stimulates association with 50S subunit = 70S
initiation complex
Name and describe three Efs EF-G: enzyme that translocates Aas (mRNA moves through ribosome by 1 codon
distance) and hydrolyzes GTP
GTP hydrolysis induces conformational in EF-G, forcing tail deeper into A site, and
hence complete shift of peptidyl-tRNA into P site and of free tRNA into E site for
release
EF-Tu: (G protein) uses GTP to bind aminoacyl tRNA and bind ribosome, two
functions:
1. protect ester linkage in aminoacyl-tRNA from hydrolysis
2. GTP  GDP hydrolysis when appropriate complex between EF-TU-
aminoacyl-tRNA complex and ribosome formed (no codon-anticodon = no
hydrolysis, no tRNA transfer), ∆ G of GTP hydrolysis contributes to
fidelity of protein synthesis, release EF-Tu from ribosome
*doesn’t recognize fmet-tRNAf, recognizes Met-tRNAm and other tRNAs (opposite
of IF2)
EF-Ts: induces dissociation of GDP from EF-Tu (GTP binds Ef-Tu and EF-Ts is
released)
Release Factors RF3: mediates reaction between RF ½ and ribosome (similar to G protein Ef-Tu)
RF 1 UAA, UAG, RF 2: UAA, UGA: recognize codons and promote release of
completed protein from the last tRNA
EF-G and RRF dissociates complex
Differences between Eukaryotic and Chr Euk Pro
prokaryotic translation initiation Ribosomes 60S + 40S = 80S 50S + 30S = 70S
initiator tRNA normal Met-tRNA formylMet-tRNAf
Initiation 40S ribosome w/ bound Shine Dalgarno
Met-tRNA attaches cap sequence, purine rich,
of 5’ end of euk. mRNA, attaches to 16S RNA
scans to 1st AUG,
helicase
# Start sites 1 start site Many start sites
5’ end Capped: mRNA circular: eIF Accessible to proteins
-4E binds cap, eIF-4G binds 2
polyA binding proteins
(PABPI)
GTP form delivers EF1 α Ef-tu
aminoacyl tRNA to A site
Catalyzes exchange of EF1 β γ EF-Ts
GTP for bound GDP
GTP driven translocation EF2 EF-G
Release factor(s) eRF1 RF 1, 2, 3
Prevents reassociation on eIF-3 IF3
ribosome subunits
without initiator complex
What kind of amino acids can be used L-Amino acids
for peptide bond formation
Ionisine and purpose Deaminated adenosine, maximizes number of codons that can be read by a tRNA
molecule, can base pair with U, C, or A
Explain mechanism behind wobble First two bases checked more heavily: 16S RNA in 30S subunit forms H bonds on
amino-groove with correctly formed bps of codon-anticodon duplex interactions that
check first two positions
3 major proteins cleaved 1. secretory proteins – exported, lysosomal protein, proteins spanning plasma
membrane
Describe process by which proteins Start translation on free ribosome, halts, directed to cytoplasm side of ER, ribosome
leave cell docks on membrane, protein synthesis begins again, during translation, polypeptides
goes through membrane  lumen of ER
4 components of ribosome 1. signal sequence: 9-12 hydrophobic aa, near amino terminus
translocation to ER 2. signal recognition particle (SRP): stop EF-binding site, halt protein synthesis
3. SRP Receptor (SR) – SRP-ribosome diffuse to ER, binds SR
4. Translocon – translocation machinery, integral and peripheral membrane
proteins, protein conducting channel (opens when ribosome binds, resumes protein
synthesis), polypeptide goes to lumen of ER
Mechanisms of ribosome catalysis Induced fit = conformational change upon binding correct A site substrate: important
to exclude H2O
Position effects = Entropy trap
Transition state stabilization
Maximal rate of each ribosome ~15 codons/sec
Error rate ~ 10-4 per residue
Binding energy of codon: anticodon Binding energy of codon: anticodon interaction is ~12 kJ/mol, corresponding to an
interaction affinity constant of about 100. Therefore, codon: anti-codon interactions should have
an error rate on the order of 10-2 per residue.
A kinetic Proofreading mechanism for - rate of productive transpeptidation, initiated by dissociation of Ef-Tu-GDP is
selecting a correct codon-anticodon independent of which tRNA is bound
interaction - rate of amino-acyl tRNA release, while retaining EF-Tu-GDP is larger when a non-
cognate compared to the cognate tRNA is bound.
∴ Incorrect amino-acyl tRNA is more likely to be released before transpeptidation,
proofreading depends on differences in the rate of a reaction when the correct vs.
incorrect substrate is bound to the ribosome.
Cross-pathway Comparisons FA activation: acceptor of acyl group = CoA
Describe acyl adenylate intermediates AA activation: acceptor of acyl group = tRNA
in two different pathways Both: irreversible by hydrolysis of pyrophosphate
Flexible arms Protein synthesis: flexible short stretch of polynucleotides in tRNA synthases edit
AA linkage without dissociation
Carboxylases: pyruvate carboxylase has a long flexible arm that allows prosthetic
group to rotate from one active site of enzyme to other (ATP-bicarbonate site to
pyruvate site -- gluconeogenesis), Propionyl carboxylase (degradation of odd chain
FAs converted to succinyl CoA, enters CAC), acetyl CoA carboxylase (FA
synthesis),
Pyruvate  Acetyl CoA: E2 transacetylase has flexible lipoamide cofactor attached
to lysine residue, carries substrate from active site to active side
Carboxylases An enzyme that catalyzes a carboxylation or decarboxylation reaction.
Acetyl CoA  malonyl CoA (acetyl CoA carboxylase): FA synthesis
Propionyl CoA  Succinyl CoA (Propionyl CoA carboxylase): after odd chain FA
degradation, conversion to succinyl CoA before entering CAC
Pyruvate  OAA (pyruvate carboxylase) gluconeogenesis

Biotin activates CO2 group, covalently attached to ε amino of Lys


Carboxy-biotin intermediate (CO2-biotin-enzyme) formed by hydrolyzing ATP
Activated CO2 transferred to acetyl CoA, pyruvate
G proteins G proteins bind guanyl nucleotides, stimulates adenylate cyclase activity, increases
concentration of cAMP
EF-Ts catalyzes exchange for EF-Tu activated receptor (GDP  GTP)
Heterotrimeric G protein in signaling pathways
Error Frequencies: DNA, RNA, protein DNA: 10-10: 3’5’ exonuclease
RNA: 10-5: backtracking stimulated by accessory protein allows mistakes to be
Repair mechanism corrected
Protein: 10-4: editing site from aminoacyl synthase, EF-Tu specific transport of
amino acyls
Activated complexes: E1 enzyme activates ubiquitin by adenylation (hydrolyzes ATP  Ppi)
Activation part leaves FA activation, AA degradation (activation by adenylation) – pg. 622 + 862
Maintains whole Activating group is released from growing chain: fatty acid synthesis, cholesterol
synthesis, protein synthesis
Activating group is released from incoming unit as it is added to growing chain:
Glycogen synthesis (UDP-glucose), DNA synthesis (dNTP), RNA synthesis
Ub activated by E1 by adenylation before transfer to protein marked for destruction
UDP-glucose – activated glucose
ATP – activated form of orthophosphate
Acetyl CoA – activated acetate
Water excluded from active site of Ribosome
Glycogen phosphorylase (cleave phosphorylytically, not hydrolytically to keep G1P
in cell)
Hexokinase on binding glucose, water would prevent phosphorylation
Pyrophosphate hydrolysis Drives: RNA synthesis, DNA synthesis, activates amino-acyl to charge tRNA,
citrulline  arginosuccinate (urea cycle), formation of UDP-glucose in glycogen
synthesis, drives activation of FA to acyl CoA before degradation
Urea cycle: Citrulline + asparatate + ATP  AMP + Ppi + argininosuccinate
(argininosuccinate synthetase)
Uses ATP hydrolysis ρ protein
Topoisomerase II
Helicase
Clamp loader
Ubiquitination
P loop NTPase DnaB – Helicase A2 (pg. 797)
DnaA – binds OriC during replication
19S component of proteasome: isopeptidase cleaves off Ub
α subunit of heterotrimeric G protein (P loop participates in nucleotide binding)
small G protein (Ras) – monomeric, cycle between GTP and GDP-bound form. – in
EGF pathway
α3β 3 subunits in hexameric ring of ATP synthase
ENZYME MECHANICS: Conformational change in an enzyme to form the substrate binding site induced by
Induced fit binding of the substrate
adenylate kinase, Hexokinase, Phosphoglycerate kinase, Pyruvate kinase
Citrate Synthase: binds OAA, then acetyl CoA and only hydrolyzes citryl CoA
Approximation The binding of two substrates to a single surface on the enzyme to increase the rate
of their reaction (correct orientation of binding increases reaction)
NMP kinases – pyrimidine metabolism 713
Carbonic anhydrase CO2  bicarbonate
Stabilization of Th intermediate The interaction among enzyme groups and the transition state of a reaction that
lower the activation energy of the reaction and hence increase its rate
Serine proteases: chymotrypsin, subtilisin (Serine’s –OH acts as a nucleophile
Peptidyl transferase center of ribosome (on 23S rRNA)
Rnase P (remove nts from precursor tRNA, and produces correct 5’ termini)
Multi-subunit enzymes α-1,6-glucosidase and transferase on 1 polypeptide chain. (glycogen degradation)
FA synthase: has acetyl Transacylase, malonyl transferase, acyl-malonyl ACP
condensing enzyme (domain 1), (domain 2) 3-hydroxybutyrl ACP dehydratase,
enoyl ACP reductase, β -ketoacyl ACP reductase, ACP, (Domain 3): thioesterase
Bifunctional PFK2 enzyme (controls glycolysis/gluconeogenesis): FBPase2/PFK2
Processivity In DNA synthesis (DNA pol III + β 2 sliding clamp)
RNA synthesis (RNA pol II)
glycogen phosphorylase activity (30Å from catalytic to glycogen binding site)
Primer needed Glycogenin – glycogen synthesis
DNA synthesis – RNA primer
2nd messengers cAMP, calcium, IP3, lipid DAG

Which dehydrogenase can utilize either Glutamate dehydrogenase (amino acid degradation)
NAD+ or NADH+?
Enzymes that move around phosphate Phosphoglycerate mutase (3PG  2,3 BPG  2 PG) – glycolysis
groups Phosphoglucomutase (G1P  glucose 1,6 BP  G6P) – glycogen breakdown
Use PLP Aldehyde group of coenzyme forms a Schiff base with a specific lysine side chain of
the enzyme.
Transaminase uses PLP during amino acid degradation (forms Schiff base)
Glycogen phosphorylase uses PLP to cleave glycogen phosphorylytically
Where is FADH2 made? Succinate dehydrogenase – CAC
Glycerol phosphate dehydrogenase – oxidize glycerol
Fatty acyl CoA dehydrogenase – oxidize fats
Transfer electrons from FADH2 to Q to form ubiquinol (QH2)
Effect of phosphorylation Pyruvate dehydrogenase: inactivated
Glycogen phosphorylase: activated
Glycogen synthase: inactivated
PK: inactivated
HMG-CoA
PFK2/FBase2: turns on PFK2
Use Mg2+ DNA polymerase
Hexokinase, Adenylate kinase, other kinases
Dehydrogenases GAP, alcohol, lactate, isocitrate, α-ketoglutarate, succinate, malate DH, an enzyme
that oxidizes a substrate by transferring one or more protons and a pair of electrons
to an acceptor, usually NAD/NADP

Anda mungkin juga menyukai