• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/323

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

323 Cards in this Set

  • Front
  • Back
In serpentine receptors, the carboxyl terminmal undergoes what?
Reverisible Ser/Thr phosphorylation
Ligand binding causes a what?
A confomational change which permits G-protein binding
Activated G-proteins do what?
• Activate or inhibit effector enzymes
What is an example of a serpentine receptor? And what is it mediated by?
β-Adrenergic Receptor Mechanism
• Mediated by epinephrine
What is an agonist and antagonist?
• Agonist-structural analogs that bind to a receptor and mimic the normal ligand (activators)
• Antagonist-analogs that bind to a receptor and block its activity (inhibitors)
What are the 4 heterotrimeric G-proteins?
Gs (stimulatory), Gi (inhibitory), Gr, Gq
What is adenylate cyclase?
An integral protein (not a signaling molecule)
What are the steps to the β-Adrenergic Receptor Mechanism?
1. Epinephrine binds to receptor causing a conformational change
2. The conformational change promotes binding of Gs to receptor
3. Gs-α GTP activates adenylate cyclase
4. Adenylate cyclase converts ATP to camp (second messenger)
5. G-proteins recycle, re-associate
What happens during desensitization of the β-Adrenergic Receptor Mechanism
1. Receptor/ligand complex binds β-ARK
2. β-ARK phosphorylates a Ser on receptor
3. phosphorylation induces binding of β-Arrestin
4. receptor-arrestin complex enters the cell by endocytosis
What does arrestin do?
• Prevents receptor interaction GTP-protein
• Recycles receptors
• Signals for endocytosis
Describe the downstream effect.
Conversion of glycogen to glucose through cAMP
• cAMP activates cAMP-dependent protein kinase (PKA)
• PKA molecules activate other enzymes
• Amplification
What is PKA and what does it do?
• An Allosteric enzyme
• Regulates a number of enzymes via Ser/Thr phosphorylation
What are PKA and PKC?
Antagonists
What does phospholipase C do?
Converts PIP2 to diacylglycerol and IP3 (2 important 2nd messengers)
What happens in plasma hormone receptors with to intrinsic enzyme activity?
1. hormone binds to receptor
2. GDP is exchanged for GTP on Gq
3. Activated Gq activates membrane phospolipase C
4. IP3 diffuses into endoplasmic reticulum and causes the release of intracellular Ca2+
5. Ca2+ binds to PKA
6. PKA becomes insoluble and associates with membranes
7. Diacylglycerol binds to PKC as a cofactor-PKC is activated
8. PKC phosphorylates a number of substrates on Ser/Thr
What are some responses to increased intracellular calcium?
• Exocytosis
• Muscle contraction
• Cytoskeletal rearrangements
• PKA and phosphatase activity increases
• Activation of calcium channels
• Calcium binding by intracellular proteins
What is Calmodulin?
Ca2+ binding protein
• 4 binding sites
• Ca2+ binding is cooperative (hill plot, similar to Hb)
• A subunit of phosphorylase b and of Calmodulin-dependent Kinase
Effect or responses caused by extracellular signals depends on what?
• The tissue or cells to which ligand binds
• The specific GTP-binding protein activated
• The 2nd messenger species generated
• The protein kinase activated
What is phosphorylation?
Phosphoryl group transfer
What is the feature of virtually all signal transduction mechanisms?
Protein phosphorylation
What are characteristics and tendencies of protein kinases?
• Tend to remain in the cell location containing their activators and their substrates
• Tend to be cell and tissue specific
• May be expressed as several isozymes (different form of same enzyme)
What important cell process is controlled by phosphorylation?
Cell division
What are modules?
Binding site groups (often multivalent) and domains on signaling molecules the contribute to propagating signal and regulating them.
What kind of binding domain does IRS-1 have?
PhosphoTyrosine Binding Domain (PTB)
When does signaling happen?
When new binding sites are produced from groups of proteins
IRS-1 can only bind to _______ after it is phosphorylated.
Grb-2
What are rafts?
What do they include?
Regions of membranes “enriched” in certain proteins and lipis
• Some sphingolipids and sterols
• Some GPI-linked proteins, Tyr-kinases
Rafts contain what?
And this does what?
Contain tight concentrations of signaling molecules.
Makes responses more effective
What is the refractory period?
In neuronal signaling, the time period during which receptors do not respond to ligand
Why do steroid hormones need to be bound to a serum binding protein?
Because they are hydrophobic
How do steroid hormones pass throught the lipid membrane? What are they held together by?
Carrier molecules
Held together by weak interactions
What do steroid hormones do at their target tissues?
1. Pass through membrane by simple diffusion
2. Bind to specific receptor proteins in the nucleus with good affinity
Why does hormone binding trigger a conformational change?
So they become capable of interacting with specific regulatory sequences in DNA called Hormone Response Elements (HRE).
What effects do steroid hormones have on gene transcription?
They enhance or inhibit it.
How long does it take for a full response to occur?
Hours to days
What is Tamoxifen and what does it do?
It is a breast cancer treatment.
It competes with estrogen binding (but does not enhance gene expression).
What important cell process is controlled via phosphorylation?
Cell division
What is Cyclin-dependent Kinase?
How many different CDKs do animals have?
A heterodimer of Cylcin + Catalytic subunit
8
What is cyclin and where is it synthesized?
A regulatory subunit associated with the dimer (not required).
Synthesized in cells
How is cyclin an example of amplification and specificity?
• Amplification- increases catalytic activity
• Specificity- animal cells have 10 different cyclins
What does CDK do?
It phosphorylates specific proteins at timed intervals to orchestrate cell division.
CDK activity is stimulated by what?
CDK activities occur with what?
• Stimulated by cyclin
• Occur with regular oscillations
What is a destruction box and what does it do?
It is a motif
Causes cyclin to bind several molecules of Ubiquitin
Ubiquitin-tagged proteins are degraded by proteosomes.
What causes cyclin degradation?
CDK phosphorylating DBRP (destruction box recognition protein)
How is most energy in a system lost?
As heat
Biological energy flow requires what?
• Conservation of energy
• Efficient utilization of free energy
• A constant source of energy
Define bioenergetics
• The quanititative study of biological energy transduction (movement of energy from one molecule to another one)
• The study of chemical processes that yield or use energy
Define: Gibb’s Free Energy, Enthalpy, Entropy
• Gibb’s Free Energy (ΔG)- the amount of energy capable of doing work during a reaction.
• Enthalpy (ΔH)- how many bonds and what kind are made and broken during a reaction.
o Exothermic=release of heat
• Entropy (ΔS)- Quantity of randomness or order; increases with disorder
Relate free energy, enthalpy, and entropy in an equation.
ΔG=ΔH-TΔS
What are the symbols for standard transformed conditions?
ΔG’° and K’eq
How are ΔG’° and K’eq related?
ΔG’° = -RTln K’eq
What are ΔG’° and ΔG?
The maximal free energies available under ideal conditions
What lowers the activation energy?
Catalysis
If there is a common intermediate, what can you do with ΔG and Keq?
ΔG-add
Keq-multiply
Reactions in which the terminal phosphoryl group of ATP is transferred to an acceptor molecule to produce a covalent bond is an example of what?
Group tranfer (NOT simple ATP hydrolysis)
What are the theoretical and actual energy values obtained from hydrolysis of ATP? Why are they Different?
Theoretical: -30.5 kj/mol
Actual: -51.5 kj/mol
ATP and ADP form a complex with MG2+ and free energy change is depended on that complex.
What is Phosphorylation Potential?
Actual energy available for biochemical reactions.
What class of reactions does ATP use to provide energy for biological processes?
Group Transfer
In hydrolysis reactions, why are the products more stable than the reactants?
1. Bond strain due to electrostatic repulsion is relieved by charge separation
2. Products are stabilized by:
a. Ionization
b. Isomerization
c. Resonance
A large and negative free energy (ΔG’°) results from coupling what?
1. Hydrolysis of the P-O bond
2. Formation of a reactive intermediate
3. Summation of all free energies
What are the 3 different groups ATP can transfer to other molecules in reactions?
1. ADP (phosphoryl) α
2. AMP (pyrophosphoryl) β
3. PPi (adenyl) γ
What is adenylylation and what is an example?
When ATP transfers adenylate as an adenylyl group to the nucleofile
Ex) fatty acid activation before oxidation
What is a transphosphorylation?
Transfer of a phosphoryl group
What are phosphoryl group donors?
What is the primary one?
All nucleoside triphosphates (NTPs) and deoxynucleoside triphosphates (dNTPs)
ATP
What are examples of enzymes that carry phosphoryl groups from ATP to the other nucleotides?
• Nucleoside disphosphate kinase
• Adenylate kinase
• Creatine kinase
What happens to the transfer of phosphoryl group with nucleoside diphosphate kinse?
1. Phosphoryl group from ATP transfers to an active site on His
2. Phosphoryl group transferred from enzyme His to NDP or dNDP
What happens in transphosphorylation with adenylate kinase?
• ADP accumulates under periods of high metabolic activity (interferes with muscle contraction)
• Adenylate kinase removes the ADP
What is oxidation and reduction?
• Oxidation-electron loss
• Reduction-electron gain
The flow of electrons in an oxidation/reduction reaction is dependent upon what?
The differences in affinity for electrons between 2 molecules
What is the force generated as a result of differences in electron affinity is defined by what? Measured in what?
Defined by Electromotive Force (EMF)
Measured in Volts
What is a dehydrogenation reaction and what enzyme catalyzes?
Conversion of an alkane to an alkene
Catalyzed by dehydrogenases
What is a reducing and oxidizing agent?
• Reducing- electron donor (reductant)
• Oxidizing- electron acceptor (oxidizer)
What is defined as a reducing equivalent in biological systems?
One electron moving by any of the 4 mechanisms of electron transfer
How many reducing agents do most biochemical enzymatic dehydrogenations transfer?
2
What are the 4 mechanisms of electron transfer?
1. Direct transfer of the electron
2. Transfer of a hydrogen atom (acid base)
3. Hydride ion transfers
4. Direct combination with oxygen (hydrocarbon is the e- donor and oxygen is the e- acceptor)
What is the standard reduction potential?
E°, Measure of the affinity of the electron acceptor for a pair of electrons
What is the Nernst Equation?
E=E° + [(RT)/(nF)]ln(e- acceptor)/(e- donor)
What equation relates standard reduction potential to free energy?
ΔG=-nFΔE
What are water-soluble electron carriers that move between several enzymes?
NAD+ and NADP+
What are water-soluble electron carriers that do NOT move between several enzymes, but serve as prosthetic groups?
What do they bind to?
FMN and FAD (flavonucleotides)
Bind to flavoproteins
NAD+ and NADP+ (names)
Nicotinadmide Adenine Dinucleotide
Nicotinadmide Adenine Dinucleotide Phospohate
What are NADH and NADPH derived from?
Niacin
What does difficiency of NADH and NADPH cause?
Pellagra, dermatitis, diarrhea, dementia, death
When a substrate oxidizes, what happens to the 2 H+ it gives up?
1. NAD+ or NADP+ accept a hydride ion
a. Both become reduced
b. Both forms absorb light at 340 nm
2. The other proton is released into the environment and buffered.
What functions to NADH and NADPH have?
• NAD+>>NADH, favors flow of electrons to carrier
o Catabolic oxidations
• NADPH>>NADP+, favors transfer of electrons to substrate
o Anabolic reductions
What are flavin nucleotides (FMN and FAD) derived from?
Riboflavin
FMN and FAD (names)
Flavin Mononucleotide
Flavin Adenine Dinucleotide
Why do FMN and FAD participate in a wider range of reactions than NAD and NADP?
They accept 1 or 2 electrons
What bio molecules are membranes composed of?
• Proteins
• Polar Lipids
• Carbohydrates
Constituents of membrane lipids vary with what?
Not with what?
• Vary with climate
o Nature of cell membrane is temp dependent
• NOT with diet
o Determines what is available, not how it is used
The fact that each membrane source has characteristic lipids implies that there are regulatory controls on what?
The fact that each membrane source has characteristic lipids implies that there are regulatory controls on what?
Which membrane lipids are found on the outer monolayer of the plasma membrane?
• Phosphatidylcholine
• Sphingomyelin
Which membrane lipids are found on the innermonolayer of the plasma membranes?
• Phosphatidylethanolamine
• Phosphatidylserine
• Phosphatidylinositol
• Phosphatidylinolsitol 4-phosphate
• Phosphatidylinositol 4,5-bisphosphate
Prosthetic group of Lipoproteins and example
• Lipids
• Ex) B-lipoprotein of blood
Prosthetic group of Glycoproteins and example
• Carbohydrates
• Ex) Immunoglobulin G
Prosthetic group of Phosphoproteins and example
• Phosphate groups
• Ex) Casein of milk
Prosthetic group of Hemoproteins and example
• Heme (iron pophyrin)
• Ex) Hemoglobin
Prosthetic group of Flavoproteins and example
• Flavin nucleotides
• Ex) Succinate dehydogenase
Prosthetic group of Metalloproteins and examples
• Iron- Ferritin
• Zinc- Alcohole dehydrogenase
• Calcium- Calmodulin
• Molybdenum- Dinitrogenase
• Copper- Plastocyanin
What is meant by the term “functional asymmetry” in membrane proteins?
Protein domains exposed on one side of the bilayer are different from those exposed on the other side.
What does “semi-permeable” mean in reference to the lipid bilayer?
It is permeable to nonpolar compounds.
Which way do nonpolar and polar (head groups) face on the lipid bilayer?
• Nonpolar-internally
• Polar-outward
Why is the term “fluid mosaic” used to describe the lipid bilayer?
It is constantly moving due to weak (noncovalent) interactions
What does the term “sidedness” mean in reference to the lipid bilayer?
The way the proteins are distributed causes the membrane to be asymmetric.
• Carbohydrate is always on outside, it is normally attached to a protein/glycoprotein.
Proteins interspersed through the lipid bilayer are stabilized by what?
Hydrophobic interactions
What are the 3 classes of membrane lipids that biological membranes are composed of?
1. Glycerophospholipids
2. Sphingolipids
3. Sterols
What are the 3 structures that membrane lipids form and which is the most stable?
1. Micelle
2. Bilayer
3. Liposome
Liposome is most stable
What are the individual subunits of micelles and bilayers?
What are the individual subunits of micelles and bilayers?
Micelles form easily from what?
1. Free fatty acids
2. Lysophospholipids
3. Detergents
What 2 factors affect the degree of mobility in a membrane?
1. Temperature
2. Lipid composition
What state do lower/higher temperatures favor and what state is preferred by cells?
• Low-paracrystal
• High-fluid state (liquid disordered)
• Gel is preferred by cells (liquid ordered)
What is phase change defined as?
Transition Temperature
What does adding more unsaturated fatty acids do to a membrane lipid?
It lowers the transition temperature
What do sterols do to the transition temperature of a membrane lipid and why?
Increase transition temperature
• Prevent ordering of acyl chains
• Favors liquid-ordered state
What are the 3 levels of membrane lipid mobility? (example of the rare one)
1. Lipid acyl chains move freely, but whole lipid does not move
2. Lipid acyl chains move freely and rapidly through the plane of the membrane
3. Mobility from one side of the bilayer to another (rare, requires enzyme: flipase)
Ex) phospolipid biosynthesis in bacteria
What feature about the primary structure of a protein is considered evidence of a trans-membrane domain?
The presence of unbroken sequences of more than 20 hydrophobic residues in a membrane
How can the “sidedness” of a protein be checked?
Hydropathy Plot- Used to predict trans-membrane domains of proteins (not absolute). Amino sequence often indicates secondary structure (α-helixes, β-sheets).
When secondary structure of proteins is known, hydrophobicity is revealed.
Hydropathy Index
A scale that expresses the relative hydrophobic and hydrophilic tendency of a chemical group
What are the 4 functions of metabolic pathways?
1. Obtain chemical energy from capturing solar energy or from degradation of energy rich nutrients.
2. Conversion of nutrient molecules into cellular precursors.
3. Polymeration of monomeric precursors into macromolecules.
4. Synthesis and degradation of specialized cellular biomolecules.
What is catabolism?
the degradative phase of metabolism where organic nutrient molecules (carbs, fats, proteins) are converted into smaller, simpler end products (lactic acid, CO2, NH3)
What happens in a catabolic pathway?
It releases energy, some is conserved in the form of ATP and NADH, NADPH, FADH2, the rest is lost as heat
What is anabolism?
small, simple precursers are built into larger and more complex molecules (lipids, polysaccharides, proteins, nucleic acids)
What does anabolism require?
an input of energy, usually in the form of phosphoryl group transfer of ATP and reducing power of NADH, NADPH, FADH2.
How were the metabolic pathways elucidated?
1. Metabolic inhibitors, growth studies, and biochemical genetics
2. Isotopes-atoms with same number of protons but different number of neutrons-specific labeling wihtout changing the chemical properties of the metablites
What Precurser of Heme was figured out by an isotope?
Glycerine
What can you use to detect isotopes?
NMR
What does the P-NMR do?
monitors levels of ATP, ADP, and Pi; it is useful to study energy metabolism in muscle.
What are the 5 general principles of Metabolic Pathways?
1. Metabolic Pathways are irreversible
2. Catabolic and Anabolic Process must differ
3. Every metabolic pathway has a committed step (usually the rate determining step)
4. All metabolic pathways are heavily regulated
5. Metabolic pathways in eukaryotic cells occur in specific compartments
What are metabolic pathways irreversible?
It is a highly exergonic reaction-it goes to completion
If a highly exergonic reaction is part of a multistep pathway the entire pathway is irreversible
Why does every metabolic pathway have a committed step?
early in each pathway, there is an irreversible reaction which "commits" the intermediate that is produces to continue down the pathways.
What are metabolic pathways regulated by?
What is usually regulated?
Regulated by the law of supply and demand.
The rate limiting step (usually the committed step) is usually regulated
Why do metabolic pathways in eukarytic cells occur in specific compartments?
Example?
different metabolites can operate in different locations and in different pathways.
Example: ATP is synthesized in mitochondria but used in cytosol
Acetyl-CoA is produced in mitochondrion but utilized in cytosol
What metabolic pathways occur in the mitochondria?
*citric acid cycle
*electron transport
*oxidative phosphorylatioins
*fatty acid oxidation
*amino acid breakdown
What is caveolin and what does it do?
It is an integral membrane proteins with 2 globular domains connected by a hairpin-shaped hydrophobic domain.
Where does caveolin bind?
What does it bind?
To the cytoplasmic leaflet of the plasma membrane.
It binds cholesterol in the membrane.
Caveolae involve what?
Both leaflets of the bilayer
Cytoplasmic-where the caveolin globular domains project
Endoplasmic-typical sphingolipid/cholestrol raft with associated GPI-linked proteins
What cellular functions is caveolae implicated in?
What cellular functions is caveolae implicated in?
Describe the movement of Glucose across the intestinal epithelial cells
1. glucose enters cells on the apical (luminal side)
Na+ gets in at the same time (Na+/glucose symporter)
2. glucose moves through the cell and passes into the blood via GLUT2
3. Na+/K+ transporter continues to pump Na+ outward to maintain the Na+ gradient that drives glucose uptake
What are examples of a Gated Ion Channel?
Na+/K+ ATPase
Nicotinic Acetylcholine Receptor
Neuronal Signaling
What are examples of Receptor enzymes?
Insulin Receptor
Guanylyl Cyclase
What are examples of Steroid Hormones?
Regulation of Transcription
Tamoxifen
Regulation of the cell cycle by protein kinases
What are the main points of the Insulin Receptor Signal Transduction?
1. Autophosphorylation
2. Tyr-Kinase activity
3. IRS-1 is phosphorylated
4. Sequential formation of complexes
5. Low Mr G-proteins are activated
6. Protein phosphorylation cascade
7. Nuclear event
What are the responses of the insulin signal transduction pathway?
*inducing the expression of proteins
*glycogen synthesis
What is the Scatchard analysis used for?
Estimation of the dissociation constant (Kd)
Number of receptor-binding sites in a given preparation
What can we find with the Scatchard analysis?
Kd
Bmax-then number of unoccupied sites
What are SH2 domains and what do they do?
(insulin receptor signal transduction)
Bind phosphotyrosine residues with high affinity
Present on many signaling proteins
What do G-proteins do? What is it stimulated by?
Stimulated by activated receptor
Exchanges bound GDP for GTP
What are the 5 Main Classes of Reactions that cells use in Metabolic Pathways?
1. Oxidation-reduction
2. Reactions that make or break C-C bonds
3. Internal rearrangements, isomerizations, and eliminations
4. Group Transfer Reactions
5. Free radical Reactions
Phosphoryl groups yield energy as what?
Group transfer reactions
The PPi product of adenylylation is hydrolyzed by what?
Inorganic pyrophosphatase
Reaction that Nucleoside diphosphate kinase catalyzes
ATP + NDP (or dNDP) ←→ADP + NTP (or dNTP) G’°= 0
Reaction that adenylate kinase catalyzes
2ADP←→ATP + AMP G’°= 0
Reaction that creatine kinase catalyzes
ADP + PCr←→ATP + Cr G’°= -12.5
Reaction that polyphosphate kinase-1 catalyzes
ATP + polyPn ←→ ADP + polyPn+1 G’°= -20
Reaction that polyphosphate kinase-2 catalyzes
GDP + polyPn+1 ←→ GTP + polyPn
What metabolic pathways occur in the cytosol?
Glycolysis, pentose phosphate pathway, fatty acid biosynthesis, gluconeogenesis,
What metabolic pathways occur in the ER (rough)?
Protein synthesis
What metabolic pathways occur in the ER (smooth)?
Lipid and steroid biosynthesis
Where does gluconeogenisis and storage of fats take place?
Gluconeogensis-liver
Storage of fats-adipose tissue
What are the 3 pathways of glucose utilization?
1.glycogen, starch, sucrose (storage)
2. pyruvate→citric acid cycle (energy)
3. Ribose 5-phosphate→nucleotide biosynthesis
What are the 3 fates of pyruvate? What conditions allow them to happen?
1. Aerobic conditions: 2 Acetyl CoA oxidized via citric acid cycle to 4CO2 + 4H2O
Animal, plant, and many microbial cells
2. Anaerobic conditions: 2 ethanol + 2 CO2
Fermentation to alcohol in yeast
3. Anaerobic conditions: 2 lactate
Fermentation to lactate in vigorously contracting muscle, erythrocytes, some other cells, and in some microorganisms
What is the main goal of glycolysis?
To convert energy from glucose into ATP
What are 2 ways energy is stored in the form of ATP?
Substrate-level phosphorylation
S-P + ADP → S + ATP
Oxidative phosphorylation
NADH → NAD+ + H+
What drives ATP-synthesis?
Proton gradient across the mitochondrial membrane
What processes of glycolysis liberate energy? What happens?
Oxidation processes. Glucose is oxidized and NAD+ is reduced to NADH
Phosphate is ionized what what pH? What does this do?
7 (net negative charge)
prevents glucose to diffuse across the membranes
Phosphoryl groups do what to help make ATP?
They conserve energy used for ATP
What does binding of Mg2+ -phosphate group to active site do?
Provides additional binding energy that lowers activation energy
What is a kinase?
Enzymes that catalyze the transfer of the terminal phosphoryl group from ATP to an acceptor nucleophile
Glycolysis: 1st step of the preporatory phase + enzyme
Phosphorylation of Glucose:
Glucose is phosporylated by ATP at C-6 to yield glucose 6-phosphate
Catalyzed by hexokinase
Which kinase requires Mg2+ for its activity?
Hexokinase
What are isozymes?
2 enzymes that catalyze the same reaction but are encoded in different genes
Glycolysis: 2nd step of the preporatory phase + enzyme
Isomerization
Conversion of glucose 6-phosphate to fructose 6-phosphate (an aldose to a ketose)
Catalyzed by phosphohexose isomerase
Glycolysis: 3rd step of the preporatory phase + enzyme
Phosphorylation (1st committed step)
Fructose 6-phosphate is phosphorylated by ATP to yield fructose 1,6-bisphosphate
Catalyzed by phosphofructokinase-1 (PFK-1)
Glycolysis: 4th step of the preporatory phase + enzyme
Cleavage of 6-C sugar into 3-C units
Fructose 1,6-bisphosphate is cleaved into glyceraldehydes 3-phosphate (aldose) and dihydroxyacetone phosphate (ketose)
Catalyzed by aldolase
Glycolysis: 5th step of the preporatory phase + enzyme
Interconversion of the triose phosphates
Dihydroxyacetone phosphate is converted to glyceraldehydes 3-phosphate
Catalyzed by triose phosphate isomerase
Glycolysis: 1st step of the payoff phase + enzyme
Oxidation of glyceraldehyde 3-phosphate and formation of NADH
Glyceraldehydes 3-phosphate is oxidized to yield 1,3-bisphosphoglycerate
Catalyzed by glyceraldehydes 3-phosphate dehydrogenase
1st energy-conserving reaction of glycolysis
Energy of the oxidation of glyceraldehyde 3-phosphate is conserved how? Why?
In the formation of acyl phosphate.
It has very high phosphoryl group transfer potential
Glycolysis: 2nd step of the payoff phase + enzyme
Substrate level phosphorylation: formation of the 1st 2 ATP molecules
Phosphoryl transfer from 1,3-bisphosphate to ADP yields 3-phosphoglycerate and ATP
Catalyzed by phosphoglycerate kinase
Glycolysis: 3rd step of the payoff phase + enzyme
Conversion to form a 2nd molecule with high phosphoryl group transfer potential
Enzyme is initially phosphorylated on a His residue. Phosphoryl group transfers to 3-phosphoglycerate, forming 2,3-BPG. Phosphoryl group is transferred to same His residue, producing 2-phosphoglycerate.
Catalyzed by phosphoglycerate mutase
Glycolysis: 4th step of the payoff phase + enzyme
Conversion to form PEP
A molecule of water is removed from 2-phosphoglycerate to yield phosphoenolpyruvate (PEP)
Catalyzed by enolase
Glycolysis: 5th step of the payoff phase + enzyme
Substrate level phosphorylation: formation of the 2nd 2 molecules of ATP
Phosphoryl group is transferred from phosphoenolpyruvate to ADP, yielding pyruvate
Catalyzed by pyruvate kinase
Which step of glycolysis is most heavily controlled?
The 3rd: phosphorylation
Anaerobic and aerobic glucose consumption deliver how many molecules of ATP?
Anaerobic-2
Aerobic-32
Equation for anaerobic glucose consumption
Glucose + 2 ADP + 2 Pi + 2 NAD+ → 2 pyruvate + 2 ATP + 2 NADH + H+ + 2 H2O
Equation for aerobic glucose consumption
Glucose → 6 CO2 + 2 ATP + 2 GTP + 10 NADH + 2 FADH2
What is the main purpose of pyruvate’s 3 fates?
To regenerate NAD+
Cancer cells depend on what for energy production?
Glycolysis
What is fermentation?
Process in which energy is extracted without the consumption of oxygen
What ferments milk to yogurt?
Lactobacillus bulgaris
What ferments milk to propionic acid and CO2?
Propionibacterium freudenrechii
The reduction of pyruvate to lactate is catalyzed by what?
Lactate dehydrogenase
What is the 1st step of converting pyruvate to ethanol + enzyme
Pyruvate is decarboxylated in an irreversible reaction yielding acetaldehyde
Catalyzed by pyruvate decarboxylase. (enzyme our body doesn't have)
What is the 2nd step of converting pyruvate to enthanol + enzyme
Acetaldehyde is reduced to enthanol
Catalyzed by alcohol dyhydrogenase
What does pyruvate decarboxylate require? And what is its tightly bound coenzyme?
Mg2+
Thiamine pyrophosphate (TPP)
What happens when your body digests carbohydrates? (steps)
Starch, glycogen, sucrose→oligosaccharides, di- and trisaccharides→oligosaccharides, di- and trisaccharides (chyme)→lactose→glucose/galactose
What enzymes catalyze the digestion of carbohydrates?
Salivary amylase in mouth/esophagus (degrades and hydrolyzes sugar)
Acid hydrolysis in stomach (degrades further)
α-amylase in small intestine
lactase
What does glycogen phosphorylase do? What kind of reaction does it take place in?
Catalyze an attack by Pi on the cose residues at the nonreducing end, generatings glucose 1-phosphate and a polymer one glucose unit shorter
Phosphorolysis
What removes the branches (α1→6) for glycogen phophorylase?
Debranching enzyme
What is a mutase?
An enzyme that catalyzes the transfer of a functional group from one position to another in the same molecule
What must happen to disaccharides before they enter cells?
They must be hydrolyzed to monosaccharides
Conversion of galactose 1-phosphate to glucose 1-phosphate involves with 2 nucleotide derivatives?
UDP-galactose and UDP-glucose
What is glycogenolysis?
Catabolic Conversion of glycogen to glucose 6-phosphate
What is glycolysis?
Catabolic Conversion of glucose 6-phosphate to pyruvate
What is gluconeogenisis?
Anabolic Conversion of pyruvate to glycogen
What is glycogenesis?
Anabolic conversion of glucose to glycogen
What is glycogen?
What is the structure?
Where is it located?
It is a Major storage form of glucose in animals
α1→4 glycocidic bonds and every 8-10 glucose molecules is α1→6 glycolytic branch
Mainly in the liver and muscle
What amount of glucose is stored as individual glucose and glycogen in hepatocyte?
Individual glucose: .4M
Glycogen: .01 μM
What does the brain rely on for fuel?
Glucose, not fatty acid
How long is liver glycogen around for?
1 day
Where is glycogen stored?
Large cytosolic β-granules, 20-40 granules form rosettes
What does glycogen phosphorylase do?
Attacks α1→4 glycosidic bond at the non reducing end
What enzyme is used in glycogenolysis?
Phosphoglucomutase
What does a debranching enzyme do?
Shifts 3 glucose molecules to nearby non reducing end. Single glucose is released.
What happens with glycogen mobilization in the muscle?
Glycogen is converted to gluc-6-P, which directly enters glycolysis
What happens with glycogen mobilization in the liver?
Glycogen is converted to gluc-6-P to glucose, which is released into the blood stream to supply brain with glucose during meals
What is our body’s daily demand for glucose? What is our supply?
Demand: ~160 g (120 g for brain)
Supply: ~20 g body fluids, ~190 g glycogen
What does gluconeogenesis do? Occurs where? What does it usually start with?
Maintains steady glucose level in the blood for muscles and brain
Occurs mainly in liver and kidney
Starts usually with pyruvate and lactate
What 3 enzymes in glycolysis are not in gluconeogenesis?
Hexokinase
Phosphofructokinase-1
Pyruvate kinase
What does the first bypass in gluconeogenesis involve? + enzyme
Conversion of pyruvate to oxaloacetate, catalyzed by pyruvate carboxylate
Pyruvate is reduced to malate, catalyzed by malate dehydrogenase
Oxaloacetate is converted to PEP, catalyzed by phosphoenolpyruvate carboxykinase
What does the 2nd bypass in gluconeogenesis involve? +enzyme
Conversion of fructose 1,6-bisphosphate to fructose 6-phosphate
Catalyzed by fructose 1,6-bisphosphatase
What is the 3rd bypass in gluconeogenesis involve? +enzyme
dephosphorylation of glucose 6-phosphate to glucose
Catalyzed by glucose 6-phosphatase
What does pyruvate carboxylate require?
Acetyl-CoA as a positive effector
What are precursors to gluconeogenisis. In animals, plants, and both.
Animals-lactate
Plants-CO2 fixation
Both-triacylglycerols
What is the formula for gluconeogenesis?
2 pyruvate + 4 ATP + 2 GTP + 2 NADH + 6H2O → glucose + 4 ADP + 2 GDP + 6Pi + 2 NAD+ + 2 H+
What are the intermediates of the citrate acid cycle? What is special about these?
Citrate
Isocitrate
α-ketoglutarate
succinyl-CoA
succinate
fumarate
malate
they can all undergo oxidation to oxaloacetate
What are glucogenic intermediates?
1. citric acid cycle intermediates
2. glucogenic amino acids
Do mammals have a net conversion of fatty acid into glucose? Why?
No. there is no pathway that converts acetyl-CoA into pyruvate
Where does glycogen synthesis mainly happen? What is its starting point?
Mainly in the liver and muscle
Starting point is gluc-6-P
What enzyme converts glucose-6-P to Gluc-1-P?
Phosphoglucomutase
What enzyme converts gluc-1-P to UDP glucose?
UDP-glucose pyrophosphorylase
UDP glucose are key intermediates for what?
Polymeration reactions
Vit. C synthesis
What is an excellent leaving group on UDP-glucose?
Nucleotidyl group
Formation of what is irreversible?
UDP-glucose
What enzyme is used in the reaction converting UDP-glucose and glycogen to glycogen and UDP?
Glycogen synthase
What is glycogenin?
The primer on which new chains are assembled during glycogen synthesis and the enzyme that catalysizes their assembly
What is the main goal of metabolic pathways?
To maintain homeostasis
Why do metabolic pathways want to maintain AMP?
AMP changes very dramatically because AMP starts out so low to begin with
What enzyme catalyzes the conversion of 2 ADP to AMP and ATP?
Adenylate kinase
What does the regulation of carbohydrate catabolism do?
• Prevent futile cycles (glycolysis-gluconeogenisis)
• Partition metabolites appropriately between alternative pathways
• Shut down pathways when products accumulate
• Use pathways best suited for the organism
What characteristics do the most heavily controlled (usually irreversible) steps of carbohydrate catabolism have?
• Equilibrium is far on the side of the product
• Regulated by the enzyme activity
• Become the rate limiting steps of the pathway (valve)
• Sit on branching points of a pathway (committed step)
What are the 4 isozymes of hexokinase?
• Muscle hexokinase I-III
• Liver hexokinase IV (glucokinase)
What inhibits muscle hexokinases I and II?
Their product: glucose-6-phosphate
What inhibits hexokinase IV?
The reversible binding of a regulatory protein specific to liver.
What is the most important control element in mammalian glycolysis?
Phosphofructokinase-1
What is Phosphofructokinase-1?
Most heavily regulated protein of glycolysis
Tetrameric protein with catalytic sites and regulatory sites in the same subunit
What happens when PFK is active and inactive?
Active-glycolysis
Inactive-pentose phosphate pathway (ribose biosynthesis)
What regulates PFK-1?
ATP-ATP binds to allosteric site and lowers the affinity for fruc-6-P
What is a very potent activator of PFK? What does it do?
Fructose-2,6 bisphosphate
It is an allosteric activator that shifts the conformation from the T-state to the R-state
It increases the affinity for fruc-6-P and dimishes the inhibitor effect of ATP
What does pyruvate kinase do?
Controls the outflow from glycolysis
Describe allosteric regulation of pyruvate kinase
• ATP inhibits
• Fruc 1,6-bisphosphate activates
Describe the covalent regulation of pyruvate kinase
• L-type pyruvate kinase (liver isozyme) is reversibly phosphrylated when glucose level is low-inactivates enzyme
What inhibit all isozymes of pyruvate kinase?
• High concentrations of ATP
• Acetyl-CoA
• Long-chain fatty acids
What is the decision point of gluconeogenesis vs. glycolysis?
Fruc-1,6 BP/Fruc-6-P
What are the 3 key enzymes of glycolysis?
1. hexokinase
2. phosphofructokinase-1
3. pyruvate kinase
What sugars enter the glucose feeder pathway?
• Trehalose
• Lactose
• Sucrose
• Fructose
• Galactose
What does fructose 6-phosphate do?
Activates glycolysis
Inactivates gluconeogenisis
High glucose level:
What is released?
What happens?
What enzyme?
Insulin is released=raised levels of Fruc-2,6-BP=activates PFK-1=glycolysis
Causes glycogen formation
Enzyme is glycogen synthase
Low glucose level:
What is released?
What happens?
What enzyme?
Glucogon is released=raised levels of cAMP=raised levels of PKA=lower levels of F-2,6-BP=gluconeogenesis
Causes glycogen mobilization
Enzyme is glycogen phosphorylase
Glycogen phosphorylase is regulated alssosterically by what? Covalently by what?
Allosterically by glucose (negative regulator)
Covalently by hormone-controlled phosphorylation (postive regulator)
What are the 3 major hormones that regulate fuel metabolism?
Epinephrine
Insulin
Glucogon
Epinephrine:
Secreted from what?
Produced when?
Increases what?
Results in what?
Secreted from adrenal medulla
Produced at moments of stress
Increases strength of heart beat, raises BP, increases flow of oxygen and nutrients to tissue
Results in Increase in blood glucose
Glucogon:
Released by what?
Prime target is what?
Results in what?
Released by pancreas
Prime target is liver
Results in increase in blood glucose
Insulin:
Produces where?
Stimulates what?
Results in what?
Produced in the pancreatic β-cells
Stimulates glucose uptake in muscle
Stimulates fatty acid synthesis
Results in decreased blood glucose level, glycogen formation in muscle, fatty acid synthesis in adipose tissue
What happens in diabetes?
Cells do not have enough glucose (starving)
Triacylglycerol hydolysis, gluconeogenesis and ketone body formation is accelerated
High blood glucose level, glucose spills into urine
Ketone bodies are acidic, kidneys try to maintain blood pH and secret H+ and other ions=dehydration.
What happens in Type 1 diabetes?
Autoimmune disease where antibodies destroy β cells of pancreas=no more insulin
Degenerative disease:
Blindness (high glucose levels)
Kidney failure
What happens in Type 2 Diabetes? Multi-step process:
1. Insulin resistance-cells are unable to use insulin efficiently normal or elevated levels
2. Hyperglycemia-stimulates additional insulin secretion; glucose is fairly constant but insulin is highter
3. Pre-Diabetes-β-cells become exhausted and can no longer produce enough insulin=blood glucose is higher
4. Diabetes-untreated pre-diabetes
What are 3 hormones that fat cells secrete? Which one is related to diabetes?
1.leptin (low in diabetes)-after each mean, fat cells signal appetite
2. resistin-too much causes insulin resistance
3. adiponectin-to little causes insulin resistance
What molecules can be transformed into acetyl-CoA?
• Carbohydrates
• Fatty acids
• Amino acids
What are the 3 stages of cellular respiration?
1. pyruvate, fatty acids, and a.a. are oxidized to 2-C fragments (acetyl-CoA)
2. Acetyl groups are fed into citric acid cycle and oxidized to CO2; energy is conserved as reduced NADH and FADH2
3. Reduced NADH and FADH are oxidized and e- are transferred to oxygen via a chain of e- carrying molecules (respiratory chain), this induces the formation of a proton gradient across the mitochondrial membrane which drives ATP synthesis (oxidative phosphorylation)
What is the decarboxylation of pyruvate to acetyl-CoA catalyzed by?
Pyruvate dehydrogenase complex (PDC)
What does Pyruvate dehydrogenase complex (PDC) consist of?
Dihydrolipoyl DH
Pyruvate DH
Dihydrolipoyl transacetylase
Pyruvate dehydrogenase:
Number of chains
Prosthetic group
Reaction catalyzed
12
TPP
Oxidative decarboxylation of pyruvate
Dihydrolipoyl transeacetlyase:
Number of chains
Prosthetic group
Reaction catalyzed
24
lipoamide
transfer of the acetyl group to CoA
Dihydrolipoyl dehydrogenase
Number of chains
Prosthetic group
Reaction catalyzed
24
FAD
Regeneration of the oxidized form of lipoamide
What are 2 stoichiometric cofactors of PDC?
Coenzyme A
NAD+
Oxidative decarboxylation of pyruvate to acetyl-CoA by the PDH complex:
Step 1 + enzyme
Decarboxylation of pyruvate
Catalyzed by pyruvate dehydrogenase component
Oxidative decarboxylation of pyruvate to acetyl-CoA by the PDH complex:
Step 2 + enzyme
Oxidaton of hydroxyethyl-TPP and transer onto dihydrolipoyl transacetylase
Catalyzed by pyruvate dehydrogenase component
Oxidative decarboxylation of pyruvate to acetyl-CoA by the PDH complex:
Step 3 + enzyme
Oxidation of hydroxyethyl-TPP and transfer onto dihydrolipoyl transacetylase
Catalyzed by dihydrolipoyl transacetylase
Oxidative decarboxylation of pyruvate to acetyl-CoA by the PDH complex:
Step 4 + enzyme
Oxidation of hihdrolipoamide
Catalyzed by dihydrolipoyl dehydrogenase
What is Beriberi?
Based on what?
What is dry/wet beriberi?
Neurological and cardiovascular disorder
Based on thiamine (Vit. B1) deficiency
Dry beriberi- damage to peripheral nervous system-pain in limbs, weakness of muscles, distorted skin sensation
Wet beriberi- damage to cardiovascular system-enlarged heart, increased cardiac output
Thiamine deficiency:
Affects what?
Increases in what?
Affects the multienzyme complexes that contain enzymes with TPP cofactor:
1. Pyruvate dehydrogenase complex
2. α-ketoglutarate dehydrogenase complex
3. transketolase
Increases in pyruvate and α-ketoglutarate dehydrogenase concentration
What happens with arsenic and mercury poisoning?
What happens with arsenic and mercury poisoning?
Krebs cycle
1st step + enzyme
Condensation of acetyl-CoA with oxaloacetate to for citrate
Catalyzed by citrate synthase
Krebs cycle
2nd step + enzyme
Transformation of citrate (to cis-aconitate) to isocitrate
Catalyzed by aconitase
Krebs cycle
3rd step + enzyme
Oxidation of isocitrate to α-Ketoglutarate, formation of NADH
Catalyzed by isocitrate dehydrogenase
Krebs cycle
4th step + enzyme
Oxidation of α-Ketoglutarate to Succinyl-CoA + CO2, formation of NADH
Catalyzed by α-ketoglutarate dehydrogenase complex
Krebs cycle
5th step + enzyme
Conversion of Succinyl-CoA to Succinate
Catalyzed by succinyl-CoA synthaetase
Krebs cycle
6th step + enzyme
Oxidation of Succinate to Fumarate, formation of FADH2
Catalyzed by Succinate Dehdrogenase
Krebs cycle
7th step + enzyme
Hydration of Fumarate to L-Malate
Catalyzed by fumarase
Krebs cycle
8th step + enzyme
Oxidation of L-Malate to Oxaloacetate, formation of NADH
Catalyzed by malate dehydrogenase
How many ATP does one turn of the citric acid cycle produce?
10
What does 1 glucose yield?
2 pyruvate + 2 ATP
What does 1 pyruvate yield?
2 acetyl CoA + 2 CO2 + 2 NADH
What does 1 acetyl CoA yield?
4 CO2 + 6 NADH + 2 FADH2 + 2 GTP
What does 1 glucose ultimately yield?
6 CO2 + 2 ATP + 2 GTP + 10 NADH + 2 FADH2
What are 3 ways you can regenerate oxaloacetate?
Pyruvate (gluconeogenesis)
Phosphoenolpyruvate
Malate
What is biotin?
Cofactor of pyruvate carboxylase
Carrier or CO2
What is the 1st level of regulation of the citric acid cycle?
Control: Entry
Amount of acetyl-CoA that enters cell cycle is strictly regulated (Pyruvate dehydrogenase complex)
What is the 2nd level of regulation of the citric acid cycle?
Control
The irreversible reactions of the citric acid cycle:
• citrate synthase
• isocitrate dehydrogenase
• α-ketoglutarate dehydrogenase
How are different levels of the citric acid cycle inhibited?
• Product inhibition
• Allosteric feedback inhibition
• Inhibition by intermediates that reflect high energy state.
What is the pyruvate dehydrogenase activated and inactivated by?
• Activated by molecules that signal high energy level (ATP, NADH, fatty acids)
• Inactivated by molecules that signal low energy level (AMP, CoA, NAD+, Ca2+)
What is glyoxylate cycle and where does it take place?
Converstion of acetate to succinate (→ glucose)
Takes place in certain plants and microorganisms
Why doesn’t glyoxylate take place in animals?
Formation of acetyl-CoA is too exergonic to e reversible
Glyoxylate Cycle: Bacteria and bacteria/plants
Bacteria: grow on acetate as only carbon sourse
Bacteria and plants: convert acetyl-CoA to succinate
What are the 2 unique enzymes in the glyoxylate cycle?
Isocitrate lyase
Malate synthase
Acetyl Co-A in glyoxylate cycle vs. citric acid cycle
Glyoxylate: 2
Citric acid: 1
What is the 1st step of the glyoxylate cycle + enzyme?
Conversion of Acetyl-CoA to Citrate
Catalyzed by citrate synthase
What is the 2nd step of the glyoxylate cycle + enzyme?
Conversion of citrate into isocitrate
Catalyzed by aconitase
What is the 3rd step of the glyoxylate cycle + enzyme?
Conversion of isocitrate into glyoxylate, release of succinate
Catalyzed by isocitrate lyase
What is the 4th step of the glyoxylate cycle + enzyme?
Conversion of glyoxylate into malate, addition of Acetyl CoA
Catalyzed by malate synthase
What is the 5th step of the glyoxylate cycle + enzyme?
Oxidation of malate into oxaloacetate, releases 1 NADH
Catalyzed by malate dehydrogenase
Why is isocitrate a crucial intermediate?
Isocitrate dehydrogenase acts as a covalent modifier
• Phosphorylation inactivates
• Dephosphorylation activates
• Reduced cellular energy supply activates phosphatase
Where does the glyoxylate cycle take place?
In plants in the glyoxysomes (specialized peroxisomes)
What does glyoxylate cycle allow plants and some microorganisms to do?
Use fat to make carbohydrates
What happens to succinate after it leaves the glyoxylate cycle?
It is exported to the mitochondria, where it is transformed to malate.
What are the 3 stages of the Calvin cycle?
1. CO2 fixation into 3-phosphoglycerate
2. Reduction: Converstion of 3-phosphoglycerate to glyceraldehyde 3-phosphate
3. Regeration: regeneration of ribulose 1,5-bisphosphate from triolose phosphates
Calvin cycle: 1st step
3 CO2 + H2O combine with ribulose 1,5-bisphosphate to form phosphoglycerate
Calvin cycle: 2nd step
Phosphoglycerate to 1,3-bisphosphoglycerate
6 ATP to 6 ADP
Calvin cycle: 3rd step
1,3-bisphosphoglycerate to glyceraldehyde 3-phosphate/dihydroxyacetone phosphate
6 NADPH + 6 H+ to 6 NADP+ + 6 Pi
Calvin cycle: 4th step
release glyceraldehyde 3-phosphate
Calvin cycle: 5th step
glyceraldehyde 3-phosphate/dihydroxyacetone phosphate to ribulose 5-phosphate
release 2 Pi
Calvin cycle: 6th step
ribulose 5-phosphate to ribulose 1,5-bisphosphate
3 ATP to 3 ADP