• 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

1/856

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;

856 Cards in this Set

  • Front
  • Back
In the "native structure" of a protein, where are the hydrophobic residues found? Hydrophilic residues?
- Hydrophobic residues are mostly buried in the interior
- Hydrophilic residues are mostly on the surface
Which of the amino acid residues is NOT achiral / optically inactive? Why?
- Glycine
- It's α-Carbon is attached to 2 H atoms
What are the two mirror image forms of a chiral amino acid called?
Enantiomers
Which configuration are all amino acids (except glycine) found in mammalian proteins?
L-configuration
How do you draw an amino acid in the "L-configuration"?
- α-C in center
- NH3+ wedged to left
- COO- dashed to top
- H wedged to right
- R group dashed down
What are the two categories of amino acids and the two sub-categories for each?
Hydrophobic / Apolar:
- Aliphatic
- Aromatic
Hydrophilic / Polar:
- Uncharged
- Charged
Which amino acids are categorized as hydrophobic/apolar and aliphatic?
- Glycine (Gly, G)
- Alanine (Ala, A)
- Valine (Val, V)
- Leucine (Leu, L)
- Isoleucine (Ile, I)
- Methionine (Met, M)
- Proline (Pro, P)
What is unique about the amino acid Proline (Pro, P)?
- Proline has an imino group (2 N-C bonds)
- All other amino acids have an amino group (1 N-C bond)
Which amino acids are categorized as hydrophobic/apolar and aromatic?
- Phenylalanine (Phe, F)
- Tyrosine (Tyr, Y)
- Tryptophan (Trp, W)
How can protein concentration be estimated?
Measuring the absorbance at 280 nm; aromatic side chains (Trp, Tyr, Phe) have characteristic UV absorbance at 280nm
Which amino acids are categorized as hydrophilic/polar and uncharged?
- Serine (Ser, S)
- Threonine (Thr, T)
- Asparagine (Asn, N)
- Glutamine (Gln, Q)
- Cysteine (Cys, C)
Which amino acids are categorized as hydrophilic/polar and charged? What charges do these residues have?
- Lysine (Lys, K) +
- Arginine (Arg, R) +
- Histidine (His, H) +
- Aspartate (Asp, D) -
- Glutamate (Glu, E) -
Positively charged residues are acidic or basic? Which residues?
Basic: Lys, Arg, His
Negatively charged residues are acidic or basic? Which residues?
Acidic: Asp, Glu
What does the pKa of a group tell you?
The pKa is the pH at which it is 50% protonated and 50% unprotonated
When the pH is LOWER than the pKa, what is the status of that group?
- Higher concentration of H+ in solution
- >50% of group is protonated
When the pH is HIGHER than the pKa, what is the status of that group?
- Lower concentration of H+ in solution
- <50% of group is protonated
What equation can be used to determine the ratio of protonated (HA) to unprotonated (A-) forms?
Henderson-Hasselbalch Equation:
pH = pKa + log[A-]/[HA]
What does the isoelectric point, pI represent?
An intermediate pH characteristic of the amino acid, peptide or protein in which the net charge is zero
What does it mean for the pH to be "buffered"?
When the pH is near the pKa of the residue/protein it takes significantly more acid or base to change the pH than when further from the pKa; therefore the solution is STABILIZED near the pKa values of the ionizable groups
Which important molecules in the body help "buffer" the intracellular and extracellular fluids to keep cells, tissues, and organisms surviving?
- Amino acids, peptides, proteins
- Phosphate
- Bicarbonate
In what type of proteins are disulfide linkages common? Where are they uncommon?
- Common: extracellular proteins
- Uncommon: intracellular proteins
Which hormone is highly dependent upon the increased conformational stability due to disulfide bonds?
Insulin (contains 3 disulfide bonds)
What features of the peptide bond limit the number of conformational states possible for a polypeptide?
1. Strong electronegativity of C=O relative to amide N leads to resonance structures; prevents bond rotation around the amid bond
2. Trans conformation of R groups is preferred due to lower energy / more stable / less steric hindrance
Simply, what determines the protein's folding and 3D structure?
The sequence of amino acids in a protein (the primary structure)
What are the two types of secondary structures of proteins?
- Right-handed α-helix
- β-pleated sheet (parallel or antiparallel or mixed)
What is it called when secondary structures make stable associations with one another?
Super-secondary structures
What is a domain?
A structure above super-secondary structures that is the smallest thermodynamically stable unit of protein structure; often accounts for part of an enzyme's active site or may catalyze one part of a complex reaction sequence
What does the tertiary structure represent? How many domains are in a tertiary structure?
- Final thermodynamically stable structure of a single polypeptide chain
- One or more domains (often 2 or 3)
When multiple polypeptide chains come together to form a globular protein, this level of structure is called what?
Quaternary structure
What are three examples of fibrous proteins we need to know about?
- α-Keratin
- Collagen
- Elastin
What is α-Keratin a primary component of?
Hair and nails
What is the structure of α-Keratin?
- Two right handed α-helices
- α-helices are intertwined to form a left-handed supercoil called an α-coiled coil
- These can bond non-covalently (H-bonds, hydrophobic intxns, and ionic bonds) and covalently (disulfide bonds)
What distinguishes the α-Keratin found in hair with that which is found in nails?
- α-Keratin in hair has a moderate number of disulfide bonds
- α-Keratin in nails has a high number of disulfide linkages to make it rigid
What is the most abundant protein in the body?
Collagen (20-25% of total protein)
What is the structure of Collagen?
- Three collagen chains -Gly-X-Y-(X often proline, Y often hydroxyproline or hydroxylysine), called α-chains, are left-handed helices
- These three are wrapped around each other in a right-handed triple helix
- Multiple triple helices interdigitate to form collagen microfibrils
What amino acids are important to the structure of Collagen? Why?
- Proline - stack long outside of helix providing rigidity
- Glycine - found internally in the helix due to small R group (every third residue)
Which amino acids are often found on the outside of the triple helix of Collagen? Why is this significant?
- Proline, hydroxyproline and hydroxylysine
- These form bonds between neighboring chains resulting in aggregation into long fibers
Why is Vitamin C important for Collagen formation?
- Vitamin C / Ascorbate is required to catalyze the hydroxylation reactions to form hydroxyproline and hydroxylysine
- These residues are important for stabilizing the triple-helical structure
If there is a vitamin C / ascorbate deficiency, what can happen?
Scurvy (symptoms include bleeding gums) due to decreased tensile strength of collagen
Where is the protein elastin commonly found?
Connective tissue: lungs, large arterial walls, elastic ligaments
What amino acids predominantly make up elastin?
- Small, non-polar amino acids (e.g., glycine, alanine, and valine)
- Proline and lysine
Which connective tissue protein has rubber-like properties?
Elastin
What does the enzyme elastase do? What can inhibit elastase?
- Elastase is an enzyme inhibitor of elastin (which can destroy the alveolar epithelium)
- Elastase is inhibited by α1-antitrypsin
What clinical problem can occur if there is not enough of α1-antitrypsin (elastase inhibitor)?
Emphysema (due to the destruction of the alveolar epithelium)
What is a gene family?
- Similar genes formed by duplication of an original gene; the genes have similar biochemical functions
- Hemoglobin has two gene families, α-globin and β-globin
What happens within a gene family during development? Specifically with globins?
- The expression of members of a gene family change during development
- Composition of the Hb tetramer changes with gestational and post-partum age
- I.e., ζ (zeta) α-globin is expressed briefly embryonically and then only α is expressed throughout fetal growth and adulthood
- I.e., ε (epsilon) β-globin is expressed embryonically, followed by γ (gamma) during fetal growth and later by β
How does the structure of myoglobin compare to hemoglobin?
- Myoglobin is a monomeric protein
- Hemoglobin is a heterotetrameric protein (αβ)2
- The α and β subunits are similar sequentially and structurally and evolutionarily related to myoglobin
- Both contain a Fe2+-protoporphyrin IX prosthetic group that binds O2
How does the function of myoglobin compare to hemoglobin?
- Hemoglobin transports O2 from the lungs to peripheral tissues
- Myoglobin is found in muscle and is designed to store O2
Why is it necessary for hemoglobin to demonstrate cooperativity of oxygen binding?
- When Hb is in the muscles (resting or working) the pO2 is much lower (due to the consumption of O2)...this leads to Hb releasing its O2
- In the lungs, the pO2 is higher, which leads to increased binding of O2 to Hb
How does hemoglobin demonstrate cooperativity of oxygen binding?
- Binding of a single oxygen to one Hb subunit induces a conformation change that is partially transmitted to adjacent subunits
- This induces an increased affinity for oxygen
- As more oxygens bind to subunits the affinity is increased and the molecule transitions from the T (tense) state to the R (relaxed) state
In general, how do H+, CO2, and 2,3-DPG regulate O2 binding to hemoglobin?
All act as allosteric regulators/effectors, which means they bind at a site other than the active site to induce a conformational change that reduces the Hb's affinity for O2
Specifically, how does [H+] regulate O2 binding to hemoglobin?
- As pH lowers (or [H+] increases), the affinity of Hb for O2 decreases
- This leads to an increase in release of O2 in the tissues that need it
- Catabolism can continue
- H+ binding results from a shift in pKa of specific residues
Specifically, how does [CO2] regulate O2 binding to hemoglobin?
- As [CO2] increases, there is a lower affinity for O2
- CO2 builds up in tissues that are actively catabolizing fuel molecules, therefore more O2 is required to continue working
What kind of effectors / regulators are H+ and CO2?
Heterotropic Negative Allosteric Effectors
- Heterotropic because they are not O2
- Negative because they decrease affinity for O2
- Allosteric because they bind to a site other than that which O2 binds to
Specifically, how does 2,3-DPG regulate O2 binding to hemoglobin?
- 2,3-DPG is a negative allosteric effector
- It binds to a cleft between the β subunits
- Binding is stabilized by ionic intxns
- RBCs have regulatory mechanisms to control the conc. of 2,3-DPG so as to fine tune the affinity of Hb for O2
How does O2 affect O2 binding to hemoglobin?
- O2 is a positive homotropic allosteric effector of O2 binding
- It is a negative allosteric effector of H+ and CO2 binding
- This reciprocal relationship between O2 and H+ is termed the Bohr effect or isohydric shift
What is the Bohr effect (aka isohydric shift)?
- O2 is a negative allosteric effector of H+
- H+ is a negative allosteric effector of O2
- This reciprocal relationship between O2 and H+ is called the Bohr Effect
What is the molecular basis of sickle cell anemia?
- Homozygous recessive disease
- Caused by a point mutation in adult β-globulin gene, leads to substitution of Valine (Val) from Glutamic Acid (Glu)
- Val is hydrophobic (Glu is hydrophilic) and creates a sticky patch on deoxyHb that leads to polymerization of Hb tetramers
- This leads to the sickle cell shape which often blocks microcirculation as well as a shorter half-life
What are the four common forms of Sickle Cell Disease (SCD)?
- Homozygous HbS disease (HbSS)
- HbS/βo thalassemia
- Hbs/β+ thalassemia
- HbSC
The "S" mutation of Hb causes what?
- Glu6 --> Val
- Glu is hydrophilic, Val is hydrophobic
- Val creates sticky patch on deoxyHb; leads to polymerization of Hb tetramers
- Leads to sickled shape and reduced deformability of RBCs
The "C" mutation of Hb causes what?
- Glu --> Lys
- Negative to positive charge
- Interacts with membrane causing formation / sickling problems
The βo thalassemia mutation leads to what in Hb?
- Lack of β chain
- Still have two α chains
The β+ thalassemia mutation leads to what in Hb?
- Reduced β chain
- RBC are smaller due to less Hb
What are common complications of SCD?
- Vaso-occlusive crises
- Hemolysis and anemia
- Ischemia
- Chronic pain
- Dactylitis - painful/swelling of hands and feet (infants)
- Frequent infections (spleen is busy metabolizing RBC and is less capable of fighting infections)
- Delayed growth
- Vision problems
- Chest pain
- Renal problems
- Pulmonary hypertension
- Necrosis of bones
What components make up a nucleic acid?
- Nitrogenous base (purines and pyrimidines)
- Ribose or deoxyribose
- Phosphate group
What are the different nitrogenous bases and how are they categorized? How many rings does each kind have?
- Purine: Adenine, Guanine (2 rings)
- Pyrimidine: Cytosine, Uracil, Thymine (1 ring)
How can you distinguish between the purines, adenine and guanine?
- Both have two rings
- Guanine has a carbonyl carbon on the 6 membered ring and an amino (NH2) group
- Adenine has only an NH2 group on the 6 membered ring (no carbonyl)
How can you distinguish between the pyrimidines, cytosine, uracil and thymine?
- All have one ring
- Cytosine has a carbonyl and an amino (NH2) group
- Uracil has two carbonyl groups
- Thymine has two carbonyl groups and a methyl group
How do you distinguish between the two nucleic acid sugars, deoxyribose and ribose?
- Both have OH groups at 1', 3' and 5' positions
- Deoxyribose has an H at the 2' position (total of 3 OH groups)
- Ribose has an OH group at the 2' position (total of 4 OH groups)
What's the difference between a nucleoTide and a nucleoSide?
- NucleoTides have a base, sugar AND a phosphate
- NucleoSides have ONLY a base and a sugar
How are the components of a nucleotide connected?
- Sugar attaches to phosphates at 5' OH and 3' OH via phosphodiester bridge (backbone)
- Sugar attaches to the base at the 1' position
How do DNA and RNA differ?
- DNA uses the sugar deoxyribose
- RNA uses the sugar ribose
- DNA uses the base thymine
- RNA uses the base uracil
Describe the structure of double-stranded DNA.
- Right-handed, B-form helix is most common
-Major groove (12A) and minor groove (6A)
- Complementary strands run antiparallel to each other
- Bases are on inside of helix with bases stacked perpendicular to helix axis (promotes stability)
- Repeats after 10.5 residues / 36A
- Held together by hydrogen base pairing (AT and CG)
How many hydrogen base pairs keep adenine and thymine/uracil together?
Two hydrogen bonds
How many hydrogen base pairs keep guanine and cytosine together?
Three Hydrogen bonds
What are the three forms of DNA helices?
- A form = more symmetrical right handed helix
- B form = major and minor grooves; right handed; most common
- Z form = left handed helix
What are two ways to separate a double helix by disrupting hydrogen bonds?
Changes in pH and temperature
How does ionic composition affect denaturation of DNA?
- Ionic composition (salt concentration) effects denaturing and annealing RATES
How can the denaturation and renaturation of DNA be measured?
- Spectrophotometrically at 260 nm
- Single strands absorb better (free to absorb light)
- Double stranded DNA doesn't absorb as well because the stacking of bases blocks the UV light from absorption
If a DNA helix is being denatured, how will the absorbance at 260 nm be affected?
There will be an increase in absorbance due to less base stacking
If a DNA helix is being renatured, how will the absorbance at 260 nm be affected?
There will be a decrease in absorbance due to increased base stacking
What is the melting temperature (Tm) of DNA? What does it depend upon?
- Defined as the temperature at which half (50%) of the helical structure is lost
- Depends on % GC content
- Also affected by ionic strength
How is the Tm affected by a higher GC content?
The Tm will be reached at a higher temperature because GC base pairs have 3 H bonds vs 2 H bonds for AT pairs
How is the Tm affected by ionic strength? Explain.
- High salt favors duplex
- Low salt favors denatured
- Electrostatic repulsion caused by charged phosphate backbone; if there is more salt to neutralize those charges then the DNA strands will be less repelled by each other
How do linear and circular DNA differ?
- Circular can be relaxed or supercoiled due to the nature of twists adding tension
- Linear can feel some tension due to twisting if ends are anchored by proteins
Most DNA is negatively or positively supercoiled?
Negatively supercoiled; may aid in transcription and replication
What is the function of topoisomerases?
- Enzymes that change the topological state of circular DNA but not its covalent state
- Two types
Compare and contrast the differences between Type 1 and Type 2 Topoisomerases.
- Time 1: create transient single-stranded breaks in DNA; nicking-closing enzymes; can relax both negatively and positively supercoiled DNA; removes 1 twist at a time
- Type 2: create transient double-stranded breaks; requires ATP to complete cycle; removes 2 twists at a time
What are the steps by which Type 1 Topoisomerases function?
- Cleave on strand of DNA
- Pass a segment of DNA through the break
- Reseal the break
- (NO energy required)
What are the steps by which Type 2 Topoisomerases function?
- Two strands of DNA are cleaved (ATP required)
- DNA is passed through break
- Break is resealed
When is the function of topoisomerases necessary?
- When unzipping DNA twists are added to remaining helix
- Topoisomerases are important to reduce the tension
What drugs inhibit E.coli DNA gyrase (type 2 topoisomerase)? What are these drugs used to target?
- Nalidixic acid and Ciprofloxacin (quinolones)
- By only targeting bacteria they make for effective antibiotics
- Ciprofloxacin is used for anthrax
What drugs inhibit eukaryotic Type II topoisomerases? What are these drugs used to target?
- Etoposide and doxorubicin
- Used in cancer chemotherapy
- Specifically inhibit the Topo ATPase activity (increases rate of cleaving but reduces rate of resealing); this results in the arrest of DNA replication and RNA transcription
- Cancer cells are rapidly replicating and have higher levels of Type II topoisomerases making these drugs lethal to these cells and less damaging to slow-growing cells
What is the general hierarchy of chromatin packing?
- 1st order: nucleosome
- 2nd order: 30-nanometer fiber
- 3rd order: chromatin loop
- 4th order: miniband
- 5th order: chromosome
Describe the first order of chromatin structure.
- Nucleosome consists of ~200bp of DNA wrapped around octamer of core histone proteins
- Covalent modifications of core histones determine transcriptional activity of packaged genes
Describe the second order of chromatin structure.
- 30-nanometer fiber
- Six 200bp nucleosomes packaged in a solenoid arrangement into one 1200bp 30nm fiber
- Binding of linker histone is required to stabilize 30nm fiber
Describe the third order of chromatin structure.
- Chromatin loop
- Fifty 1200bp 30nm fibers form loops of chromatin containing ~60,000bp DNA
- Each loop is attached to the nuclear matrix at the base
- Contains one to several genes
Describe the fourth order of chromatin structure.
- Miniband
- Consists of a tandem of 18 chromatin loops = ~1 million bp DNA
- Encircles axis of mitotic chromosome
Describe the fifth order of chromatin structure.
- Chromosome
- Contains a single linear DNA molecule, packaging an average of 75 million bp of DNA (75 x minibands stacked on top of one another)
- Includes a centromere and two telomeres, loci, or repetitive DNA located in the middle and at ends of chromosome, respectively
What is "Chromatin"?
- Genomic DNA
- Nuclear proteins
What is the difference between "euchromatin" and "heterochromatin"?
- Heterochromatin - 10% of chromatin in typical cell; highly condensed and usually transcriptionally inactive
- Euchromatin - 90% of chromatin in typical cell; less condensed and usually transcriptionally active
Which level of the hierarchy of chromatin structure is related to looking like "beads on a string"? What makes up the "beads" and the "string"?
- 1st order: nucleosome
- Beads = nucleosome (half protein - octamer of histone and a few non-histones/half DNA)
- String = linkers (H1 histones)
What histones are found in a nucleosome?
- Two of each core histone:
- H2A
- H2B
- H3
- H4
- Form an octamer
Are histones acidic or basic? Which residues make them this way?
Basic; lots of lysine+ and arginine+
How can we know that histones are vital cellularly?
Highly conserved sequences
What is the function of histones?
- Structural role (wrapping DNA for packing)
- Inhibition of transcription
What is necessary for transcription of a gene to occur (re: histones)?
- Histones must be modified via acetylation of the Lysine+ and Arginine+ residues
- This neutralizes the positive charges
- Results in repulsion of histones from DNA gene sites causing chromatin "opening" allowing in transcriptional machinery
How can histones be modified to inhibit transcription?
- Methylation of Histone H3 causes formation of the non-transcribable "hetero"-chromatin
- OR Phosphorylation of histones usually repressed transcription by preventing the condensation chromatin into recognizable chromosomes as cell enters M-phase (mitosis)
What is meant by the term "non-histone"?
- Applies to any chromatin protein that is not a histone
- Includes DNA repair enzymes, DNA and RNA polymerases, and transcription factors
What are all of the ways in which histones can be modified? Which residues are impacted directly by these modifications?
- Acetylation - lysine
- Methylation - lysine and arginine
- Phosphorylation - serine and threonine
- Ubiquitination - lysine
- Sumoylation - lysine
- Usually affects residues on amino-terminal that protrude from nucleosome
By which distinct mechanisms do histone modifications affect chromosome function?
- Effects on electrostatic charge of histone and resulting changes in conformation (acetylation/deacetylation)
- Modifications which provide binding sites for regulatory proteins containing protein domains that recognize these modifications
What are epigenetic modifications?
Heritable changes in gene function that occur without a change in the sequence of the DNA
Functionally how does DNA methylation by DNA methyltransferases (DNMTs) affect gene expression?
DNA methylation represses gene expression by blocking binding of transcription factors that normally activate gene expression
Structurally how does DNA methylation by DNA methyltransferases (DNMTs) affect gene expression?
- Methylated CpG in DNA is recognized and bound by methyl CpG binding proteins (MBPs)
- MBPs recruit histone deacetylases (HDACs)
- HDACs carry out histone deacetylation
- Histones in de-acetylated state bind DNA more tightly which inhibits gene transcription by blocking binding of transcription activator proteins
- Additionally MBPs can recruit histone methyltransferases (HMTs)
- Methylated histones are recognized and bound by HP1 protein, completely silencing gene expression
How can epigenetic modifications directly affect cancer?
- These epigenetic modifications (methylation or deacetylation) may lead to the suppression of expression of tumor suppressor genes
- Leads to high rate of uncontrolled cell growth
What is the function of tumor suppressor genes?
- Help control cell growth by regulating the processes of mitosis and cell division
What is the role of Histone H1?
- Acts as the linker protein for the first level of chromatin packing (connects nucleosomes)
- In second order packing, it participates in packaging the 6-200bp nucleosomes into one-1200bp 30nm fiber
Which order of chromatin packing looks like a solenoid?
Second order: 30nm fiber
What is the nuclear matrix? Which order of chromatin hierarchy is this an important aspect of?
- Nuclear matrix is a proteinaceous structure that contains structural and gene regulatory machinery
- Important for the 3rd order: loops which attach to it
At what order of chromatin hierarchy does "supercoiling" become possible?
- 3rd order: loops
- By being bound to the nuclear matrix, the DNA can become supercoiled; effectively circularizes the DNA
Besides conferring supercoiling abilities at the 3rd order of chromatin hierarchy (loops), what other advantages does this level confer?
Genes located in one loop can interact with up- and downstream regulatory sequences (e.g., enhancers) in an organized way
How do "loops" (3rd order) get organized into "minibands" (4th order)?
- Nuclear matrix is in the center with 18 loops spanning out around the chromosome axis
- Looks like flower petals from above
- Proximity of loops on adjacent minibands would permit regulatory sequences in distal loops to interact
Hyper-phosphorylation of chromatin proteins (especially histone H1) permits what?
Hyper-condensation of chromatin into mitotic chromosomes (5th order)
What are the features of the mitotic chromosome?
- Centromere at center
- Two telomeres at either end (repetitive DNA caps; GGGTTA for up to ~15,000 bp)
As telomeres get shorter in somatic cells with each cell division, what term does this lead to?
Cellular Senescence (deterioration with age)
How are telomeres maintained in germ-line cells?
Telomerase maintains telomere length after cell division
Why is it important for telomerase to be inactive in somatic cells?
- If mutated and telomerase is activated it can lead to abnormal maintenance of telomere length
- This leads to cellular immortality and tumor formation
Organization in the interphase nucleus is "dynamic" and "non-random", how is this organization maintained?
Nuclear matrix
What is the perceived purpose of the nuclear matrix?
To organize the genome into domains that regulate gene expression and cell replication
What are the three components of the nuclear matrix?
1. Nuclear envelope / Pore Complex / Lamina
2. Nucleolus
3. Internal Nuclear Matrix
What would be the consequences if the structure of the interphase nucleus was not organized?
- It would be very difficult to maintain normal replication and transcription of chromatin
- This could lead to carcinogenesis and disease
Describe the nuclear envelope component of the nuclear matrix.
- Double membrane
- Encloses perinuclear space
- Continuous with endoplasmic reticulum (ER)
- Divided into segments, limited by nuclear pores
- Outer membrane faces cytoplasm and contains ribosomes
- Inner membrane contains integral proteins that bind nuclear lamina, which attaches to marginal heterochromatin
Describe the nuclear pore complex found in the nuclear matrix?
- Lie between segments of nuclear envelope
- ~10nm diameter holes
- Regulates passage of proteins into, and proteins and RNAs out of, the nucleus
- Pores have three strata arranged as octamer
- Proteins in strata termed nucleoporins which act as docking sites for proteins that contain nuclear localization signals (NLSs)
How big of proteins can get through the nuclear pore complexes of the nuclear envelope?
Proteins smaller than 40kD diffuse through pores; larger proteins are under complex regulation (require nuclear localization signals, NLSs, to get in)
What is the function of nucleoporins?
- Proteins found on the strata of the nuclear pores
- Nucleoporins in cytoplasmic stratum serve as "docking sites" for proteins with nuclear localization signals (NLSs)
What are nuclear localization sequences (NLSs) required for? What common residues do they contain?
- NLSs required for import into the nucleus
- Contain positively charged basic amino acids (lysine, K, and arginine, R)
- Must be on surface of protein
What do proteins containing NLS sequences bind to? What is an example?
- NLS-Receptor Proteins
- Found in cytoplasm
- Ex: importin - heterodimer with two subunits (α and β)
Explain how the NLS receptor protein, Importin, transports proteins into the nucleus?
- Importin-α binds NLS of nuclear protein
- Complex migrates to pore where importin-β binds a nucleoporin in pore complex
- Energy-dependent transport is used to get through the pore (Ran-GTP)
- Once inside, nuclear protein is released and importin is cycled back through pore
How does phosphorylation status affect nuclear import?
- Phosphorylation status either enables or inhibits nuclear import
- Some proteins may have to be de-phosphorylated
- Some have to be released by a cytoplasmic masking protein to be recognized by importin; but masking protein requires phosphorylation to release protein
What is the purpose of Nuclear Export Sequences (NESs)? What amino acid is common in NES sequences?
- Used to export proteins and RNA sequences from the nucleus
- These sequences are leucine-rich
Are nuclear pores specific for transport of proteins or RNA?
- No, protein and RNA transport are not pore-specific
- Same pore can accommodate both
What is the energy source for nuclear import and export?
- RanGTP: GTP binding protein
- High concentration of GTP in nucleus, low conc. in cytosol
- Ran-GTP binds to empty nuclear import receptors and cargo-bound export receptors
- They exit via pore to the outside of the cell where GTP is hydrolyzed to GDP
Why is it important to understand nuclear import/export ("trafficking")?
- Drugs cannot readily penetrate the nucleus
- Nuclear structures are poor targets for therapeutic intervention
- Using knowledge explaining how substances are targeted to nucleus, approaches may be designed that enable development of drugs that can target nuclear structures and genes
Where is the nuclear lamina located?
- Adjacent to inner nuclear envelope
- Marginal heterochromatin are attached on inside of nucleus
- It is broken up by the pore complexes
What are the lamina's proteins called? What is their function?
- Lamins A, B, and C
- Lamins A and C interact with the marginal heterochromatin
- Lamin B binds the inner nuclear membrane via the Lamin-B receptor (LBR), an integral membrane protein
When the nuclear envelope falls apart, what happens to the Lamin proteins?
- Lamin B remains bound to the LBR of the inner nuclear envelope
- This mediates the reformation of the nuclear envelope segments at the start of the next interphase (Lamins A and C attach to Lamin B)
What are "Laminopathies"?
- Mutations of the LMNA gene (gene that codes for Lamin A and Lamin C)
- Lamins A and C are important for binding to heterochromatin to organize the DNA in the nucleus
- Consequences range from several rare cardiac and skeletal muscular dystrophies (wasting away) to a severe form of premature aging (progeria)
What occurs in the Nucleolus?
Ribosomal RNA (rRNA) production
Each nucleolus has how many chromosomes attached to it? How many genes for rRNA are on each chromosome? How many total genes for rRNA are there per nucleus?
10 chromosomes
x ~40 rRNA genes
= 400 rRNA genes/nucleus
Describe the nucleolus's membrane?
Trick, it doesn't have a membrane!
Why does transcription of RNA look like a christmas tree?
- Cell has a high demand for ribosomes
- Transcription of rRNA genes occurs constantly, via RNA polymerase I
- RNA transcripts attached to the gene are shorter at the 5' end and longer at the 3' end
What makes up the majority of the nuclear matrix?
The internal nuclear matrix (~98%)
What structures make up the internal nuclear matrix?
- Chromatin: DNA and histones (H1, H2A, H2B, H3, H4)
- Non-histones (transcription factors, RNA and DNA polymerases, etc)
How are chromosomes situated in the interphase nucleus? How was this proven?
- Rabl configuration: telomeres stick to nuclear envelop and centromere is stuck to opposite side (not mammalian cells, although they do occupy assigned territories)
- Laser micro-radiation showed that, once marked by laser burns, chromosomes occupy the same site of the interphase nucleus
- Also, DNA fluorescent in situ hybridization (FISH) showed that chromosomes occupy discrete territories in nucleus
What is the inactive X chromosome called? Where is it located in the interphase nucleus?
- Barr body
- Always located along the inner nuclear envelope
How does the activity/movement of chromosomes change?
- Chromosomes move around in their territories
- Activity diminishes as cells become heterochromatic and less transcriptionally active during differentiation
- This is reversed when cells become tumorigenic
Are nuclear matrix proteins disease-specific and tissue-specific? What is the significance of this?
- Yes
- Some nuclear matrix proteins in normal cells and cancer cells are different (useful in diagnosis)
What is Tg?
- Generation time
- Time it takes for the cell to go through four cell-cycle phases
What are the four phases of the cell-cycle?
- G1 phase (Gap 1)
- S-phase (synthesis of DNA)
- G2 phase (Gap 2)
- M-phase (mitosis)
Which phases make up interphase?
- G1 phase (Gap 1)
- S-phase (synthesis of DNA)
- G2 phase (Gap 2)
(NOT MITOSIS)
What can a cell do during the G1 phase (Gap 1)?
- Senescence (i.e., Go = nothing happens)
- Differentiation (also Go) = specialize and never divide
- Apoptosis = cell death
- Proliferation = entry into cell cycle (prepare for S-phase)
What happens to a cell during S-phase?
- Synthesis of DNA
- Chromosomes are duplicated from 2N to 4N
- Histones are synthesized to make up the new chromosomes
What happens to a cell during G2 phase (Gap 2)?
- Preparation for mitosis occurs
- Late in G2 the centrosome (not centromere) is duplicated
- Hyper-phosphorylation of histone and non-histone proteins occurs late in G2
What happens to a cell during M-phase?
Mitosis:
- Cells become spherical
- Nuclear membrane disintegrates
- Chromatin condense into chromatids (2/chromosome)
- Chromosomes align on equatorial plate, then chromatids segregate into daughter cells
Which length of the cell cycle is predictable in terms of time? How long does it last? Which part is variable?
- S, G2, and M are relatively constant (12-24 hours)
- G1 can be very brief of very long (as in Go phase)
What are the two types of factors that control the cell cycle?
- External factors: 1' messengers
- Internal factors: 2' messengers
What kind of molecules are 1' messengers, aka external factors?
- Secreted molecules and peptides: growth factors, cytokines, hormones
- Interact with cognate receptors that are in cell membrane or cytoplasm
What are some examples of external factors (1' messengers) that activate the cell cycle?
- FGFs = fibroblast growth factor
- IGFs = insulin growth factor
- Wnts
What are some examples of external factors (1' messengers) that inhibit the cell cycle?
- TGF-β = transforming growth factor
What are the different kinds of internal factors (2' messengers)?
- "Early response" genes: c-myc, fos, jun - rapidly respond to growth signals (~15 min.)
- "Delayed response" genes: cdks and cyclins
When CDKs and cyclins are combined, what do they form? What is this unit called?
- Heterodimers
- Cyclin dependent protein kinases
What do kinases do?
Phosphorylate target proteins
What is the function of the CDK subunit? What are some examples?
- Catalytic subunit
- Phosphorylates the target (substrate)
- CDKs 1, 2, and 4
How does the content of each CDK change throughout the cell cycle?
It remains constant
What is the function of the cyclin subunit? What are some examples?
- Regulatory subunit
- Regulates activity of kinase heterodimer
- Cyclins D, E, A, and B
How does the content of each cyclin change throughout the cell cycle?
It increases (hence name - cycles...cyclin)
What are proto-oncogenes? Examples?
- When it is mutated the protein it encodes becomes pathologically over-active; thus abnormally stimulating the cell-cycle to induce cancer
- Ex: growth factors, growth factor receptors, early response genes, CDKs/cyclins
What residues do kinases phosphorylate?
Residues with OH groups:
- Serine
- Threonine
- Tyrosine
When is regulation by growth factors predominant?
- During early G1 phase
- Diminishes by end of G1 before restriction point (R)
What molecules regulate discrete steps during interphase?
- Intracellular cyclins
- (although cyclins are important throughout)
When do each of the four Cyclins (D, E, A, and B) reach their peak concentrations?
- Cyclin D - increases near beginning of G1, peaks late G1, stays high through mitosis
- Cyclin E - peaks at end of G1 phase
- Cyclin A - increases beginning of S phase; peaks at beginning of G2 phase (where it abruptly goes down)
- Cyclin B - Increases during G2 but peaks at beginning of mitosis
What does p27 have to do with Cyclins D, E, A, and B?
- p27 is a cell-cycle inhibitor (directly inhibits CDK-2 and CDK-4)
- It is high during Go phase
- Drops at beginning of G1 phase
What molecule(s) is G1 phase activated by?
CyclinD/CDK4 heterodimer (first checkpoint)
Describe the activation of Cyclin D which is required for initiating G1 phase.
Growth factors induce increase in Cyclin D:
- Wnt growth factors induce increase of cytoplasmic β-catenin
- This migrates to nucleus to activate transcription of c-myc gene
- This activates cyclin D gene
- Growth factors also induce intracellular ras (GTPase-binding protein) which activates MAP kinases to effect cyclin D production
What happens to Cyclin D in order to initiate G1 phase?
- Cyclin D binds CDK4 to form heterodimer
- CyclinD/CDK4 phosphorylates retinoblastoma (Rb) protein forming P~Rb
- P~Rb releases transcription factor protein E2F
- E2F activates genes encoding Cyclin E and Cyclin A (for 2nd checkpoint)
Retinoblastoma (Rb) protein controls the cell cycle how?
- Rb inhibits cell-cycle
- Phosphorylated Rb (via CyclinD/CDK4) releases inhibition of protein E2F
- E2F is necessary to activate genes for Cyclin E and A
What is the S-phase / 2nd checkpoint activated by?
- CyclinE/CDK2
- Cyclin A/CDK2
How do you make the cell commit to completing the cell cycle?
- Get passed the restriction checkpoint (R)
- E2F production increases amounts of Cyclins E and A
- When E increases it forms the heterodimer CyclinE/CDK2 which breeches the restriction checkpoint
- Now cell is committed to completing cycle
How do "DNA replication complexes" get activated for synthesis?
CyclinA/CDK2 heterodimer phosphorylates DNA replication complexes which are poised at multiple origins of DNA replication, leading to activation of synthesis
How does the cell ensure that all of the DNA is replicated as well as preventing the genome from being replicated more than once?
- CyclinA/CDK2 phosphorylates DNA replication complexes
- This ensures that each daughter cell gets exactly what it is supposed to
How is mitosis (M-phase) activated?
- Dephoshorylation of CyclinB/CDK1 heterodimer
- Mediated by phosphatase: cdc25
- After being dephosphorylated, CyclinB/CDK1 dimer enters nucleus to phosphorylate target proteins
When the CyclinB/CDK1 dimer is dephosphorylated it enters the nucleus to phosphorylate target proteins; what changes does this cause? (beginning of M-phase)
- Nuclear envelope breakdown (NEB)
- Mitotic spindle assembly
- Metaphase arrest
How is anaphase activated?
Mid-metaphase by Anaphase Promoter Complex (APC)
How many cells are there in the human body?
10^13 (ten trillion)
What is the full sequence of events that leads to activation of the cell-cycle?
1. Stimulation of cell proliferation via external growth factors
2. Transcription of Cyclin D
3. Cyclin D protein produced in cytoplasm
4. Binding of Cyclin D to CDK4 = active heterodimer
5. Phosphorylation of Rb protein and release of E2F protein
6. E2F induced transcription of Cyclins E and A
7. Cyclin E and A proteins produced in cytoplasm; bind to CDK2 = active heterodimers --> DNA synthesis
7a. p53 mediated transcription of p21
7b. p21 inhibition of CyclinE/CDK2 (prevent cell from going through process again)
8. Cdc25 induced dephosphorylation of CyclinB/CDK1 heterodimer
- CyclinB/CDK1 phosphorylation of targets, inducing (i) nuclear envelope breakdown (NEB), (ii) spindle assembly, (iii) metaphase arrest
Very generally, what is cancer?
- Genetic disease
- Loss of cellular differentiation
- Increased proliferation and invasiveness of cells
- Changes in chromosomes: re-arrangement, loss, gain
Very generally, what causes cancer?
- Inherited mutations and/or environmental "insults" to DNA during aging
- Cells evolve towards malignancy with age (accumulation of mutations)
What are the five gene categories that are mutated in cancer?
1. Proto-oncogenes
2. Tumor Suppressor Genes
3. Genes that regulate Apoptosis
4. Genes that Induce Cellular Immortality: Telomerase
5. Genes that Repair DNA
When proto-oncogenes are mutated, what are they called?
Oncogenes
What are the consequences of proto-oncogenes being mutated?
- Genes are either over-expressed or have intensified activity
- Either way, leads to UP-regulation
What are some examples of oncogenes?
- ras (GTP binding protein)
- src (tyrosine kinase cell surface receptor)
- cyclin D
- EGFR (epidermal growth factor receptor)
- Myc (transcription factor)
What does a normal tumor suppressor gene do? What happens when it is mutated?
- Inhibit cell-cycle normally
- When mutated they no long work
What are some examples of tumor suppressor genes?
- p21: inhibits CDKs 2 and 4
- p53: activates p21
- Rb: binds E2F (to prevent it from inducing cyclins E and A)
- BRCA: usually repairs broken DNA (common in breast cancer)
Which tumor suppressor gene is often mutated in breast cancer?
BRCA
Why is apoptosis essential?
- Must be a balance between cell renewal and cell death
- Diseased cells must be removed
- Stimulation of apoptosis causes tumors to regress
What is the sequence of apoptosis?
- Macrophages release TNF (tumor necrosis factor)
- TNF binds TNFR cell membrane receptor
- Balance between pro-apoptotic and anti-apoptotic factors is established
- Pro-apoptotic signals induce 'leakiness' of outer membrane of mitochondria
- Cytochrome-C escapes mitos into cytoplasm where it activates Caspase
- Caspase is a protease that destroys chromatin
- Blebs appear on cell surface and cell defoliates = Apoptosis
What gene normally inhibits apoptosis? When it is mutated, how can this lead to cancer?
- bcl-2
- When mutated it is abnormally active
- Too much inhibition of apoptosis occurs
- Leads to tumor formation (ex: in B cell lymphoma)
What gene induces cellular mortality?
Telomerase
What is the "telomere hypothesis"?
When telomere reaches a certain length it sends a signal to cell to become senescent
What kind of cells have the ability to restore the telomere length after every cell division?
- Embryonic stem cells
- Adult sperm and egg cells (germ cells)
What happens if telomerase is mutated in somatic cells?
- It may become active
- This would render them immortal
- Usually they die after so many cell cycles
- Can result in carcinogenesis
What consequences can occur if genes that repair DNA are mutated?
- These proteins can be inactivated
- Clones of abnormal cells arise
In what cancer has it been seen that mutations to genes that repair DNA was responsible?
Colorectal cancer
What are three specific approaches to cancer treatment (to avoid wide-ranging side effects of chemo and radiation)?
- Target metastasis
- Target angiogenesis
- Target specific, sick molecules
How can targeting metastasis help treat cancer specifically?
- Specific protease-inhibitor molecules inhibit a protease enzyme's active site
- In cancer, proteases such as metalloproteinases facilitate metastatic spread by enabling "primary" cancer cells to eat through extracellular matrix
- "TIMPS" = tissue inhibitors of metalloproteinases are being studied in clinical trials
How can targeting angiogenesis help treat cancer specifically?
- Angiogenesis is process of new blood vessels developing
- Tumors must be within 200μm of blood supply
- If angiogenesis is prevented, tumors can be starved to death
How can targeting specific, sick molecules help treat cancer specifically?
- Small inhibitor RNAs (siRNAs) target and destroy pathogenic RNAs and viruses via RNA inhibition (RNAi)
- This could be used to target molecules that activate the cell-cycle in tumor cells but not in normal cells
How does the use of small inhibitory RNAs (siRNAs) work to target specific, sick molecules, in theory?
- siRNAs complementary to pathogenic RNA are introduced to cell
- siRNAs bind to enzyme complex termed RISC
- RISC/siRNA complex base-pairs with complementary pathogenic RNA
- RISC cleaves the pathogenic RNA, eliminating it
What are some problems with siRNAs as a specific cancer treatment?
- Must prevent "off-target effects" on normal molecules (caused by mis-matches)
- Must develop methods to deliver siRNAs to patients' tumors in vivo
What occurs during the Go phase of the cell cycle?
- Terminally differentiated cells withdraw from cell cycle indefinitely
- A cell returning from Go enters at early G1 phase
What occurs during the G1 phase of the cell cycle?
- RNA and protein synthesis
- No DNA synthesis
- Restriction point at end: cell that passes this point is committed to pass into S phase
What occurs during the S phase of the cell cycle?
- DNA synthesis doubles amount of DNA in cell
- RNA and protein also synthesized
What occurs during the G2 phase of the cell cycle?
- No DNA synthesis
- RNA and protein synthesis continue
What occurs during the M phase of the cell cycle?
- Mitosis (nuclear division)
- Cytokinesis (cell division)
- Yields two daughter cells
What are the four basic properties of DNA replication?
1. Semiconservative
2. Synthesized in 5'-->3' direction
3. A 3'-OH primer and a template are required
4. Semi-discontinuous
What does semiconservative DNA synthesis mean?
- Produce two molecules with both old and new DNA (strand of each)
- Each strand acts as a template for the synthesis of a new DNA molecule
The 3'-OH primer required for DNA replication does what to initiate?
3'OH performs a nucleophilic attack on α phosphate of incoming dNTP
What does it mean for DNA replication to be semi-discontinuous?
- In order to replicate in 5'-->3' direction, there will be a leading strand and a lagging strand
- Only short piece of template is exposed on lagging strand
- This generates Okazaki fragments (each is separated by a gap and piece of RNA)
Why is a RNA primer used to begin DNA replication on the lagging strand?
- DNA synthesis requires a free 3'-OH primer
- On lagging strand must substitute for RNA primer
What are the four basic steps of DNA replication?
1. Separation of two complementary strands at origin or replication
2. Formation of replication fork (primers and Okazaki fragments)
3. Chain elongation
4. Removal of primers
How many origins of replication are on prokaryote DNA? Eukaryotes?
- Prokaryotes: one origin per piece of circular DNA
- Eukaryotes: many origins in order to be most efficient
What are the three catalytic activities of E. coli DNA polymerase I?
- 5'-->3' polymerase
- 5'-->3' exonuclease
- 3'-->5' exonuclease
What is the purpose of the 5'-->3' and 3'-->5' exonucleases of E. coli DNA polymerase I?
- Cleave phosphodiester bonds
- Proofreading activity: removes incorrectly base paired nucleotides
What is the main difference between the enzymatic function of DNA polymerase I and III?
- Pol I has 5'-->3' and 3'-->5' exonucleases
- Pol III has no exonuclease activity (no proofreading)
What is the proofreading mechanism of DNA Pol I?
- Mispaired 3'OH end of growing strand blocks further elongation
- DNA polymerase slides back to position the mispaired base in the 3'-->5' exonuclease active site
- Mispaired nucleotide is removed
- DNA pol slides forwards and resumes polymerization activity
Due to the functions of DNA poly I and III, what role does each primarily play?
- DNA Pol III = synthesizes DNA
- DNA Pol I = erases primer and fill gaps; repair
What does the term processivity mean?
- Number of catalytic events before dissociation from template
- e.g., how many nucleotides are added by DNA pol before it falls off?
Does DNA Pol III or I have higher processivity?
- DNA pol III is very processive: synthesizes >500,000 nucleotides before dissociating
- DNA pol I is not very processive: synthesizes only 3-200 nucleotides (more involved in repair)
What is the role of DNA helicase?
Unwinds the double helix of DNA
What is the role of single-stranded DNA-binding proteins (SSB)?
- Stabilizes single-stranded regions
- Prevents premature re-ligation
What is the role of Primase?
Synthesizes RNA primer for DNA replication on lagging strand
What is the role of DNA Ligase?
Connects DNA fragments after RNA primers are replaced with DNA nucleotides by DNA Pol I
What enzyme is responsible for removing the RNA primers from DNA (during replication)?
DNA Polymerase I
Describe the steps of DNA replication on the lagging strand.
1. RNA primer is elongated by DNA polymerase III until another stretch of RNA is encountered
2. RNA primer is excised by DNA polymerase I, one ribonucleotide at a time
3. Gap is filled by DNA polymerase I
4. Remaining nick is sealed by DNA ligase
Why does the telomere get shortened with successive replications?
- Primer dependence and 5' to 3' directionality of DNA polymerase results in incomplete daughter strand synthesis at telomere
How does telomerase lengthen the telomere?
- Telomerase is a reverse transcriptase that carries its own RNA template
- RNA component of telomerase is complementary to telomere DNA repeats
At which atom does the purine base attach to the sugar?
N9 (nitrogen on 5-membered ring)
At which atom does the pyrimidine base attach to the sugar?
N1
At what carbon does the glycosidic linkage occur on the pentose sugar?
C1'
At what carbon does the phosphate linkage occur on the pentose sugar?
C5'
What is the name of the purine base "adenine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = adenosine
- Ribonucleotide = adenylic acid or AMP
What is the name of the purine base "guanine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = guanosine
- Ribonucleotide = guanylic acid or GMP
What is the name of the purine base "hypoxanthine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = inosine
- Ribonucleotide = inosinic acid or IMP
What is the name of the purine base "xanthine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = xanthosine
- Ribonucleotide = xanthylic acid or XMP
What is the name of the pyrimidine base "uracil" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = uridine
- Ribonucleotide = uridylic acid or UMP
What is the name of the pyrimidine base "cytosine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = cytidine
- Ribonucleotide = cytidylic acid or CMP
What is the name of the pyrimidine base "thymine" when it is in a ribonucleoside (w/o phosphate)? ribonucleotide (w/ phosphate)?
- Ribonucleoside = thymidine
- Ribonucleotide = thymidylic acid or TMP
What is the starting molecule for purine de novo synthesis?
PRPP (phospho-ribosyl-pyrophosphate)
Does purine de novo synthesis start with a sugar and phosphate or end with it?
Starts with it as base
Does pyrimidine de novo synthesis start with a sugar and phosphate or end with it?
Adds it at end
Which molecules are necessary to build the purine rings and in what order?
1. PRPP (ribose and sugar)
2. Glutamine (amino group)
3. Glycine (entire residue; attaches at carbonyl)
4. N-Formyl tetrahydrofolate (aldehyde)
5. Glutamine (amino group)
6. CO2
7. Aspartate (amino group)
8. N-Formyl tetrahydrofolate (aldehyde)
What are the steps of the synthesis of de novo purine nucleotide synthesis, starting with PRPP?
1. Displacement of pyrophosphate by amino group of glutamine
2. Addition of glycine
3. Formylation by folic acid derivative N-Formyl tetrahydrofolate
4. Transfer of another nitrogen from glutamine
5. 5-member ring closure by dehydration
6. Carboxylation
7. Addition of aspartate
8. Elimination of fumarate (leaves amino group of aspartate)
9. Formylation by another N-Formyl tetrahydrofolate
10. 6-member ring closure by dehydration: forms IMP
Once the purine nucleotide IMP is created, how is it differentiated into the more common AMP?
1. Aspartate is added to IMP; GTP hydrolyzed; form adenylosuccinate
2. Fumarate is removed leaving behind amino group from aspartate; AMP is formed
Once the purine nucleotide IMP is created, how is it differentiated into the more common GMP?
1. NAD+ accepts electrons resulting in a carbonyl being formed (produces XMP)
2. Glutamine donates an amino group; ATP is hydrolyzed to form GMP
How is the starting substrate PRPP produced?
- PRPP synthetase uses ATP to convert D-ribose-5-P to PRPP
- Essentially adds pyrophosphate group to C1' of ribose sugar to activate it for ATase activity
Why does PRPP have the pyrophosphate group on it?
- This activates the ribose for addition of amino group from glutamine
- Occurs via ATase enzyme
Which step is the rate-limiting step of the de novo synthesis of purines?
- First step w/ ATase
- Substituting amino group from glutamine for pyrophosphate group on PRPP
Which molecule is the first product that contains the completed purine ring?
IMP (inosinic acid)
How are the monophosphate nucleosides (GMP and AMP, etc) converted to diphosphates and triphosphates?
- Base-specific monophosphate kinases convert AMP and GMP to ADP and GDP respectively; phosphate donated by ATP (leaving ADP for both)
- NDP converted to NTP via another non-base specific kinase and again ATP donates phosphate
Adenylate kinase maintains what equilibrium among ATP, ADP, and AMP?
- 100x ATP
- 10x ADP
- 1x AMP
What regulation controls the purine de novo pathway?
- End product regulation (GMP/GDP/GTP and AMP/ADP/ATP) inhibits PRPP synthetase, ATase and each branch point (IMP --> XMP or adenylosuccinate)
- Stimulation of branch point synthesis by end-product of opposite branch (i.e. GTP stimulates IMP conversion to adneylosuccinate, which forms AMP; ATP stimulates ITP conversion to XMP, which forms GMP)
What steps take place in the degradation of AMP?
- Dephosphorylation and deamination to form inosine (either one can go first)
- Phosphorolysis (removal of sugar) to form Hypoxanthine
- Oxidation to form Xanthine
- Further oxidation to form Uric Acid
In the degradation of AMP, why would it be important to deaminate first (producing IMP)?
- In vigorously exercising skeletal muscle more ATP is needed
- Production of phosphorylated catabolite IMP facilitates resynthesis of ATP
In the degradation of AMP, why would it be important to dephosphorylate first (producing adenosine)?
- In ischemic or anoxic heart (heart attack) adenosine is needed
- Adenosine is a diffusible compound that is a potent coronary vasodilator, which facilitates oxygen delivery to damaged tissues
What steps take place in the degradation of GMP?
- Dephosphorylation to guanosine
- Phosphorolysis (removal of sugar) to form guanine
- Deaminatioin to form Xanthine
- Oxidation to form Uric Acid
What enzymes are responsible for dephosphorylation during the degradation of purine nucleotides?
5'-nucleotidases (5'-NT)
What enzyme is responsible for phosphorolysis during the degradation of purine nucleotides?
- Purine nucleoside phosphorylase (PNP)
- Removes the pentose sugar
What enzymes are responsible for deamination during the degradation of purine nucleotides?
- AMP deaminase
- Adenosine deaminase (ADA)
- Guanase
What enzyme is responsible for oxidation of hypoxanthine and xanthine during the degradation of purine nucleotides? What does this produce?
- Xanthine dehydrogenase (XDH)
- Converts hypoxanthine to xanthine
- Xanthine to uric acid
What does xanthine dehydrogenase (XDH) require to oxidize the end products of purine degradation to uric acid?
NAD+ = electron acceptor
If there is a decrease in intracellular oxygen a protease is activated and converts xanthine dehydrogenase (XDH) to what? What are the implications?
Xanthine Oxidase (XO)
- O2 acts as electron acceptor and generates H2O2 (free radicals-->damage)
What is the term for overproduction or underexcretion of uric acid?
Hyperuricemia
Which is the least soluble purine base?
Uric acid
Why is it bad if there is overproduction or underexcretion of uric acid?
- Uric acid is the least soluble purine base and exists at, or near, saturating levels
- When it exceeds saturation limits it leads to deposits in the blood, joints, tissues, and urine = Gout
What disorder is characterized by hyperuricemia (high levels of uric acid) due to inherited metabolic abnormalities?
Primary gout
What disorder is characterized by excessive uric acid production due to a coexisting acquired condition?
Secondary gout
What are the most likely causes of primary gout?
- Excess uric acid production (overexpression of PRPP synthetase and a partial or complete deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT)
- Defect in excretion (80%) - defect in family of renal urate transport proteins
What are some examples of causes secondary gout?
Individuals who overproduce or under excrete uric acid in association with drug intake or unusual dietary habits
What is the number one treatment of both kinds of gout? What does it do?
- Allopurinol
- Inhibits rate of uric acid production
- Analog of hypoxanthine
- Converted to alloxanthine by xanthine dehydrogenase (XDH) which in turn inactivates the enzyme
What is "Xanthinuria"?
- Inherited disorder
- Second most insoluble purine base (after uric acid)
- Precipitates when overproduced
- Most commonly caused by deficiency of xanthine dehydrogenase (XDH)
What causes the "bubble boy syndrome"?
- AKA: SCID (severe combined immunodeficiency disease)
- Adenosine deaminase deficiency
- Associated with severe combined immunodeficiency involving T-cell and B-cell dysfunction
- Likely due to accumulation of dATP in lymphocytes
- dATP is an effective inhibitor of ribonucleotide reductase and consequently inhibits DNA synthesis and cell division
What happens if there is a deficiency of purine nucleoside phosphorylase (PNP)?
- Impairment of T-cell function
- Individuals underexcrete uric acid and over excrete PNP substrates (inosine and guanosine)
- Accumulation of dGTP in T-cells inhibits ribonucleotide reductase and consequently inhibits DNA synthesis and cell division
Why is the salvage pathway of purine nucleotide synthesis preferred?
- De novo synthesis is metabolically expensive
- At least 5 ATP molecules are consumed per purine formed
What is the use of PRPP in the salvage pathway of purine synthesis?
- PRPP is used to attach ribose-5-P directly to a purine base
- Adenine + PRPP --> AMP + PPi (via adenine phosphoribosyltransferase, APRT)
- Hypoxanthine or Guanine or Xanthine + PRPP --> IMP or GMP or XMP + PPi (via HPRT)
What happens if there is an inherited deficiency of APRT (adenosine phosphoribosyltransferase)?
- Inability to salvage adenine (to form AMP)
- Instead adenine is catabolized to 2,8-DHA which is very insoluble and cant lead to urinary stones
On average, about how much of catabolized purine nucleotides are excreted as uric acid, while how much is salvaged?
- 10% excreted as uric acid
- 90% salvaged
What is the major method of purine synthesis?
Salvage pathway
What inherited disorder of purine salvage synthesis affects the production of IMP, GMP, and XMP?
- Lesch-Nyhan Syndrome
- HPRT is lacking or is present at very low levels (<1% of normal)
- HPRT locus is on X chromosome making this disease only affect males
What are the symptoms of Lesch-Nyhan Syndrome (lack of HPRT enzyme for purine salvage)?
- Aggressive behavior
- Self-mutilation
- Mental retardation
- High levels of uric acid which leads to gout
- Increased rates of de novo synthesis (to compensate)
What are the treatments for Lesch-Nyhan Syndrome (lack of HPRT enzyme for purine salvage)?
- Allopurinol is administered to relieve complications of gout
- No known treatment for neurological problems however
What residues/molecules contribute to the make-up of a pyrimidine residue?
- Glutamine (amino group)
- Aspartate (C-C=C-N)
- CO2
What are the characteristics of the enzyme that is responsible for the first three steps in the de novo synthesis of a pyrimidine base?
- Carbamoyl-phosphate synthetase (CPS II) (2) = uses glutamine as source of amino group and adds to phosphate
- Aspartate transcarbamoylase (2) = combines first product with aspartic acid
- Dihydro-orotase dehydrogenase (DHODH) (3) = removes water from second product
- Abbreviated "CAD"
What step is rate-limiting in the de novo pyrimidine synthesis?
- First step: CPS II
- Adding amino group from glutamine to phosphate from ATP
What is the first pyrimidine synthesized by the de novo synthesis?
UMP (uridylic acid)
How is the pyrimidine nucleotide CTP generated?
- UMP is phosphorylated by a specific and non-specific kinase to UTP
- UTP is converted to CTP by enzyme CTP synthetase; glutamine is source of amino group; ATP is used
What is the process of pyrimidine degradation and excretion?
- Pyrimidine is dephosphorylated to nucleoside derivative
- If Cytidine, must be deanminated to uridine
- Undergoes phosphorolysis (removal of sugar) to generate Uracil (or thymine if starting with dTMP)
How does pyrimidine degradation differ from purine degradation?
- Pyrimidines use reduction reactions to break down the ring structure into smaller components (uracil and thymine)
- Purines use oxidation reactions
What happens to uracil and thymine in pyrimidine nucleotide degradation?
- Additional steps are perform to degrade uracil and thymine to amino acids
- They can then be further degraded by amino acid pathways
Why is the salvage pathway much more common than the de novo pathway for pyrimidine synthesis?
- De novo costs 5 moles of ATP / mol UMP
- Salvage costs 1 mole of ATP / mol UMP
What is the most salvageable pyrimidine products?
Nucleosides; these can be used to generate nucleotides
What are four inherited disorders of pyrimidine nucleotide metabolism?
1. Orotic aciduria-UMP synthase deficiency - excessive orotic acid in urine; often megaloblastic anemia
2. Pyrimidine 5'-nucleotidase deficiency - increased erythrocyte pyrimidine ribonucleotides; leads to anemia
3. Dihydropyrimidine dehydrogenase deficiency - elevated uracil and thymine levels; convulsive disorder characterized by microencephaly
4. Dihydropyrimidinase deficiency - elevated dihydropyrimidines; variable neuro symptoms
What is the treatment for Orotic aciduria-UMP synthase deficiency (which leads to excessive amounts of orotic acid)?
- Lethal unless treated by pyrimidine replacement, i.e. uridine
- Uracil is an ineffective therapy as it can't be converted into uridine (which can be salvaged into other pyrimidine nucleotides)
How are ribonucleotides converted to deoxyribonucleotides?
- Reduced by ribonucleotide reductase (RR)
- Substrate is any ribonucleoside diphosphate (NDP)
What provides the reducing power for ribonucleotide reductase?
- NADPH + H+
(note: not NADH)
Which molecule can inhibit ribonucleotide reductase?
dATP - this ends up preventing DNA synthesis and cell division
How can thymidine be synthesized?
- First UDP must be converted to dUDP by ribonucleotide reductase
- dUDP is converted to dUMP
- dUMP is converted to dTMP by thymidylate synthase
What are four inherited disorders of pyrimidine nucleotide metabolism?
1. Orotic aciduria-UMP synthase deficiency - excessive orotic acid in urine; often megaloblastic anemia
2. Pyrimidine 5'-nucleotidase deficiency - increased erythrocyte pyrimidine ribonucleotides; leads to anemia
3. Dihydropyrimidine dehydrogenase deficiency - elevated uracil and thymine levels; convulsive disorder characterized by microencephaly
4. Dihydropyrimidinase deficiency - elevated dihydropyrimidines; variable neuro symptoms
What is the treatment for Orotic aciduria-UMP synthase deficiency (which leads to excessive amounts of orotic acid)?
- Lethal unless treated by pyrimidine replacement, i.e. uridine
- Uracil is an ineffective therapy as it can't be converted into uridine (which can be salvaged into other pyrimidine nucleotides)
How are ribonucleotides converted to deoxyribonucleotides?
- Reduced by ribonucleotide reductase (RR)
- Substrate is any ribonucleoside diphosphate (NDP)
What provides the reducing power for ribonucleotide reductase?
- NADPH + H+
(note: not NADH)
Which molecule can inhibit ribonucleotide reductase?
dATP - this ends up preventing DNA synthesis and cell division
How can thymidine be synthesized?
- First UDP must be converted to dUDP by ribonucleotide reductase
- dUDP is converted to dUMP
- dUMP is converted to dTMP by thymidylate synthase
What are four inherited disorders of pyrimidine nucleotide metabolism?
1. Orotic aciduria-UMP synthase deficiency - excessive orotic acid in urine; often megaloblastic anemia
2. Pyrimidine 5'-nucleotidase deficiency - increased erythrocyte pyrimidine ribonucleotides; leads to anemia
3. Dihydropyrimidine dehydrogenase deficiency - elevated uracil and thymine levels; convulsive disorder characterized by microencephaly
4. Dihydropyrimidinase deficiency - elevated dihydropyrimidines; variable neuro symptoms
What is the treatment for Orotic aciduria-UMP synthase deficiency (which leads to excessive amounts of orotic acid)?
- Lethal unless treated by pyrimidine replacement, i.e. uridine
- Uracil is an ineffective therapy as it can't be converted into uridine (which can be salvaged into other pyrimidine nucleotides)
How are ribonucleotides converted to deoxyribonucleotides?
- Reduced by ribonucleotide reductase (RR)
- Substrate is any ribonucleoside diphosphate (NDP)
What provides the reducing power for ribonucleotide reductase?
- NADPH + H+
(note: not NADH)
Which molecule can inhibit ribonucleotide reductase?
dATP - this ends up preventing DNA synthesis and cell division
How can thymidine be synthesized?
- First UDP must be converted to dUDP by ribonucleotide reductase
- dUDP is converted to dUMP
- dUMP is converted to dTMP by thymidylate synthase
What is required for RNA transcription?
- DNA template
- Ribonucleoside 5'-triphosphates
- NO primer
What are the three major types of RNA and what percent do they make up?
- mRNA (5%)
- tRNA (~15%)
- rRNA (~80%)
- snRNA (small nuclear RNA)
In prokaryotes how many RNA polymerases are needed to synthesize mRNA, tRNA, and rRNA?
Only 1
What subunit of RNA polymerase allows it to recognize promoter regions?
σ subunit
What kind of enzyme is RNA polymerase?
Zn2+ metalloenzyme
What kind of proofreading mechanism does RNA polymerase have?
No 3'-->5' proofreading activity
What is the consequence of not having 3'-->5' proofreading activity on RNA polymerase?
Error rate is much higher than during DNA synthesis (10^4 - 10^5)
What are the three components of the "Operon"?
- Promoter = site for binding RNA polymerase
- Operator = binding sites for repressor of activator (not in eukaryotes)
- Structural gene(s)
What is the function of the lac operon?
- Regulating the production of enzymes that allow for the utilization of lactose as a carbon source
Can multiple genes be on one operon in eukaryotes?
No, there is a new operon for each gene; unlike in prokaryotes where multiple genes can be controlled by a single operon
What are the steps of RNA transcription?
- Initiation
- Elongation
- Termination
(- Protein-Independent Termination)
Which step is paramount in regulating whether a gene is made into a protein?
Initiation of Transcription
What are the two regions of the promoter in prokaryotes that are important for the polymerase to recognize?
-35 region
-10 region
When does the "closed complex" form?
- When the RNA polymerase σ unit recognizes -35 region of DNA
After the RNA polymerase forms a closed complex at -35 region, what happens?
It migrates to -10 region; DNA is unwound and forms an open complex
What is the key initiation event of transcription?
Transition from closed complex to open complex; this commits transcription to happening
As RNA polymerase proceeds past -10 region, what happens to the σ subunit that was important for recognizing the regions?
It's released
What is the purpose of having different σ subunits on RNA polymerase?
They recognize different promoters
How does the σ subunit affect RNA polymerase?
Increases specificity but decreases affinity
What is the energy source for transcription elongation?
- Cleavage of phosphate bond between ribonucleotide
- Subsequent hydrolysis of pyrophosphate to inorganic phosphate
Can more than one RNA polymerase be involved in transcription of one gene at the same time?
- Yes
- Multiple RNA polymerase complexes load onto the promoter region in sequential fashion
How processive is RNA polymerase?
Highly; it can synthesize thousands of nucleotides before falling off
When does a transcription bubble form?
- During elongation
- Negative superhelicity in double-stranded DNA facilitates melting of two strands of DNA
Ahead of the advancing transcription complex, how is the DNA supercoiled?
Positively supercoiled
Behind of the advancing transcription complex, how is the DNA supercoiled?
Negatively supercoiled
Do the mechanisms of transcription termination involve proteins?
Yes and No; there is protein-dependent and protein-independent mechanisms
What structure do protein-dependent and protein-independent transcription termination mechanisms rely upon?
Hairpin loop
What interaction does the protein-dependent transcription termination depend upon?
Interaction of terminator protein Rho (ρ) with RNA polymerase elongation complex as it pauses at a hairpin loop
Describe the Rho (ρ) protein that is important for protein-dependent transcription termination.
- Binds to single-stranded RNA
- ATP-dependent (ATPase activity allows it to move towards complex)
- RNA-DNA helicase: disrupts RNA-DNA hybrid, leading to disassembly of elongation complex
How is the Rho (ρ) protein able to disrupt transcription and ultimately terminate it?
Transcription complex stalls at hairpin loop which signals termination
How does protein-independent transcription termination occur?
- Hairpin loop (GC rich) causes transcription complex to pause
- Run of adenylates in template bound to run of uridylates in transcript are weakly bound
- Stalling causes RNA-DNA hybrid of A-U pairs to destabilize and disengage the complex
- dA-rU is weak compared to dA-dT (therefore DNA duplex reforms, removing RNA transcript)
- RNA polymerase has less affinity for double-stranded DNA than single-stranded RNA therefore it dissociates
How does eukaryotic transcription termination occur?
- More similar to protein independent mechanism (GC hairpin stalls transcription complex; AU region of bonding between transcript and template falls apart)
How many RNA polymerases are there? What do they synthesize?
- I = 18S, 5.8S, and 28S rRNA
- II = mRNA precursors and snRNA
- tRNA and 5S RNA
How can you distinguish between the three eukaryotic RNA polymerases besides based on their cellular transcripts?
Sensitivity to α-Amanitin (mushroom toxin)
- I = insensitive
- II = strongly inhibited
- III = inhibited by strong concentrations
What are some drug inhibitors of transcription?
- Rifampicin (inhibits bacterial RNAPs)
- α-amanatin (inhibits euk RNAP II)
- Actinomycin D (intercalates between two G-C base pairs in DNA)
- Daunorubicin (intercalates between base pairs)
- Cordycepin (chain terminator that lacks 3'OH)
When are transcription and translation separated in space and time?
In eukaryotes (NOT in prokaryotes)
How are transcription and translation separated in time and space in eukaryotes?
- Nuclear membrane
- Transcription inside nucleus
- Translation outside of nucleus
What is a benefit of eukaryotes separating transcription and translation physically?
Allows for transcriptional regulation because translation does not begin immediately
What are some methods of transcriptional processing in eukaryotes?
- Capping to 5' end
- Poly-adenylation (at 3' end)
- Intron splicing by splicosomes
What is the purpose of adding a cap to the 5' end of transcripts?
- Enhances stability: protection from nucleases / degradation
- Enhances translation efficiency
What are examples of 5' capping of transcripts?
- 7-methyl-guanosine (added by guanylyltransferase)
- Methyl groups added to riboses (methyl transferases using S-Adenosyl Methionine, SAM, as donor)
What kind of RNA can be capped?
Only mRNA
What is the purpose of adding the poly-A tail to transcripts?
- Increases mRNA stability
- Helps in translation
What is the mechanism of mRNA splicing in eukaryotes?
1. 2'OH attacks 5' splice
2. Newly formed 3'OH attacks 3' splice and exons joined
3. Lariat formed of intron with 2'-5' bond formed (one end is bonded to the middle of the chain, like the letter 'd')
What molecules are involved in the precise recognition of splice sites and splicing mechanism?
snRNAs (small nuclear RNAs)
What plays a key role in alignment and catalysis of splicing introns?
RNA molecules (snRNAs)
Does splicing require energy?
Yes, ATP is involved in unwinding RNA duplex intermediates and catalyzing release of RNPs mRNA precursors and products
What is the purpose of introns?
Provides another method of generating protein diversity
Approximately what percent of human genetic diseases are possibly due to splicing defects?
~15%
What is MARFAN Syndrome (MRS)?
- Defect in fibrillin component of connective tissue
- Splicing defect of fibrillin mRNA
- Leads to defects in microfibrils and elastic fibers surrounding aorta
What kind of symptoms does someone with MARFAN Syndrome (MRS) show?
- Chest pain
- Dyspnea (difficult/labored breathing)
- MRI shows aorta weakness (and blood seepage) - aortic aneurysm (weak and bulging area in aorta)
- Limbs are disproportionally long and thin, muscle development can be poor, spine can be curved abnormally
- Dural ecstasia - brain and spinal cord surrounded by fluid contained by dura mater
What is a possible therapy for MARFAN Syndrome (MRS)?
- Blocking TGF-β with neutralizing antibodies
- Leads to normalized lung development
Which enzyme hydrolyzes lactose to galactose and glucose?
β-galactosidase
How are functionally related genes in prokaryotes organized?
Into Operons
At what level is regulation of operons in prokaryotes primarily exerted?
Transcription
What is the function of "inducers"?
Inducers bind to the repressor to weaken affinity for the operator (thus up-regulating transcription)
What are the inducers for the lac operon?
- Allolactose
- IPTG
In what configuration does the Lac repressor bind to the operator?
Tetramer in a tethered dimer configuration
What characterizes the binding site of the Lac repressor?
Palindromic sequence (symmetrical)
What is necessary of glucose concentration before the Lac Operon will be activated?
Glucose needs to be used up
What happens when glucose is consumed in E.coli?
- cAMP levels increase
- cAMP binds to CAP (catabolite activator protein)
- cAMP-CAP binds to promoter, binding facilitates RNAP binding to promoter
What is the "glucose effect"?
- When glucose levels are high, the lac operon is not activated
- When glucose levels are low, the lac operon is activated so as to create β-galactosidase so that lactose can be used as an energy source
Under what circumstances is the Lac Operon ON?
- Low glucose
- High lactose
Why is the Lac operon off when both glucose and lactose levels are high?
- Glucose high, so cAMP levels are low
- cAMP does not bind to CAP
- cAMP-CAP complex necessary to bind to promoter
If lactose levels are low, why is the Lac Operon OFF?
- Lac Repressor is bound
What are four types of molecules that are involved at the promoter for regulation of eukaryotic gene transcription?
- Basal Transcription Machinery
- Regulating proteins
- Coactivators
- Insulators
What is the function of basal transcription machinery / basal factors?
- In response to injunctions from activators, these factors position RNA polymerase at start of protein-coding region
- Composed of RNA Polymerase and general transcription factors (including TATA binding protein)
What binds to the core promoter for transcription?
Basal Factors / Basal Transcription Machinery
What are the two types of regulating proteins for gene regulation in eukaryotes?
- Activators and Repressors
What do activators and repressors bind to?
Enhancer sequences or silence sequences (a distance from the core promoter)
What links the basal transcriptional machinery to the activators?
Coactivators (mediators)
What is the function of insulators?
Define regulatory regions of DNA
What is the order of assembly of the basal transcription factors?
1. TATA-binding protein (TBP) binds first
2. Other basal factors bind at promoter region
3. RNA Polymerase II is recruited to the promoter in a non-phosphorylated state by the basal factors
4. Following assembly into the transcription complex, the C-terminal domain (CTD) of RNA polymerase II is phosphorylated and begins transcriptional elongation
What is the rate limiting step of transcriptional initiation?
Formation of PIC (pre-initation complex)
What are the four structural motifs that are found in DNA-binding proteins in eukaryotes?
1. Helix-turn-helix proteins
2. Zinc finger proteins
3. Leucine zipper proteins
4. Helix-loop-helix proteins
Describe the structural features of a helix-turn-helix protein.
- Two α-helices separated by a β-turn "Recognition helix" fitting in major groove of DNA
Describe the structural features of a Zinc finger protein.
- Contains zinc bound to Cys and His side chains
- Mediates DNA binding and can also serve as a protein:protein interface
Describe the structural features of a leucine zipper protein.
- Two α-helices, one with basic residues for DNA binding, one with regularly space Leu for dimerization
- Protein:protein interaction that allows dimerization of transcription factors that contain basic DNA interaction regions (doesn't actually bind DNA)
Describe the structural features of a helix-loop-helix protein.
- DNA-binding α-helix and two dimerization helices separated by a nonhelical loop
- One helix is smaller, which because of flexibility of loop allows dimerization by folding against another helix
- Larger helix contains DNA-binding regions
Which structural motif found in DNA binding proteins in eukaryotes mediates DNA binding of regulatory proteins?
Helix-Turn-Helix Motif
What are the components required for Protein synthesis?
- Amino acids
- tRNAs
- mRNAs (template)
- aa-tRNA synthetases
- Ribosomes
- Initiation, elongation, and termination factors
- ATP and GTP (energy sources)
What makes up a ribosome?
- rRNA
- protein
What are the rRNA and subunits of a prokaryotic ribosome?
- rRNA: 23S, 5S = 50S subunit
- rRNA: 16S = 30S subunit
- total: 70S subunit
What are the rRNA and subunits of a eukaryotic ribosome?
- rRNA: 28S, 5.8S, 5S = 60S subunit
- rRNA: 18S = 50S subunit
- total: 80S subunit
What are the properties of the genetic code that gives triplets (called codons)?
- Specific (codon always codes for same aa)
- Universal (although mito is slightly different)
- Redundant (degenerate, a given aa may have more than one codon)
- Non-overlapping and comma-less (read from fixed starting point as a continuous sequence)
What are the termination codons?
UAG
UGA
UAA
What is a silent mutation?
Codon base is changed to codon that codes for same aa
What is a missense mutation?
Codon base is changed to codon that codes for different aa
What is a nonsense mutation?
Codon base is changed to become a termination codon (UAG, UGA, UAA)
What is a frameshift mutation?
When the reading frame is altered
What is the Wobble Hypothesis?
tRNAs can recognize more than one codon for a specific amino acid due to 'wobble' at 5' end of tRNA anticodon, forming non-traditional base pairs
What are the useful consequences of the Wobble Hypothesis?
Don't need 61 tRNAs to read 61 codons
What are nonsense suppressors?
tRNAs whose anticodons have been mutated such that they incorporate an amino acid at termination codons
What are the major steps of protein synthesis?
1. Activation of amino acids
2. Initiation
3. Elongation
4. Termination and ribosome recycling
5. Folding and posttranslational processing
What occurs during the first step of protein synthesis?
- Activation of amino acids by aa tRNA synthetase
- Requires Mg2+, ATP, amino acid, and tRNA
- Creates aminoacyl-tRNA
What does it mean to be "polycistronic" vs "monocistronic"? Which kind of cells have which kind?
- Polycistronic - mutliple genes / mRNA = prokaryotes
- Monocistronic - single gene / mRNA = eukaryotes
How does the ribosome recognize the correct translation initiation codon?
- Consensus sequence (Shine-Dalgarno sequence) just upstream from initiation codon
- Pairs with 16S rRNA to position ribosome
What transcription factor facilitates initiation? What recognizes the initiation codon?
- IF-2 in E.coli and eIF2 in humans
- Initiator tRNA carries N-formyl methionine (bacteria and mitos)
- No formylation in humans
What occurs during initiation of protein translation (step 2)?
- IF3 keeps 30S and 50S subunits apart
- 30S initiation complex binds
- tRNA carrying N-formyl methionine binds at P site
- 50S subunit binds and GTP is hydrolyzed (70S initiation complex formed)
- Release of IFs
What occurs during elongation of protein translation (step 3)?
- Elongation factor Tu (EF-Tu) delivers next aminoacyl-tRNA to A site
- Hydrolysis of EF-Tu (this sets rate of synthesis)
- Peptide bond formation by 23SrRNA ribozyme
- Transfer of amino acid in P site to A site
- Deacylated tRNA leaves via E site
- tRNA with amino acids on it moves to P site via EF-G translocase
- Repeat
How does protein translation end (step 4)?
- Release factor (RF) recognizes stop codon at A site
- RFs change specificity of peptidyl transferase to recognize water instead of amino group as acceptor of activated peptide moiety
- Peptide released
What is the initiator tRNA in prokaryotes? In eukaryotes?
- Prokaryotes - N-formylmethionine
- Eukaryotes - MET-tRNAi
Is the Shine-Dalgarno sequence important for prokaryotes or eukaryotes?
Prokaryotes - signals initiation of translation
Eukaryotes do not use the Shine-Dalgarno sequence to indicate where translation should begin; how do they know where to begin?
- In eukaryotes the AUG nearest the 5' end of the mRNA is generally utilized as the initator MET
- 40S ribosome loads at the 5' end of the mRNA and "scans"
- Stepwise movement requires ATP and helicases
What translational initiation factor in eukaryotes recognizes and binds the 5' cap of mRNA and promotes binding of mRNA to 40S subunit?
eIF4E (subunit of eIF4)
What does the Polio Virus have to do with the eukaryotic translation initiation factor eIF4E?
Causes proteolysis of cap binding protein allowing its own uncapped mRNA to be translated
What does Diptheria Toxin do?
- Blocks eukaryotic translation by inhibiting translocation
- ADP ribosylation of EF2-translocase
What are the different types of lipids?
- Fatty acids
- Waxes
- Sterols
- Phospholipids
- Fat-soluble vitamins
- Mono-, di-, and tri-glycerides
What are the main biological functions of lipids?
- Energy storage
- Signaling molecules
What are the major structural components of biological membranes?
** Glycerophospholipids (phospholipids)
- Also sphingomyelin and cholesterol
What is the structure of phospholipids?
- Glycerol backbone
- 2 ester linkages to fatty acid-derived tails
- C1 = C16 and C18 saturated FA
- C2 = C18 and C20 unsaturated FA
- Phosphodiester linkage of polar head group to C3
What molecule is a common precursor in the synthesis of phospholipids and triglycerides
Phosphatidic Acid
Where is phosphatidic acid (PA) synthesized (de novo)?
- Endoplasmic Reticulum (ER)
- Outer mitochondrial membrane
How is phosphatidic acid (PA) synthesized (de novo)?
- Glucose derivative dihydroxyacetone phosphate is reduced to Glycerol-3-phosphate (alternatively, glycerol can be phosphorylated to glycerol-3-P)
- 2 FA (from acyl-CoA derivatives) are added to glycerol-3-P by acyl transferases to create Phosphatidic Acid (PA)
What molecule is very important in the synthesis of phosphatidic acid (PA)?
Dihydroxyacetone Phosphate (glycolytic intermediate)
Alternatively, how can DAG be used to synthesize Phosphatidic Acid (PA)?
Diacylglycerol (DAG) can be phosphorylated by DAG kinase to PA
What are phosphatidates? Examples?
- Derivatives of phosphatidic acid (PA)
- Additional polar group is esterified to phosphate moiety
- Examples: phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), phosphatidylinositol (PI)
What is necessary for phosphatidic acid (PA) to be converted to the various phosphatidates derivatives?
- Hydroxyl group must be activated by attachment of cytidine diphosphate (CDP) nucleotide
- Cytidine monophosphate (CMP) is displaced in a nucleophilic attack by other hydroxyl
With what pathway does the phosphatidic acid get activated by CDP in order to form phosphatidate derivatives?
Kennedy Pathway (two ways)
1. 1,2-diacylglycerol is activated with CDP (polar head group remains inactive)
2. CDP-activated polar head group attaches to phosphate of phosphatidic acid
Eukaryotic cells can form phosphatidates from PA by one or two strategies? What pathway is this?
Kennedy Pathway
- Both strategies:
1) DAG activated w/ CDP
2) CDP-activated polar head group attached to PA
Prokaryotic cells can form phosphatidates from PA by one or two strategies? What pathway is this?
Kennedy Pathway
- Only one strategy:
1) DAG activated w/ CDP
What are the important functions of CTP?
1. Nucleic acid biosynthesis
2. Phospholipid biosynthesis (important for activation in formation of phosphatidates)
What are the important functions of CTP?
1. Nucleic acid biosynthesis
2. Phospholipid biosynthesis (important for activation in formation of phosphatidates)
What are the important functions of CTP?
1. Nucleic acid biosynthesis
2. Phospholipid biosynthesis (important for activation in formation of phosphatidates)
What are the important functions of CTP?
1. Nucleic acid biosynthesis
2. Phospholipid biosynthesis (important for activation in formation of phosphatidates)
How is phosphatidylglycerol (PG) synthesized?
- Phosphatidic Acid (PA) is condensed w/ CTP to form CDP-diacylglycerol (releasing PPi)
- Nucleophilic attack by C1-OH of glycerol 3-P (releasing CMP) to form phosphatidylglycerol 3-P
- Phosphatase cleavage of Pi to leave Phosphatidyl Glycerol (PG)
Phosphatidylglycerol (PG) is abundant in bacteria and is used for what?
Necessary for protein translocation across membranes
How is phosphatidylglycerol (PG) in animals important?
- 2nd most abundant phospholipid in lung surfactant
- May be responsible for spreading of surfactant over alveolar wall surfaces of lung
Which phospholipid increases during fetal development?
Phosphatidylglycerol (PG)
How is cardiolipin synthesized?
- Phosphatidylglycerol (PG) is condensed with CDP-diacylglycerol to form cardiolipin
Where is cardiolipin important in a cell?
- Inner mitochondrial membrane (20% of lipid)
- Many enzymes require it for metabolic functions
- Mediates translocation of cholesterol from outer to inner membrane
- Activates mitochondrial cholesterol side chain cleavage
- Imports protein into mitochondrial matrix
How is phosphatidylinositol (PI) synthesized?
- Condensation of CDP-diacylglycerol with inositol
What phosphatidate derivatives is CDP-diacylglycerol important for in the synthesis of?
- Cardiolipin
- Phosphatidylinositol
What is the primary source of arachidonic acid? What is arachidonic acid required for?
- PI (phosphatidylinositol) is primary source of arachidonic acid
- Required for biosynthesis of eicosanoids
What is the main function of phosphatidylinositol (PI)?
- Functions as negatively-charged building blocks of membranes
- Regulates non-specific electrostatic interactions with proteins
What is the function of phosphorylated derivatives of phosphatidylinositol (PI)? What enzyme is responsible for doing this?
- These derivatives are involved in regulation of cell structure and metabolism
- Plays a role in signal transduction
- PI kinases convert PI to derivatives
What is created when the PI derivative PIP2 (phosphatidylinositol 4,5-bisphosphate) is cleaved?
- DAG (diacylglycerol)
- inositol 1,4,5-triphosphate (IP3)
What is the response to the cleavage of PIP2 to DAG and IP3 (which is responsible for this signal response)?
- Calcium release
- Activation of Protein Kinase C
- IP3 is responsible
What is the second most abundant lipid in the membrane phospholipid?
Phosphatidylethanolamine (PE)
By which Kennedy Pathway strategy is phosphatidylethanolamine (PE) synthesized?
- Strategy 2
- Phosphorylation and activation of head group by CMP is followed by condensation with DAG
- Also PS can be decarboxylated to PE
Where does synthesis of Phosphatidylethanolamine (PE) take place?
- ER
- Also in outer leaflet of inner mitochondrial membrane (by decarboxylation of PS)
What is a possible function of Phosphatidylethanolamine (PE)?
Membrane fusion/fission events
Which neurotransmitter is Phosphatidylethanolamine (PE) a precursor of?
N-acylethanolamine (in the brain)
Phosphatidylethanolamine (PE) can donate its ethanolamine group towards the synthesis of what?
- Glycosylphosphatidylinositol anchors (GPI) for many cell-surface proteins
- Plasmalogens
How is phosphatidylserine (PS) synthesized?
Derived from PE or PC via one of two head-group exchange reactions carried out in ER
What is phosphatidylserine (PS) necessary for?
- Activation of signaling proteins including Protein Kinase C and neutral sphingomyelinase
- Activity of dynamin-1, Na+/K+ ATPases
- Interaction w/ Hsp70 induces formation of ion channels in plasma membrane
- On activated platelets to initiate blood-clotting cascade
- On sperm cells to initiate sperm maturation
What happens to phosphatidylserine (PS) during apoptosis?
- It is normally enriched in the inner leaflet of membrane bilayer
- Becomes exposed to outside during apoptosis
How is phosphatidylcholine (PC) synthesized?
- Strategy 2 of Kennedy Pathway: Phosphorylation (phosphocholine) and activation of choline (CDP-choline) is followed by condensation with DAG
- PEMT pathway: methylation of PE (3x)
How can phosphatidylcholine (PC) be synthesized from phosphatidylethanolamine (PE)? Compare structures.
- PEMT pathway: methylation of PE 3x
- PE head group: -CH2-CH2-NH3+
- PC head group: -CH2-CH2-N(CH3)+
- So H's on PE are substituted for methyl groups
For each PC synthesized by the PEMT pathway, what else is produced? What is the significance of this molecule?
- 3 S-adenosylhomocysteine molecules
- Independent risk factor for cardiovascular disease
Why is there a need for PC to be synthesized by two pathways (Kennedy strategy 2 and PEMT)?
- In PEMT- mice, the Kennedy pathway was increased by 50%
- If these PEMT- mice are fed a choline-deficient diet, mice exhibit liver failure in 3 days (choline is necessary for Kennedy pathway)
- Decreased PC in membranes is replaced by PE, which compromises the integrity of the membrane (liver leaks hepatic enzymes leading to inflammation and rapid liver failure)
What happens to a wild-type mouse if it is fed a choline deficient diet? Why?
- It is normal
- It has the PEMT pathway to generate PC
What is the significance of a lack of Phosphatidylglycerol (PG)?
Major surfactant deficiency in premature infants relates to lack of PG, even though it comprises less than 5% of pulmonary surfactant phospholipids
What is the significance of a lack of cardiolipin synthesis?
- Barth syndrome
- Causes infantile death
- Mutation in gene coding tafazzin (enzyme involved in synthesis of cardiolipin)
- Cardiolipin is important for ATP production; w/o enough you get abnormal mitos
- Cardiomyopathy and general weakness
- Supplemental cardiolipin treats symptoms and prevents infections
If the asymmetrical transbilayer distribution of PE in sarcolemmal membranes in the heart is altered, what can happen? What causes this dis-regulation?
- Leads to sarcolemmal disruption and irreversible cell damage
- Caused by ischemia
What alternative linkage can there be in a phospholipid at the C1 of glycerol? How is this made?
- Ether linkage (as opposed to an ester linkage)
- Synthesized by dihydroxyacetone, long chain fatty acids and long-chain fatty alcohols (ether linkage)
Where is there a higher level of ether-linked lipids (as opposed to the usual ester-linked lipids)?
- Vertebrate heart tissue
- Plasma membrane of metastatic cancer cells
What is the term for "phosphatidates" that have ether linkages at C1 instead of ester linkages? How else do they differ?
Plasmalogens
- Ether linked fatty acyl chain at C1 is unsaturated
Which type of enzyme is used to catalyze the desaturation reaction for converting the saturated C1 fatty acyl chain to an unsaturated chain?
Mixed-function oxidase
Where are plasmalogens commonly found?
Heart - half of heart phospholipids are plasmalogens
What is usual and unusual about the phospholipid structure of Platelet-Activating Factor (PAF)?
- Ether-linked (less normal) fatty acyl chain is usually saturated (normal)
- C2 contains an acetyl residue instead of a long-chain fatty acid (not normal)
- Head group is usually choline
What are the consequences of Platelet-Activating Factor (PAF) having an acetyl residue at C2 instead of a long-chain fatty acid?
Increases solubility enabling it to function in an aqueous environment
What kind of cells make platelet-activating factor (PAF)?
- Platelets
- Leukocytes
- Monocytes
- Endothelial cells
What are the functions of platelet-activating factor (PAF)?
- Mediates allergic and inflammatory responses
- Released from leukocytes (basophils) and induces platelet aggregation and release of serotonin (vasoconstrictor) from platelets
- Stimulates smooth muscle contraction and activation of immune cells
- Mediates hypersensitivity and anaphylactic shock
How is Platelet-Aggregating Factor (PAF) involved in allergic responses?
- Mediates hypersensitivity and anaphylactic shock - a severe and sometimes fatal immune response
- Causes these effects by binding to an extracellular receptor on target cells
What is the head group on phosphatidic acid (PA)?
No head group: just an H
What is the head group on phosphatidylethanolamine (PE)?
Ethanolamine:
-CH2-CH2-NH3+
What is the head group on phosphatidylcholine (PC)?
Choline:
-CH2-CH2-N(CH3)3+
What is the head group on phosphatidylserine (PS)?
Serine:
-CH2-CH-(COO-)-NH3+
What is the head group on phosphatidylglycerol (PG)?
Glycerol:
-CH2-CH(OH)-CH2-OH
What is the head group on phosphatidylinositol 4,5-bisphosphate (PIP2)
Myo-inositol 4,5-bisphosphate:
- 6 membered ring w/ 3 free OH groups and 2 phosphates (where OH's bonded to P's)
What is the head group on cardiolipin?
Phosphatidylglycerol
What are the two signaling pathways we should know performed by phospholipids?
- PLC cleavage, activation of Ca2+ release by IP3 and PKC activation
- Anchoring of membrane-associated proteins (targeting sequences needed to direct them to membrane)
Where is PIP2 located and used for?
- Found on cytoplasmic side of plasma membrane
- Serves as a specific binding site for a subset of cytoskeletal and soluble proteins
- Also serves as a reservoir of messenger molecules that are released inside cell in response to extracellular signals that interact w/ spec receptors on outside surface of plasma membrane
What are the effects of IP3 cleavage from PIP2?
- IP3 signals Ca2+ release from ER
- Ca2+ has a variety of intracellular effects, one of which is working with DAG to activate Protein Kinase C (PKC)
- PKC has many intracellular substrates and they perform many functions when phosphorylated
What kind of proteins can be covalently attached to the outer surface of the plasma membrane? How are they linked?
- Extracellular proteins (e.g., lipoprotein lipase, acetylcholine esterase)
- Glycosyl-phosphatidyl-inositol (GPI) anchor
Describe the structure of a GPI anchor.
- Phosphatidylinositol derivatives contain a short oligosaccharide (N-acetylglucosamine and mannose) covalently joined to carboxyl-terminal residue of protein through phosphatidylethanolamine
How are GPI-linked proteins usually arranged?
Arranged in clusters
How can GPI-linked proteins be released?
Phospholipase cleavage
Are intracellular proteins capable of being linked by GPI-linkages?
No, GPI-linkages are only for extracellular proteins
How are intracellular proteins linked to the membrane?
- Long-chain fatty acids (e.g., palmitic acid (16:0) attached to an internal Cys resi; muristic acid (14:0) attached to an N-terminal glycine resi)
- Isoprenoids such as polyisoprene units (farnesyl [C15] and geranylgeranyl [C20]) that attach to carboxyl-terminal Cys resi)
How do membrane-associated proteins get linked to the protein?
- Targeting sequences in primary translation products direct these proteins to membranes
Where are sphingolipids found?
- Membranes of all cells
- Highest conc. in cells of central nervous system
What is the general structure of a sphingolipid?
- Sphingosine backbone: similar to glycerol except:
- C3 has unsaturated 15C chain (not ester linkage)
- C2 has amide linkage (not ester)
- C1 has head group
What are the three subclasses of sphingolipids?
- Sphingomyelins (phosphocholine or phosphoethanolamine as polar head group)
- Glycosphingolipids (one (cerebroside) or more (globosides) sugars attached to C1
- Gangliosides (polar head groups comprised of oligosaccharides w/ one or more N-acetylneuraminic acid at termini)
Where are sphingomyelins highly prevalent?
Myelin - membranous sheath surrounding and insulating axon of some neurons
Which of the three subclasses of sphingolipids contain a phosphate group?
Only sphingomyelins (not glycosphingolipids or gangliosides)
What is the base sphingolipid called (with just an H as the head group)?
Ceramide
What sphingolipid has a clinical relevance to cholera toxin?
A specific ganglioside located on the lumenal side of intestinal mucosal cells specifically binds cholera toxin, which stimulates excess secretion that results in severe diarrhea
What are the four steps in the biosynthesis of sphingolipids?
1. Synthesis of 18C amine sphinganine from palmitoyl CoA and serine; condensation reaction is catalyzed by serine palmitoyltransferase, driven by cleavage of thioester bond of palmitoyl CoA and release of CO2
2. Attachment of FA in amide linkage to yield N-acylsphinganine
3. Desaturation of sphinganine moiety to form N-acylsphingonine (ceramide)
4. Attachment of head group to produce a sphingolipid
How are gangliosides synthesized from the basic structure ceramide?
- Addition of sugar residues
- Requires activated sugars (including UDP-glucose, UDP-galactose, UDP-N-acetylglucosamine, and CMP derivative of N-acetylneuraminic acid (CMP-NeuAc))
- Sialic acid residue attached; then additional sugars and/or sialic acid residues can be further added
What molecule provides the reducing power for glycerophospholipid and sphingolipid synthesis?
NADPH
What are the similarities of sphingolipids and glycerophospholipid synthesis?
- NADPH provides reducing power
- Fatty acids enter pathway as their activated CoA derivatives
How does the head group attachment differ for sphingolipids and glycerophospholipid synthesis?
- PC, rather than CDP-choline, serves as the donor of phosphocholine in synthesis of sphingomyelin
What kind of linkage connects the head groups of glycolipids (cerebrosides and gangliosides)?
Glycosidic linkage
Where does degradation of sphingolipids and glycerophospholipids occur?
Lysosomes
What enzymes specifically cleave phospholipids? What are the four main ones to know?
Phospholipases: A1, A2, C, D
What is the specific function of phospholipase A1?
Removes fatty acids from C1 position of glycerol backbone (permits re-esterification rxns w/ other fatty acyl CoAs)
What is the specific function of phospholipase A2?
Removes fatty acids from C2 position of glycerol backbone (permits re-esterification rxns w/ other fatty acyl CoAs)
What is the specific function of phospholipase C?
Cleaves C3 phosphodiester to produce diacylglycerol
What is the specific function of phospholipase D?
Cleaves C3 phosphodiester to produce phosphatidic acid
Degradation of sphingolipids produces what?
Sphingosine (backbone) and fatty acids
What are the disorders of sphingolipid degradation called?
Sphingolipidoses
What happens when there is a sphingolipidose?
- Accumulation of substrate in lysozyme of tissue responsible for catabolism
- Includes phagocytic cells of reticuloendothelial system, located primarily in the liver, spleen, and bone marrow, and brain
Why is the brain particularly vulnerable to sphingolipidose (disorder of sphingolipid degradation)?
Brain contains highest concentration of gangliosides; if they aren't catabolized properly they will build up in tissues
What are two specific sphingolipidose diseases (abnormal accumulation of sphingolipids)?
1. Tay-Sachs Disease
2. Niemann-Pick Disease
What is Tay-Sachs disease a disorder of? What are the symptoms?
Ganglioside degradation
- Caused by deficiency in β-hexosaminidase
- Manifested in brain
- Neurons become swollen w/ lipid filled lysozymes and infants display motor weakness and retarded psychomotor skills
- Demented and blind by age 2 and usually die by age 3
- Aggravated by high turnover of ganglioside during neonatal period
What is Niemann-Pick disease a disorder of? What are the symptoms?
- Deficiency in sphingomyelinase (the enzyme that degrades sphingomyelin into ceramide and phosphocholine)
- Results in severe mental retardation but most of lipids are deposited in liver and spleen (enlarging them)
- Death usually in early childhood
What kind of lipids act as short-range messengers? What kind of hormones are these?
Eicosanoids
- Paracrine hormones
What are the precursors of eicosanoids?
- Unesterified arachidonic acid [20:4] which is made from:
- Essential FA linoleic acid [18:2]
How is the precursor of eicosanoid synthesis obtained?
Arachidonic acid from the membrane is cleaved by phospholipase A2 in response to hormonal or other stimuli
Do eicosanoids have a short or long half-life? Why?
- Short half-life
- Rapid turnover by mitochondrial and peroxisomal β-oxidation pathways
What are the three classes of eicosanoids?
- Prostaglandins
- Thromboxanes
- Leukotrienes
How are prostaglandins and thromboxanes synthesized?
- Cyclooxygenase pathway
- Arachidonic acid becomes "cyclized" and "oxidized" by prostaglandin G/H synthase (PGS)
- Occurs in smooth ER
What are the functions of the bifunctional enzyme prostaglandin G/H synthase (PGS)?
- Important in conversion of arachidonic acid to prostaglandins and thromboxanes
- Has both cyclooxygenase (COX) and glutathione-dependent peroxidase (PG hydroperoxidase) activity
The "COX" (cyclooxygenase) function of prostaglandin G/H synthase (PGS) requires what?
- Requires 2 molecules of O2 for catalyzing cyclization of C8-C12 of arachidonic acid
- This forms prostaglandin G2 (PGG2)
What structural feature distinguishes prostaglandins from the other eicosanoids?
5-membered ring (C8-C12)
What are the two isoforms of prostaglandin G/H synthase (PGS)?
- PGS-1/COX-1 - constitutively expressed in most tissues and cells
- PGS-2/COX-2 - inducible expression at limited number of sites (e.g. inflammation, pain, fever)
How are thromboxanes synthesized?
- After prostaglandin G2 (PGG2) is created by prostaglandin G/H synthase (PGS)
- It is converted to prostaglandin H2 (PGH2)
- Then it is converted to thromboxane by thromboxane synthase (in platelets)
What is the structural change when a prostaglandin is converted to a thromboxane?
Cyclopentane ring converted to a 6-membered Oxygen containing (oxane) ring
What are the important functions of prostaglandins?
- Stimulate uterine smooth muscle contractions during menstruation/labor
- Elevates body temperature and causes inflammation and pain
- Affects blood flow to specific organs and responsiveness of certain tissues to hormones such as epi and glucagon
How is prostacyclin (PGI2) synthesized and what is its function?
- Change in functional group around cyclopentane ring
- Inhibits platelet aggregation and dilates blood vessels
What is the function of the prostaglandin derivative PGE2?
Promotes the clotting process
What produces thromboxanes? How do they function?
- Produced by platelets
- Act in platelet aggregation and formation of blood clots, and reduction of blood flow to site of clot
How is prostaglandin synthesis regulated?
- PGS-2/COX-2 (prostaglandin G/H synthase (PGS)) is only expressed in certain tissues and cells (activated macrophages and monocytes)
- Activated in response to stimulation by a number of agents including platelet activating factor (PAF)
- Inhibited by glucocorticoids
What is PGS-2 responsible for?
Elevated levels of prostaglandin, which causes inflammation
How do corticosteroids affect prostaglandins? What are the consequences?
- They inhibit phospholipase A2 (this decreases production of arachidonic acid, a necessary precursor for prostaglandins)
- Used as anti-inflammatory agents
What are NSAIDs used for?
- Non-steroidal anti-inflammatory agents
- Block prostaglandin and thromboxane synthesis from arachidonic acid
- Inhibits COX pathway of PGS (both isoforms)
What kind of drug is aspirin and how does it specifically function?
- NSAID
- Covalently acetylates COX and blocks active site
- Wide-ranging effects: decreased inflammation, fever, pain, and blood clotting
What are the side effects of NSAIDs? How?
- Stomach irritation and more serious conditions
- Inhibition of COX-1
- NSAIDs that are specific for COX-2 were developed to prevent severe pain but connected with increased risk of heart attack and stroke
What are the contradicting effects of prostaglandins and thromboxanes?
- Prostacyclin prevents blood clotting
- Thromboxane prevents blood clotting
Which of the eicosanoid types is characterized by its linear structure?
Leukotrienes
How are leukotrienes synthesized? Which synthesizes the precursor to most important leukotrienes?
- Lipoxygenase adds one O2 to arachidonic acid
- 5-lipoxygenase (5-LO)
What kind of enzyme is lipoxygenase?
- Mixed-function oxidase
- Found in heart, brain, lung, and spleen
What does 5-lipoxygenase use as a precursor? What does it synthesize? What happens to this molecule?
- Converts arachidonic acid to 5-hydroperoxyeicosatetraenoic acid (5-HPETE) via oxygen attack
- 5-HPETE is unstable and is converted by a peroxidase to more stable 5-HETE
- 5-HPETE can also be converted to epoxide called leukotriene A4 (LTA4) via a synthase
What is the structurally defining feature of leukotrienes?
Four double bonds
(and no 5 or 6-membered ring structures)
What are lipoxins?
Products of multiple lipoxygenase pathways that are characterized by hydroxyl groups attached to C5 and C6
What molecules are "pro-inflammatory"?
Most leukotrienes
Which molecule appears to inhibit leukotrienes?
Lipoxins (anti-inflammatory)
What disorders are associated with elevated levels of leukotrienes?
- Some inflammatory and hypersensitivity disorders
- E.g., asthma and anaphylactic shock
Which molecule regulates 5-lipoxygenase (5-LO)? How?
- 5-lipoxygenase-activating protein (FLAP)
- Integral nuclear membrane protein that binds arachidonic acid and facilitates the substrate interaction of this enzyme
- Several inhibitors of leukotriene synthesis bind to FLAP and inhibit its function
What is the function of the leukotriene LTB4?
- Chemotactic agent
- Attracts neutrophils to combat infection
Which molecules induce contraction of smooth muscle lining airways to lungs? When does this happen?
Leukotrienes: LTC4, LTD4, LTE4
During anaphylactic shock
How do the clinically important drugs in asthma (montelukast/Singulair and zafirlukast/Accolate) help?
- Block binding of leukotrienes to lung smooth muscle cell receptor
- Prevents contraction of smooth muscle lining airway to lungs
How does the clinically important drug for asthma (Zileuton/Zyflo) help?
Blocks 5-LO activity, preventing synthesis of leukotrienes (LTC4, LTD4, LTE4) that induce contraction of smooth muscle lining airway
How are polyisoprenoids different from other lipids?
They are derived from 5-Carbon isoprene units, rather than fatty acids
What are some important polyisoprenoids?
- Cholesterol
- Steroid hormones
- Bile acids
- Fat-soluble vitamins (A, D, E, and K)
- Ubiquinone
- Protein-membrane anchors (Farnesyl and Geranylgeranyl)
- Dolichol
What is the precursor for steroid hormones and bile acids?
Cholesterol
What is cholesterol synthesized from?
27 molecules of acetate
What are the four key steps to cholesterol biosynthesis?
1. Condensation of 3 acetate units to form a 6-carbon intermediate: Mevalonate
2. Conversion of mevalonate to activated isoprene units (5C)
3. Polymerization of six 5C isoprene units to form 30C linear squalene
4. Cyclization of squalene to form four rings of steroid nucleus, followed by a further series of changes (oxidations, removal or migration of methyl groups) to produce cholesterol
What is the rate-limiting step of cholesterol biosynthesis?
- Reduction of HMG-CoA to mevalonate
- 2 molecules of NADPH donate 2 e- each
- Catalyzed by HMG-CoA reductase (major point of regulation for cholesterol synthesis)
How are steroids different than cholesterol/sterols?
Oxidized sterols (have sterol nucleus, but lack alkyl chain attached to ring D of cholesterol); they are also more polar
Which steroid drugs have potent anti-inflammatory activities? How is their function mediated?
- Prednisone and prednisolone
- Mediated in part by inhibition of arachidonate release by phospholipase A2
- Consequent inhibition of synthesis of eicosanoids
- Useful for treatment of asthma and rheumatoid arthritis
What is the precursor for the lipid-soluble vitamins A, D, E, and K?
Cholesterol
How is Vitamin D synthesized?
- Formed in skin from 7-dehydrocholesterol in a photochemical rxn driven by UV from sunlight
What is the function of Vitamin D?
- Following enzymatic conversion in liver and kidney to 1,25-dihydroxycholecalciferol, it becomes a hormone that regulates calcium uptake in intestine and calcium levels in kidney and bone
What happens if you have a vitamin D deficiency?
Defective bone formation
How does Vitamin A / retinol function?
Functions as a hormone and as visual pigment of eye
Which vitamin is important for the treatment of severe acne and wrinkled skin?
Vitamin A derivative: retinoic acid (found in tretinoin or Retin-A)
What is the function of Retinal (vitamin A derivative)?
Pigment that neuronal initiates the response of rod and cone cells of retina to light, producing a neuronal signal to the brain
What does a deficiency in Vitamin A cause?
- Dryness of skin
- Dryness of eyes and mucous membranes
- Retarded development
- Growth and night blindness
What is the function of Vitamin E?
Hydrophobic compound associated with membranes, biological anti-oxidant
What does a deficiency in Vitamin E cause?
Very rare but leads to fragile erythrocytes
What is the significance of Vitamin K?
- Aromatic ring undergoes a cycle of oxidation and reduction during formation of active prothrombin
- Prothrombin is a blood plasma protein essential in blood clotting
What does a deficiency in Vitamin K cause?
Very rare but slows blood clotting
What kind of molecule is Warfarin? What does it treat?
- Isoprenoid
- Inhibits formation of active prothrombin (anticoagulant used to treat excessive blood clotting)
What kind of molecule is Ubiquinone or Coenzyme Q? What does it do?
- Isoprenoid
- Lipophilic electron carriers in oxidation-reduction reactions
- Drives ATP synthesis in mitochondria
What kind of molecule is Dolichol? What does it do?
- Isoprenoid
- Intermediate in activation of sugar units destined for attachment to proteins and other lipids
What kind of molecules are bile acids and their salts? What do they do?
- Isoprenoids
- Polar/hydrophobic cholesterol derivatives that act as detergents in the intestine
- Emulsify dietary fats to make them more readily accessible to digestive lipases
- In liver they aid in digestion
What technique can be used to study the structure and fundamental properties of membranes?
- Freeze Fracture Technique
- Cells are cracked open at liquid N2 temperature
- Membranes split within hydrophobic interior because free energy of binding is lower in this region
- Proper preparation of membrane reveals the interior of the phospholipid bilayer as a smooth surface and intramembrane particles that reflect the distribution of membrane proteins
What is a common protein modification on integral membrane proteins?
Oligosaccharide (sugar) chain exposed to exterior side
What is the "glycocalyx"?
- "Sugar coat"
- Negatively charged sugar coat / oligosaccharides surrounding external membrane
What is the most common membrane protein? How are they characterized?
G protein coupled receptors (GPCRs); 7 transmembrane domains
What do peripheral membrane proteins do?
Often mediate membrane interaction (usually on cytosolic side)
What are two examples of peripheral membrane proteins?
Spectrin and ankyrin - associate with cytosolic surface of cell as part of a membrane skeleton
What are some important examples of G-protein coupled membrane receptors (GPCRs)?
- β-adrenergic receptor
- Visual pigment proteins (rhodopsin)
- Na+/K+ ATPase (sodium pump)
What are the different types of carrier proteins?
- Primary active transporters
- Passive transporters
- Secondary active transporters
What are the steps of the Na+/K+ ATPase?
1. Transporter binds 3 Na+ inside
2. Phosphorylation of enzyme (from ATP)
3. Transporter releases 3 Na+ to outside and binds 2 K+
4. Dephosphorylation of enzyme
5. Release 2 K+ inside cell
6. Repeat
What values do K+ and Na+ need to be extracellularly?
[K+] = 4 mM
[Na+] = 145 mM
What kind of transporter is the Na+/K+ ATPase?
Primary transporter - uses ATP for energy needs
What ions does the anion exchange protein exchange?
Chloride and bicarbonate
Where is the chloride-bicarbonate exchanger found?
Red blood cells (RBC)
What is the function of the chloride-bicarbonate exchanger?
- Increase rate of movement of bicarbonate ions across RBC plasma membrane more than a million fold
- Necessary for gas exchange
What kind of transporter is the chloride-bicarbonate exchanger?
Antiport transporter
When RBCs or other cells are placed in medium of low ionic strength what happens?
Hypo-osmotic - take up water and will burst
How do you make "membrane ghosts"? What is the purpose?
- Put RBCs in low ionic strength medium; cell swells and bursts (leaking all of its insides)
- Put in normal ionic strength medium and plasma membrane reseals
- Now can study properties of RBC plasma membrane
How do water molecules transfer through aquaporins?
One molecule at a time
What technique can be used to determine the diffusion coefficient (D) for lateral movement of lipids in membrane?
Fluorescence Recovery After Photobleaching (FRAP):
- Put fluorescent probes on membrane lipids
- Bleach one area with laser
- Watch how long it takes for area to contain fluorescent probes again (due to diffusion)
Why is there incomplete recovery during FRAP experiments?
Some lipids are immobile
What did SDS-PAGE analysis of the RBC Ghosts show?
- Glycophorin and anion exchange protein are principle integral membrane proteins
- Peripheral membrane proteins (spectrin, actin) are organized as a membrane skeleton
- Linkage of membrane proteins like anion exchanger to membrane cytoskeleton via linker proteins such as ankyrin
- Band 4.1 greatly restrains lateral mobility of membrane protein
What is hereditary spherocytosis?
- Gene mutations that result in abnormalities of spectrin, ankyrin, or band 4.1 in membrane skeleton
- Normally biconcave RBC lose their shape and become fragile
- RBC break up as they pass through narrow capillaries
- Leads to hemolytic anemia
In hereditary spherocytosis why do the RBC not maintain their biconcave disk shape?
- Actin/spectrin network maintains shape
- This disease causes abnormalities in these proteins
What is the function of co- and post-translational modifications (PTMs)?
Increase functional diversity of a limited gene pool (only 20 AA's)
How common is glycosylation?
~50% of all proteins undergo some type of glycosylation
Why is adding sugars via glycosylation have enormous potential for increasing diversity of proteins?
There are not only many different kinds of sugars, but there are many different linkages that can combine them
What carbohydrate is generally found in glycolipids?
Mannose
What are some negatively charged carbohydrates commonly found in mammalian glycoproteins and glycolipids?
Sialic acid, glucuronic acid, and iduronic acid
What carbohydrates are found predominantly in glycosaminoglycans?
Xylose, glucuronic acid, and iduronic acid
How must monosaccharides be modified before they can function as immediate precursors in glycoprotein and glycolipid biosynthesis?
Monosaccharides must be "activated" to high-energy compounds (i.e., nucleotide sugars)
What is the major in vivo source for synthesis of nucleotide sugars?
Glucose (Glc)
What is necessary to make the early intermediate in the pathway of creating nucleotide sugars? (e.g. sugar 1-phosphate)
ATP
What is the substrate and donor of sugar residues for the reaction catalyzed by glycogen synthase?
UDP-Glucose (UDP-Glc)
What is the substrate for several families of glucuronyltransferase?
UDP-Glucuronic Acid (UDP-GlcA)
What is UDP-Glucuronic Acid (UDP-GlcA) involved in (re: glycosylation)?
- Glycoprotein / Proteoglycan synthesis
- Drug detoxification of xenobiotics
- Excretion of steroid hormones
- Heme metabolism and excretion of bilirubin (RBC turnover)
What is the function of glycosyltransferases?
- Transfer carbohydrate residues from an activated donor substrate, usually a nucleotide sugar, to an acceptor that can be a monosaccharide, an oligosaccharide (glycan), a glycoprotein, or a glycolipid
- Some even transfer sugar to hydroxyl group of some amino acids
Where are glycosyltransferases found?
Endoplasmic reticulum and Golgi apparatus
How do proteins move through the secretory pathway?
Transport vesicles through ER, Golgi, and plasma membrane
What is the required sequence for an "N-glycosidic" linkage?
Asn-X-Ser(Thr)
Sugar binds to N of Asn
What residue is bound in an "O-glycosidic" linkage?
Serine (sugar binds to hydroxyl group)
Preassembly of a glycan (oligosaccharide) chain occurs on which lipid? What kind of lipid is this?
Dolichol Phosphate (dolichol-P)
- Membrane-associated
- Isoprenoid derivative
- Precursors: mevalonic acid and cholesterol
How is dolichol-P regulated?
Regulation of cholesterol metabolism (because cholesterol is a precursor)
Which enzyme catalyzes the co-translational transfer of the 14 residue oligosaccharide chain from dolichol-P to Asn residue for an N-linked sugar? Where does this occur?
Oligosaccharyltransferase (OST) - takes place in lumen of RER
Do all N-linked sugar modifications have the same sugar sequence?
No, although they do start with the same 14 residue oligosaccharide structure (Glc3Man9GlcNAc2) this is further processed
The final oligosaccharide structure of an N-linked sugar is based on what?
The order in which that glycoprotein encountered specific processing glycosyltransferases and glycosidases
What is the function of glycosidases?
Hydrolytic enzymes that remove carbohydrates
The original N-linked oligosacchardie structure is recognized by what? What is their purpose?
- Chaperones in the ER called calnexin and calreticulin (lectins)
- Proteins that bind to a specific glycan structure that assist newly synthesized proteins fold into native conformation
What are proteoglycans and how do they differ from glycoproteins?
- Special class of glycoproteins
- Have specific glycans called glycosaminoglycans
What is the structure of proteoglycans?
- Protein backbone to which glycosaminoglycan chain(s) are covalently attached
- Glycosaminoglycans are negatively charged polysaccharides composed of repeating disaccharide units
- Disaccharide units consist of an amino sugar (glucosamine or galactosamine) and a uronic acid (glucuronic acid or iduronic acid)
- These sugars may be sulfated
How are the glycosaminoglycans attached to proteoglycans?
Xyl-Ser O-lnkage
What is the purpose of hyaluronic acid (also called "hyaluronan")?
Serves as a scaffolding protein for large numbers of proteoglycans in ECM (it is not covalently attached to protein and not sulfated)
In what tissues are proteoglycans highly prominent?
Connective tissues
What are some examples of genetic diseases that are caused by defects in glycan synthesis?
- N-glycans (congenital disorders of glycosylation or CDG)
- Glycosaminoglycans (chondrodysplasias)
- O-glycans on α-dystroglycan (at least 5 different muscular dystrophies)
What is erythropoietin (EPO)?
Growth factor secreted by kidney that stimulates production of RBCs
What is erythropoietin (EPO) used to treat?
Anemia caused by bone marrow suppression (e.g. after cancer chemotherapy)
Describe the structure of erythropoietin (EPO).
- 165 residue protein
- Contains N-linked oligosaccharides
- Heavily glycosylated w/ carbohydrates constituting ~40% of mass
If erythropoietin (EPO) is not glycosylated, how does it function?
Only exhibits 10% of normal activity
What is the most highly used drug today? What is its function?
Heparin (Lovenox) - anticoagulant
What is the functional mechanism of Heparin/Lovenox?
- Heparin forms a high affinity complex with antithrombin via Heparin's highly sulfated domains ("NS" domains)
- Antithrombin undergoes a conformational change which increases its activity 1000-10,000 fold
- Antithrombin inhibits two principle procoagulant proteases (factor Xa and thrombin) thereby decreasing the number of fibrin clots
How does the function of Heparin/Lovenox compare to Warfarin/Coumadin?
Heparin has an immediate impact on coagulation
How do proteoglycans function in structural support?
- Glycosaminoglycans bind water molecules
- This allows cartilage to resist compression in joints
- "Bubble-packing material"
How can the structural support of proteoglycans be utilized to treat osteoarthritis (OA)?
- HYALGAN - hyaluronic acid preparation
- Injected into joints
- Helps joint resist compression
How can glycoprotein structures be used as "information"?
- Cellular recognition
- Intracellular targeting
- Binding sites for bacterial toxins/parasites
What are the antigens found on blood cells made of?
Antigens are oligosaccharides whose structures are genetically polymorphic (multiple forms)
What are changes in cell surface carbohydrates associated with?
Cancer cells
How is abnormal glycosylation a marker of cancer cells?
- Changes in activity or expression of glycosyltransferases and/or glycosidases
- Frequently observed in cancer cells
What is Glycation?
- Non-enzymatic process that generates glycosylated proteins
- Long-lived proteins of lens, plasma, and RBCs are most susceptible
What happens if hemoglobin is "glycated" (HbA1c)?
- Glucose becomes covalently attached to ε-amino groups of lysine residues in Hb
- The aldehyde form of glucose first forms a Schiff base w/ NH2 amino groups of HB
- Undergoes an Amadori rearrangement to form a more stable amino ketone linkage
- Ketoamine can further cyclize to yield glycated HbA1c
The production of HbA1c (glycated hemoglobin) is dependent upon what?
Amount of glucose in blood and duration of hyperglycemia
In prolonged hyperglycemia, how is Hb affected?
Hb becomes glycated (HbA1c) - this may rise to as high as 12% or more
How can the amount of HbA1c be used to monitor diabetic patients?
- The amount of HbA1c corresponds to amount of glucose in blood and duration of hyperglycemia
- Used to monitor effectiveness of insulin treatment
What are Advanced Glycation End Products (AGEs)?
- Heterogenous compounds formed from glycated proteins that are oxidized
- They can damage other proteins by cross-linking them, causing pathological changes
How does the influenza virus bind to the host cell?
- Influenza A virus is coated w/ several antigenic proteins including hemagglutinin (HA) and sialidase
- HA on virus binds to receptors on host cell, leading to internalization and subsequent membrane fusion events
How can avian, swine, and human influenza be distinguished? Why is there a species barrier between avian and human influenza viruses
- Avian flus contains α2,3 linked sialic acid on galactose (found predominantly in intestines and respiratory tract of birds)
- Human flus contain α2,6 linked sialic acid on galactose (found predominantly on epithelial cells of human upper respiratory tract)
- Swine flu has both α2,3 and α2,6 linkages
What drugs have been developed to treat the flu after onset?
- Oseltamivir (Tamiflu) and Zanamivir (Relenza)
- Analogs of sialic acid
- Do not kill virus, but slow replication to a level where immune system can more easily destroy it
- The active site in sialidase is invariant in majority of virus strains (making these effective inhibitors)
Where does catabolism of glycolipids, glycoproteins, and proteoglycans occur?
Lysosome
How does degradation of glycolipids, glycoproteins, and proteoglycans occur?
Carbohydrate units are degraded by the sequential action of hydrolytic enzymes (glycosidases) solely from the non-reducing end of the oligosaccharide
What are lysosomal storage disorders caused by?
- Result from deficiency of a catabolic enzyme and the storage material represents the substrate of the missing enzyme
- Usually a deficiency of a glycosidase
If you are unable to cleave a particular carbohydrate linkage, what happens?
- Accumulation of that particular substrate in the lysosome
- Cell becomes distorted, inactivated, or destroyed by accumulation
When is the protein modification "hydroxylation" particularly important?
- Collagen synthesis
- Proline is converted to hydroxyproline (necessary for helix structure)
Which enzyme hydroxylates proline for collagen synthesis?
Prolyl 4-hydroxylase
What protein modification is specific to lysine residues?
Acetylation of lysine ε-amino groups (N-acetylation) in histones
How does acetylation of lysine residues on histones affect DNA?
- Acetylation removes the positive charge on the lysine side chain
- This inhibits the ability of histones, which are rich in lysine and arginine residues to interact w/ negatively charged DNA
- This affects transcription
What residues are prone to the protein modification "phosphorylation"?
- Serine
- Threonine
- Tyrosine
(all have OH groups)
Which types of enzymes phosphorylate proteins?
Protein kinases, which use ATP as substrate
Which types of enzymes dephosphorylate proteins?
Protein phosphatases
What is the effect of a phosphorylation modification?
Addition of a bulky, negatively charged group into a region of protein that was only moderately polar previously
What can be the effects of phosphorylation?
- Dramatic change in protein conformation to convert an inactive protein into an active protein
- Serve as a docking/binding site for other proteins in signaling cascades
- Serve as a modulator of the activity of enzymes
What is an example of phosphorylation changing the protein conformation?
- Insulin receptor
- On / Off switch
What is an example of phosphorylation serving as a docking/binding site for other proteins in signaling cascades?
- Erythropoietin signaling pathway that stimulates the formation of RBCs
What is an example of phosphorylation modulating activity of enzymes?
Glycogen synthase has at least 9 different phosphorylation sites, and the pattern of phosphorylation modulates the activity of the enzyme
Which protein modification adds a bulky base containing negatively charged phosphates (more than one, not phosphorylation)?
ADP-ribosylation
catalyzed by enzymes using NAD+ as substrate
Bacterial toxins such as diphtheria, cholera, and pertussis toxins act by modifying crucial host cell proteins with what type of protein modification? Consequences? Example
- Mono-ADP-ribosylation
- Critical to pathogenic mechanism that cause these diseases
- Ex: cholera toxin modifies Gs protein rendering it permanently active; ultimately leading to massive water loss, dehydration, and electrolyte loss
How are cellular responses to DNA strand breaks in mammalian cells coordinated?
By polymerization of ADP-ribose moieties onto substrate proteins carried out by poly ADP-ribose polymerase (PARP)
Why is PARP (poly ADP-ribose polymerase) a cancer drug target?
- Involved in DNA damage repair and maintaining genome stability
- Responses to DNA strand breaks is coordinated by polymerization of ADP-ribose moieties
What are the purposes of protein degradation?
1. Proteins that are improperly folded, mis-assembled, or damaged will be removed, preventing accumulation
2. Mechanism for maintaining the appropriate levels of proteins and for permitting rapid changes in levels so that cell can adapt to conditions
What are the two major protein degradation pathways in mammalian cells?
1) proteasome
2) lysosome
What molecule tags proteins for degradation?
Ubiquitin
Which accessory protein helps ubiquitin identify which proteins should be tagged for degradation?
E3
What are the three helper proteins to ubiquitin in targeting protein degradation?
- E1 = activates ubiquitin to form thioester in ATP dependent process
- E2 = ubiquitin is transferred to E2 enzymes (ubiquitin conjugating enzyme)
- E3 = links ubiquitin to target proteins (ubiquitin ligase)
How is Ubiquitin linked to target proteins?
- Isopeptide bond between:
- ε-amino group of lysine residues of target protein and
- carboxyl-terminal glycine residue of ubiquitin
When proteins are ubiquitinated, what is their fate?
Degraded by a large complex called a proteosome
What does it mean to be ubiquitinated (must be polyubiquitinated, not monoubiquinated)?
- Contain a linear, polyubiquitin chain
- Lined up for degradatio by proteasome in cytosol
How does the proteasome degrade proteins?
- Hydrolyzes ATP to unfold proteins prior to proteolysis by inner chamber of proteasome
What are the consequences of a protein being only "monoubiquitinated"?
- Not targeted to proteasome for degradation
- Some proteins are triggered to undergo endocytosis and are subsequently delivered to lysosome for degradation
What is an example of a defect in the ubiquitin-proteasome system for protein degradation?
- Heterozygous mutations in BRCA1 (Breast cancer susceptibility locus) are seen in some cases (~10%) of breast and ovarian cancer
What is BRCA1?
- Breast cancer susceptibility locus
- Heterozygous mutations are seen in 10% of cases of breast and ovarian cancer
- BRCA1 protein is an E3 ubiquitin ligase whose activity is abolished by mutations found in familial breast and ovarian cancers
What can inhibit degradation of proteins?
- Acetylation of lysine residues on E3 protein
- E3 protein usually attaches a ubiquitin molecule to lysine
What is the common function of hydrophobic protein modifications?
Promotes interaction of proteins w/in lipid bilayer
What are two fatty acid examples of hydrophobic protein modifications?
1. Palmitoylation - palmitic acid (C16) is linked through a thioester bond to cysteine
2. Myristoylation - myristic acid (C14) is linked to amino-terminal glycine
What happens during farnesylation (C15)?
- Farnesyltransferase uses farnesyl pyrophosphate as substrate
- Enzyme requires CAAX consensus sequence at C-terminus of target protein (C=cys, A=aliphatic, X=any aa)
- Farnesylation of cysteine occurs following proteolytic removal of AAX resulting in cys at C-terminus
What is an important example of the farnesylation (C15) protein modification?
Farnesylation of Ras is required for its association w/ plasma membrane, its functioning in signal transduction, and essential for transforming activity of oncogenic variants
Which clinical correlation relates to farnesylation?
- Hutchinson-Gilford Progeria Syndrome
- Aging-like phenotypes as children (avg death at 13)
- Caused by accumulation of mutant form of prelamin A (retains a farnesyl group rather than being converted to mature lamin A which lacks farnesyl group)
What is the structure of a GPI linkage?
- GPI = glycosyl phosphatidylinositol
- GPI is attached to C-terminus of target protein through a phosphoethanolamine portion of molecule
- Two fatty acid moieties of GPI are imbedded in bilayer
How does the cell establish and maintain compartmentalization?
Selective transport molecules to various organelles; often using a "tag" that is read by specific receptors
What is the name of the space between the membrane-bound organelles and takes up ~50% of cell volume?
Cytosol
What is inside of the cytosol?
- Enzymes involved in intermediary metabolism
- Ribosomes
What is the purpose of the ribosomes found on the endoplasmic reticulum (ER)?
- Actively synthesizing
- Remove selected proteins from cytosol
- Either transmembrane proteins or water soluble proteins
How does the cell establish and maintain compartmentalization?
Selective transport molecules to various organelles; often using a "tag" that is read by specific receptors
How do transmembrane proteins and water soluble proteins differ?
- Transmembrane - only partly translocated across ER membrane and become embedded
- Water soluble - fully translocated across ER and released into lumen
What is the name of the space between the membrane-bound organelles and takes up ~50% of cell volume?
Cytosol
What are the two types of water soluble proteins?
- Secretory proteins
- Proteins destined for lysosome
What is inside of the cytosol?
- Enzymes involved in intermediary metabolism
- Ribosomes
Where does all protein synthesis start?
Cytosolic ribosomes
What is the purpose of the ribosomes found on the endoplasmic reticulum (ER)?
- Actively synthesizing
- Remove selected proteins from cytosol
- Either transmembrane proteins or water soluble proteins
Where does protein synthesis finish?
- Cytosolic ribosomes
- Ribosomes attached to ER (for transport into membrane or into lumen)
How do transmembrane proteins and water soluble proteins differ?
- Transmembrane - only partly translocated across ER membrane and become embedded
- Water soluble - fully translocated across ER and released into lumen
Ribosomes that finish synthesis in the cytosol can go where ultimately?
- Mitochondria
- Nucleus
- Peroxisomes
What are the two types of water soluble proteins?
- Secretory proteins
- Proteins destined for lysosome
Ribosomes that finish synthesis on/in the ER can go where ultimately?
- First to Golgi
- Lysosomes or secretory vesicles or cell surface
Where does all protein synthesis start?
Cytosolic ribosomes
Where does protein synthesis finish?
- Cytosolic ribosomes
- Ribosomes attached to ER (for transport into membrane or into lumen)
Ribosomes that finish synthesis in the cytosol can go where ultimately?
- Mitochondria
- Nucleus
- Peroxisomes
Ribosomes that finish synthesis on/in the ER can go where ultimately?
- First to Golgi
- Lysosomes or secretory vesicles or cell surface
How are proteins targeted to the ER membrane?
- Signal sequence on protein
- Recognized by Signal Recognition Particle (SRP)
- SRP binds signal sequence and ribosome
- SRP binds to SRP receptor on ER membrane and ribosome binds to adjacent receptors
- Peptide translocation complex sends polypeptide into ER lumen
- Signal peptidase cleaves signal sequence
- Protein folds inside lumen (SRP binding prevented protein from folding until in ER)
What is the structure of the signal recognition particle (SRP) that recognizes the signal sequence targeting a protein to the ER?
- Ribonucleoprotein
- Single 7SL RNA molecule plus 6 different polypeptide chains
What are the functions of the signal recognition particle (SRP)?
- Recognizes signal sequence on growing polypeptide
- Arrests elongation so that it may translocate across membrane
Where is the signal recognition particle (SRP) located?
Cycles between ER membrane and cytosol
What is the structure of the signal sequence that targets a protein for the ER?
- No consensus sequence
- Usually 13-48 residues
- Usually occurs at amino terminus
- Tripartite domain structure: hydrophilic amino-terminal domain w/ net positive charge, hydrophobic core domain w/ α-helix, and a polar carboxyl-terminal domain
- Different primary sequence but similar 3D conformation
What removes the signal sequence from the protein once it has successfully gotten to/in the ER?
Signal peptidase
Where is the SRP receptor located?
- Exclusively in ER
- Integral membrane protein w/ SRP binding site exposed to cytosol
What is the function of the SRP receptor?
Binds the SRP on the nascent polypeptide/ribosome complex, effectively targeting complex to ER membrane
What is the function of the ribosome receptor?
Binds to the ribosome and stabilizes the ribosome/nascent polypeptide chain complex on the ER membrane
What is the function of signal peptidase
- Integral membrane protease w/ active site facing lumen of ER
- Cleaves amino-terminal signal sequences co-translationally
What happens during the translocation process across ER membrane?
- ATP hydrolysis required
- Occurs through aqueous pore or channel called "translocon"
- Translocation and protein synthesis are usually coupled since unfolded polypeptide chains are preferred
- Translocated polypeptide chains fold in lumen of ER w/ assistance of chaperones
What are "stop-transfer sequences"?
Hydrophobic, α-helical sequences which function to anchor the protein in membrane
If a protein traverses the membrane multiple times, how many stop-transfer sequences will it have?
Multiple sequences to tell protein to cross membrane again
How are transmembrane portions of a polypeptide sequence anchored into the membrane?
Hydrophobic interactions between non-polar residues and fatty acyl groups of membrane
What is the structure of the Golgi?
- 4-12 cisternae
- Two distinct sides: cis face is juxtaposed to ER and trans face is distended into a tubular reticulum called the trans Golgi network (TGN)
What happens in the trans Golgi network (TGN)?
- Site in cell where proteins that have been synthesized on ER-bound ribosomes are sorted for transport
- Go to lysosomes, secretory granules, or plasma membrane
What happens in the constitutive pathway?
- Common to all cells
- Soluble proteins are continuously secreted without intracellular storage
What happens in the regulated pathway?
- Only in certain cells (exocrine, endocrine, and neurons)
- Subset of secretory proteins is sorted to storage organelles (called secretory granules) from which they are released only upon stimulation
What kind of cells have secretory granules? Why?
- Exocrine cells
- Endocrine cells
- Neurons
- Allows contents to be released only upon stimulation
Which organelle is like a "recycling center"?
Lysosome
What is the function of the lysosome?
- Disposal of abnormal proteins
- Downregulation of cell surface signaling receptors
- Release of endocytosed nutrients
- Degradation of pathogenic organisms (phagocytosis)
- Cellular survival (autophagy)
What kind of enzymes are found in the lysosome?
- Acid hydrolases (hydrolytic enzymes)
- Optimally active at pH of 5
How is the pH of 5 maintained in the lysosome?
V-type ATPases (V for vacuolar) couple ATP hydrolysis to transport of protons into lysosome (and endosomes)
What is autophagy?
- "Self-eating"
- Major catabolic, energy-producing pathway in eukaryotes
- Involves lysosomal degradation of cytoplasmic proteins and organelles and protein aggregates
What is the major catabolic, energy producing pathway in eukaryotes?
Autophagy (self-eating)
What is the function of autophagy?
- Eliminate damaged organelles and protein aggregates
- During starvation it dramatically increases and provides internal source of nutrients for energy generation and survival
Intracellular protein aggregates and dysfunctional organelles are common features of what types of diseases?
Neurodegenerative diseases:
- Alzheimer's
- Parkinson's
- Huntington's
- Amyotrophic Lateral Sclerosis (ALS or Lou Gehrig's)
What happens in a lot of neurodegenerative diseases?
- Toxic protein aggregates and damaged organelles accumulate within specific types of neurons
- Leads to neuronal dysfunction and ultimately neuronal death
What is a potential therapeutic target of neurodegenerative diseases?
Regulation of autophagic pathways (to degrade protein aggregates and dysfunctional organelles)
The proteins found in lysosomes are synthesized where?
- On ribosomes bound to ER membrane so that they can be transported through Golgi apparatus
- Requires that they have a signal sequence
How are soluble acid hydrolases targeted to the lysosome?
- Tagged with mannose 6-phosphate
- Directed to lysosome by mannose 6-phosphate receptors
What are 3 components of lysosomal enzyme targeting?
- GlcNAc phosphotrnasferase - recognizes a protein signal on lysosomal enzymes and transfers GlcNAc-1P to specific mannose residues
- GlcNAc glycosidase (phosphodiesterase) - cleaves off GlcNAc residue generating mannose 6-P recognition marker
- Mannose 6-P receptors - recognizes mannose 6-P on lysosomal enzymes and transports them from Golgi to prelysosomal compartment (endosomes)
What is the name for the general disease of lysosome function? What is it usually caused by?
- Lysosomal Storage Disorders (LSDs)
- Defective activity of a single lysosomal hydrolytic enzyme
What are the effects of a Lysosomal Storage Disorder (LSD)?
- Intra-lysosomal accumulation of undegraded substrates
- Leads to enlarged lysosomes ("inclusion bodies")
- Leads to progressive multi-organ involvement (neurological, liver, spleen, heart, kidney, abnormal bone formation, muscle atrophy, ocular disease)
What kind of therapies are there for Lysosomal Storage Disorders (LSDs)?
- Enzyme Replacement Therapy (ERT)
- Dependent upon receptor-mediated endocytosis of mannose 6-P tagged recombinant acid hydrolases via cell surface mannose 6-P receptors
What are some examples of Lysosomal Storage Disorders (LSDs)?
- Gaucher's
- Fabry's
- Hurler's
- Pompe's disease
What causes mucolipidosis II (I-cell disease)?
Defects in lysosomal enzyme targeting because of a deficiency in the enzyme (GlcNAc phosphotransferase) that generates mannose 6-phosphate tag on proteins destined for lysosome
What are the consequences of mucolipidosis II (I-cell disease)?
- Cells lack multiple lysosomal enzymes in lysosomes and abnormally high levels of lysosomal enzymes in plasma and other body fluids
- Severe psychomotor retardation, hepatosplenomegaly (enlargement of spleen and liver), many skeletal abnormalities, coarse facial features, and restricted joint movement
- Usually die by age 10
What does vesicular transport between organelles involved?
- Coat protein-covered vesicle - buds from donor compartment
- Coat proteins released from vesicle
- Uncoated vesicle binds to a specific target protein
- Fusion of uncoated vesicle to target membrane
How are SNARES involved in vesicular fusion?
- Integral membrane proteins
- Each vesicle has its own v-SNARE which matches to a t-SNARE found on target membrane (ensures specificity)
What are the steps of vesicular fusion with SNARE molecules?
- Vesicle approaches plasma membrane of target
- v-SNARE and t-SNARE bind to each other, zipping up from amino termini and drawing two membranes together
- Zipping causes curvature and lateral tension on bilayers, favoring hemifusion between outer leaflets
- Hemifusion: inner leaflets of both membranes come in contact
- Complete fusion creates a fusion pore
- Pore widens; vesicle contents are released outside cell
What are the effects of the Tetanus toxin released by Clostridium?
- Blocks release of inhibitory neurotransmitter γ-aminobutyric acid (GABA)
- Results in a spastic paralysis ("lockjaw")
What are the effects of the Botulinum toxin released by Clostridium?
- Blocks release of Acetylcholine (ACh)
- Results in flaccid paralysis
What was "Botox" (diluted botulinal toxin) originally approved for?
- Crossed eyes
- Eyelid tics
What is Botox (diluted botulinum toxin) used for commonly?
- Overactive muscle contraction
- Brow wrinkling
- Back pain
- Bladder spasms
- Migraine headaches
- Writer's cramp
- Incontinence
- Patients w/ cerebral palsy
How do Tetanus and Botulinum toxins function molecularly?
- Peptidases that cleave SNAREs essential for synaptic vesicle fusion to plasma membrane
- Tetanus targets GABA release
- Botulinum targets ACh release
What is "endocytosis"?
Internalization of material from the external environment enclosed within a membrane derived from the plasma membrane
What are the steps of Receptor-mediated endocytosis?
- Macromolecules bind to complementary cell-surface receptors and accumulate in coated pits
- Enter the cell as receptor-macromolecule complexes in endocytic vesicles
- Endocytic vesicles fuse with endosomes where receptors and their ligands may be sorted
How do the endosomes become acidic?
V-type ATPases (although not as acidic as lysosomes)
What occurs in endosomes?
- Receptors and ligands are sorted from one another
- Selective concentrating mechanism to increase efficiency of internalization of particular ligands
What are the four pathways of receptor-mediated endocytosis (what can happen after the receptor and macromolecule are inside the endosome)?
- Receptor recycled and ligand degraded
- Receptor and ligand recycled
- Receptor and ligand degraded
- Receptor and ligand transported
What is the major protein component of coated pits/vesicles?
Clathrin - triskelion-shaped "3-legged" protein complex is composed of noncovalently associated polypeptide chains (3 heavy and 3 light chains); chains undergo self-assembly to form polyhedral protein baskets/cages
How much of the plasma membrane surface is covered in coated pits of clathrin?
2%
What is required before the vesicle can fuse to the endosome?
Removal of clathrin coat
How can iron be transported into the cells?
Via transferrin / transferrin cycle
What is the transferrin cycle?
- Iron binds to transferrin
- Transferrin bound to iron binds to transferrin receptor
- Clathrin-coated pit buds off to form clathrin-coated vesicle
- Clathrin removed and turns into endosome
- Proton pump (V-type ATPase) acidifies endosome
- Iron is released from endosome to make heme
- Endosome fuses with membrane to obtain more iron (via SNARES)
What does familial hypercholesterolemia result from?
Mutation that affects the structure and function of the LDL receptor
What is familial hypercholesterolemia (FH) characterized by?
- Elevated conc. of low density lipoprotein (LDL) in plasma
- Deposition of cholesterol in tendons and skin (xanthomas) and in arteries (atheromas)
- Inheritance as an autosomal dominant trait w/ a gene dosage effect (homozygotes are more severely affected)
What is one of the most common inborn errors of metabolism?
Familial hypercholesterolemia (FH) (heterozygotes 1 in 500 persons; homozygotes 1 in 1 million)
What are the consequences of familial hypercholesterolemia (FH)?
Coronary heart disease begins in childhood
How can the mutations of the LDL receptor that causes familial hypercholesterolemia (FH) be classified?
5 classes:
- 1 = defect in synthesis
- 2 = defect in transport
- 3 = defect in binding
- 4 = defect in clustering
- 5 = defect in recycling
Signaling receptors are what kind of proteins? What is their function?
- Transmembrane proteins on cell surface; upon binding of ligand they become activated and generate various intracellular signals
- Also intracellular receptors which are stimulated by small hydrophobic signal molecules that can diffuse across membrane
What are the four types of signaling molecules?
- Contact-dependent
- Paracrine
- Synaptic
- Endocrine
When is contact dependent signaling particularly important/utilized?
Development and in immune responses
When does paracrine signaling occur?
- Between different cell types
- Called autocrine signaling if same cell type
- Cancer cells often rely on autocrine signaling for survival and proliferation
What is the function of synaptic signaling?
Coordinates behavior of cells in remote parts of body; very fast
What does endocrine signaling depend upon?
Diffusion and blood flow; relatively slow
Describe what kind of cell signaling changes are "slow" responses?
- Increased cell growth
- Changes in gene expression
- Changes in protein synthesis
Describe what kind of cell signaling changes are "fast" responses?
- Cell movement
- Cell secretion
- Metabolism (rapid phosphorylation of effector proteins)
The same signal can mediate many different effects, how does this happen? Example?
- Different receptor proteins
- Different intracellular machinery
- Heart muscle and salivary gland cells use same ACh receptor, but ACh induces different cellular responses (due to difference in intracellular machinery)
How is rapid relaxation of smooth muscle cells coordinated?
- Autonomic nerves in vessel wall release ACh
- ACh acts on nearby endothelial cells by stimulating NO synthesis
- NO diffuses across membrane and binds to guanylyl cyclase in smooth muscle cells
- Activated guanylyl cyclase converts GTP to cGMP
- cGMP induces rapid relaxation of smooth muscle cells
Intracellular receptors are structurally related and belong to what very large superfamily? What structural feature do they have?
Nuclear Receptor Superfamily - contain DNA binding domains that regulate transcription
How do most signaling ligands that bind to intracellular nuclear receptors enter the cell?
Via carrier proteins for transport in blood stream; dissociate from carrier proteins when they enter into cells
When steroid hormone receptors translocate into the nucleus upon ligand binding, what happens?
Ligand binding induces conformational changes in nuclear receptors and affects their interaction with either inhibitory proteins or coactivator proteins; this determines whether they regulate transcription positively or negatively
Direct stimulation of early response gene expression can occur how quickly?
In 30 minutes
What is the "delayed response"?
The gene products from the "early response" induce expression of other genes "delayed response"
What are the roles of the early response gene products?
- Induce expression of delayed response genes
- Negative feedback on primary response genes
Why is nuclear receptor signaling usually specific?
- Only certain types of cells have receptors
- Each of these cell types contains a different combination of other cell-type-specific gene regulatory proteins that collaborate w/ activated receptor to influence transcription of specific genes
What are the three largest classes of cell surface receptors?
- Ion-channel coupled receptors
- G-protein coupled receptors
- Enzyme coupled receptors
What are ion-channel coupled receptors involved in?
Rapid synaptic signaling (aka transmitter-gated channels or ionotropic receptors)
What are G-protein coupled receptors involved in?
Indirectly regulate plasma membrane-bound enzymes or ion channels, which is mediated by a trimeric GTP-binding protein
What are enzyme coupled receptors involved in?
Function as enzymes or associate directly with enzymes that they activate
What do "molecular switch" proteins do?
Relay signal to next signaling component in chain
What do "scaffold proteins" do?
Bring two or more signaling proteins together so that they can interact more quickly and efficiently
Primary signals are transduced to produce large amounts of what?
Large amounts of small intracellular mediators or many copies of a downstream signaling protein, which amplifies the signal
What is a signaling cascade?
When there are multiple amplification steps in a relay chain
What is a coincidence detector?
A molecule that receives signals from two or more signaling pathways and integrates them before relaying a signal onward
What would be the purpose of a protein anchor in a cell signaling pathway?
Some proteins anchor one or more signaling proteins in a pathway to a particular structure in cell where signaling proteins are needed
What are examples of "molecular switches"?
- Phosphorylation
- GTP-binding
- (also cAMP-binding, Ca2+-binding, and protein modifications such as ubiquitylation)
When a G-protein has GTP bound is it on or off? How can it modify this?
On - intrinsic GTPase activity to shut themselves off by hydrolyzing their bound GTP to GDP
What are the two types of GTP-binding proteins in cells?
- Monomeric GTP-binding proteins (small GTPases)
- Trimeric GTP binding proteins (G proteins)
What is the function of GTPase-activating proteins (GAPs)?
Increase rate of GTP hydrolysis of small GTPases (leads to inactivation)
What do many receptor tyrosine kinases do?
Generate specific docking sites on themselves to interact with signaling proteins
What do phosphoinositides do?
Generate docking sites in plasma membrane to recruit intracellular signaling proteins
What are some examples of highly conserved small interaction domains? What do they bind to?
- Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains bind to phosphorylated tyrosine
- Src homology 3 domains bind short proline rich amino acid sequences
- Pleckstrin homology (PH) domains bind to charged head groups of specific phosphoinositides
What interaction domains bind to phosphorylated tyrosine?
SH2 (src homology 2) and PTB (phosphotyrosine-binding)
What interaction domains bind to short-proline rich amino acid sequences?
SH3 (src homology 3)
What interaction domains bind to charged head groups of specific phosphoinositides?
Pleckstrin homology (PH)
What is desensitization / adaptation?
Prolonged exposure to a stimulus decreases the cell's response to the level of stimulus
What are some different methods of achieving desensitization/adaptation?
- Receptor sequestration
- Receptor down-regulation
- Receptor inactivation
- Inactivation of signaling protein
- Production of inhibitory protein
Which coupled receptor mediates most cellular responses to signals?
G-protein coupled receptors (GPCRs)
What kind of signal molecules are there for GPCRs?
- Proteins
- Small peptides
- Derivatives of amino acids and fatty acids
- Photon of light
- Smell and taste
What is the structure of GPCRs?
- Single peptide chain
- 7 transmembrane domains
What is a guanine nucleotide exchagne factor (GEF)?
Molecule that exchanges GDP for GTP, thus activating the α subunit
What is a regulator of G-protein signaling (RGS)?
- Increases GTPase activity
- Counterparts of GAPs that regulate small GTPases
What are the three G-proteins we need to know and what are their functions?
- Gs = (stimulatory) activates adneylyl cyclase, activates Ca2+ channels
- Gi = (inhibitory) inhibits adenylyl cyclase
- Gq = activates phospholipase C-β
What is the steps of activating Gs protein?
- Signal molecule activates GPCR
- GEF (guanine nucleotide exchange factor) activates α subunit by exchanging GDP to GTP
- Gs-α activates adenylyl cyclase
- AC converts ATP to cAMP
- cAMP binds to 2 regulatory subunits of Protein Kinase A, releasing 2 catalytic PKA subunits
- Activated PKA phosphorylates CREB
- CREB-binding protein (CBP) binds CREB
- Gene transcription is activated
How does Cholera toxin affect the Gs-protein?
- Cholera toxin transfers ADP-ribose from NAD+ to α-subunit of Gs
- Inhibits GTP hydrolysis
- Sustains active conformation of Gs and subsequently sustains adenylyl cyclase
- Leads to prolonged cAMP production in intestinal epithelial cells leading to large efflux of Cl- and water into gut
- Causes diarrhea
How does Pertussis toxin affect the Gs-protein?
- Pertussis toxin catalyzes ADP-ribosylation to α subunit of Gi
- Prevents protein from interacting with receptors
- As a result, G protein remains in GDP-bound form
- Unable to regulate target proteins
- Whooping cough
What are some responses to increases in cAMP?
- Thyroid gland: thyroid hormone synthesis and secretion via TSH
- Adrenal cortex: cortisol secretion via ACTH
- Ovary: progesterone secretion via LH
- Muscle: glycogen breakdown via adrenaline
- Bone: bone resorption via parathormone
- Heart: increase in HR and force of contraction via adrenaline
- Liver: glycogen breakdown via glucagon
- Kidney: water resorption via vasopressin
- Fat: triglyceride breakdown via adrenaline, ACTH, glucagon, and TSH
What is the function of phospholipase C-β? How is it activated?
- Activated by Gq-protein
- Cleaves phosphotidylinositol 4,5-bisphosphate (PIP2) to inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG)
How does phospholipace C-β activation by Gq-protein ultimately lead to activation of Protein Kinase C?
- Phospholipase C cleaves PIP2 into IP3 and DAG
- IP3 induces release of Ca2+ from ER
- DAG and Ca2+ activate protein kinase C (PKC)
What are some of the major responses to PLC-β?
- Liver: glycogen breakdown via vasopressin
- Pancreas: amylase secretion via ACh
- Smooth muscle: muscle contraction via ACh
- Blood platelets: platelet aggregation via thrombin
What is another function of DAG besides mediating activation of PKC?
Mediates pain and inflammatory responses by regulating synthesis of prostaglandins (which is inhibited by most anti-inflammatory drugs)
What modification is performed to desensitize GPCR proteins?
GPCRs are phosphorylated by PKA, PKC or GPCR kinases (GRKs) indicating feedback regulation
What is the function of Arrestin?
- Prevents phosphorylated GPCR from interacting w/ G proteins
- It also couples GPCR to clathrin-dependent endocytosis machinery
What five principle classes of enzyme-coupled receptors are used in humans?
1. Receptor tyrosine kinases
2. Tyrosine kinase-associated receptors
3. Receptor Ser/Thr kinases
4. Receptor guanylyl cyclases
5. Receptorlike tyrosine phosphatases
What is the function of Receptor tyrosine kinases (RTKs)?
- Directly phosphorylate specific tyrosines on themselves and on a small set of intracellular signaling proteins
- Ligand binding causes receptor chains to dimerize, bringing the kinase domains of two receptor chains together so they can become activated and cross-phosphorylate each other (transautophosphoryation)
- Tyr phosphorylation increases kinase activity
- Tyr phosphorylation creates high-affinity docking sites for binding of specific signaling proteins w/ SH2 or PTB domains
What is the function of Tyrosine kinase-associated receptors?
- Directly recruit cytoplasmic kinases to relay signals
- No intrinsic kinase activity
- Ex: Cytokine receptors mediate signaling via (Janus Kinase) JAK-STAT pathway
What is the function of Receptor Ser/Thr kinases? Example?
- Directly phosphorylate specific Ser/Thr residues on themselves and proteins they associate with
- Transforming growth factor-β (TGFβ) family members mediate development, immune responses, and cell proliferation and death
What is the function of receptor guanylyl cyclases?
- Directly catalyze production of cGMP
What is the function of receptorlike tyrosine phosphatases?
Remove phosphate groups from tyrosine of specific intracellular signaling proteins; they are receptor-like because their ligands are not known
What is an important example and exception to the normal rules of receptor tyrosine kinases (RTKs)?
- Insulin receptor, IGF1 receptor (ligand binding rearranges their transmembrane receptor chains, moving the two kinase domains close together)
- Do not generate docking sites on the receptor but use a specialized docking protein called insulin receptor substrate 1 (IRS1)
What are some functions of Ras proteins (small GTPase)?
- Molecular switch for RTK signaling
- Relays signals from RTKs