Molecules to Cells Quiz #1 Flashcards

0
Q

What is the structure of an amino acid?

A

Each amino acid (except for proline) has a carboxyl group, an amino group, and a distinctive side chain (often called the R group) bonded to the α-carbon atom.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
1
Q

All amino acids (except glycine) found in proteins are in what configuration?

A

L-configuration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Protein secondary stucture: The process of folding typically follows a hierarchical sequence in which secondary structures involving __________ between __________ groups and __________ form first.

A

H-bonds, peptide bond carbonyl (C=O) groups, peptide bond amide hydrogens (>N-H)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Cystic Fibrosis

A

Chloride ion channel inhibited

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Each amino acid (except for proline) has a ________ group, an ________ group, and ________ bonded to the ________.

A

carboxyl, amino, an R group, α-carbon atom

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Name all of the hydrophobic aliphatic amino acids.

A
GAVLIMP
Glycine
Alanine
Valine
Leucine
Isoleucine
Methionene
Proline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Name all of the hydrophobic aromatic amino acids and their characteristic UV absorbance spectra

A

WYF (wives smell good)
Tryptophan
Tyrosine
Phenylalanine

280 nm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Name all of the hydrophilic uncharged polar amino acids.

Why are these called uncharged?

A
STNQC (stinks)
Serine
Threonine
(N) Asparagine
(Q) Glutamine
Cysteine

Their side chains do not ionize at physiological pH.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Note that the _______ groups of asparagine and glutamine are _______ and never carry a __ charge. Those side-chains are ______, not _______.

A

–NH2, not ionic, +, amides, amines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Name the hydrophilic charged amino acids. Indicate which can be positively and negatively charged.

A
K HERD
(K) Lysine 
Histidine
(E) Glutamic acid (glutamate)
(R) Arginine 
(D) Aspartic acid (aspartate)

Lysine, histidine, and arginine are positively charged and glutamic and aspartic acid are negatively charged.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

The ratio of protonated (HA) to unprotonated (A) forms can be calculated from the Henderson-Hasselbalch Equation: ___________________. Note that when the pH is one unit higher than the pKa, the [A]/[HA] ratio is ___; 2 pH units higher and [A]/[HA] is ___.

A

pH = pKa + log[A]/[HA], 10, 100

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Define the isoelectric point, pI

A

The pH at which an amino acid, peptide or protein will have a net charge of zero

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q
pKa = pH -> 50% prot/50% unprot
\+1 pH unit -> \_\_% prot/\_\_% unprot
\+2 pH units -> \_\_% prot/\_\_% unprot
-1 pH unit -> \_\_% prot/\_\_% unprot
-2 pH units -> \_\_% prot/\_\_% unprot
A

+1 pH unit -> 9% prot/91% unprot
+2 pH units -> 1% prot/99% unprot
-1 pH unit -> 91% prot/9% unprot
-2 pH units -> 99% prot/1% unprot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

How do you calculate the isoelectric point (pI) from two pKa’s?

A

pI = (pK1 + pK2)/2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Explain the buffering effect of amino acids.

A

The pH of a solution is stabilized to changes from added base or acid near the pKa values of ionizable groups on amino acids.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

The _________ group of __________ can spontaneously _________ to form a disulfide bond.

A

thiol/sulfhydryl, cysteine, oxidize

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

__________ bonds are important in stabilizing the circulating peptide hormone __________

A

Disulfide, insulin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

The amino acids of proteins are joined by ________ bonds between the ________ group of one amino acid and the ________ group of the next.

A

peptide, carboxyl, amino

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Proteins are synthesized on ribosomes from __-terminal to __-terminal and they are by convention drawn with the __-terminal to the left, __-terminal to the right.

A

N, C, N, C

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Explain what limits the rotation around a peptide bond between amino acids.

A

The strong electronegativity of the carbonyl oxygen relative to the amide nitrogen within the peptide bond tends to delocalize electrons and create resonance. The C-N linkage has double bond character in 40% of structure. Since double bonds are always planar, peptide bonds exhibit this planar character with little bond rotation around the amide bond.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q
The preferred (i.e., low energy) orientation of adjacent R groups is \_\_\_\_\_\_\_\_\_\_\_\_\_ because \_\_\_\_\_\_\_\_\_\_\_\_\_ is 
minimized. This limits the number of available conformational states.
A

trans, steric hindrance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are the orientations of the bonds flanking the alpha carbon in a polypeptide chain called? How do they limit conformational possibilities of protein folding?

A

phi (N-C) and psi (C-C) angles; the rotation around these bonds is limited, many combinations
of phi and psi angles cause the side chains of amino acids to clash and are thus unfavored. (Ramachandran plot)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

The ______-handed alpha helix contains ___ residues per turn and is stabilized by ________ bonds
between the carbonyl of amino acid i and the amide proton of amino acid i+__. The H-bonds are ________ to the helix axis and the amino acid side chains (R groups) project ________________, minimizing ____________. Alpha helices on the surface of a protein are ___________, having a ___________ face projecting out into solvent and a __________ face projecting inward to the apolar core of the protein.

A

right, 3.6, hydrogen, 4, parallel, radially outward, steric repulsion, amphipatic, hydrophilic, hydrophobic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

The beta pleated sheet is stabilized by _______ bonds running between _____________ atoms and _______ protons of non-contiguous parts of the polypeptide chain. Hydrogen bonds are ________ with the beta sheet. Side chain R groups ___________________________ the plane of the sheet. Beta sheets often form _________________ of proteins.

A

hydrogen, carbonyl oxygen, amide, co-planar, alternately project above and below, the hydrophobic core

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Parallel beta sheets have ________ aligned in the __________. Anti-parallel beta sheets have ________ that ____________________.

A

peptides, same direction, peptides, alternate in direction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Which amino acid are often found in the places where α-helices or β-pleated sheets end and why?

A

Proline, because its ring structure prevents it from forming the H-bonds required for α-helices or β-pleated sheets.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is a domain?

A

Domains are the smallest thermodynamically stable units of protein structure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are metamorphic proteins?

A

Metamorphic proteins are proteins that exist in multiple distinct structures of approximately equal energy that are in equilibrium (envision a thermodynamic folding funnel with two wells of similar depth)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Where is keratin found, and explain the structure of keratin.

A

Keratin is the primary component of hair and nails.
It is a coiled coil protein that consists of two right-handed α-helices intertwined in a left-handed super-coil (called an α-coiled coil). Coils bond with each other via non-covalent (hydrophobic interactions, ionic bonds, H-bonds) and covalent (disulfide bonds) bonds.
A much higher number of disulfide cross-links makes nails (and horns, claws and hooves) rigid.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the most abundant protein in the body?

A

Collagen (20-25%)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is the structure of collagen?

A

A typical collagen molecule is a long, rigid structure in which three left-handed helices (called α-chains) are wound around each other in a right-handed triple helix (called a coiled coil or super-helix).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What amino acids are important in collagen? Explain.

A

Collagen is rich in proline and glycine. The proline rings stack along the outside of the helix giving it rigidity. Glycine, having the smallest R group, is found at every third position of the polypeptide chain (repeating sequence, -Gly-X-Y-, where X is frequently proline and Y is often either hydroxyproline or hydroxylysine) so that it can fit into the restricted central space where the three strands come together.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Describe the mechanism of scurvy

A

The enzymes that hydroxylate the prolines and lysines of collagen to stabilize the triple-helical structure require ascorbate (vitamin C). The symptoms of scurvy (e.g., bleeding gums) are due in part to the decreased tensile strength of collagen.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

“Cork screw” hair is often the earliest sign of scurvy and is thought to be due to inability to _________________________________.

A

rearrange the disulfide bonds of keratin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Outline the steps of type I collagen synthesis

A

Synthesis, post-translational modification, and triple helix assembly occur inside the cell, then the resulting procollagen molecule is secreted into the extracellular matrix where peptidases cleave N- and C-terminal propeptides. Once removed, the mature collagen triple helix (termed tropocollagen) self-assembles into fibrils, with subsequent cross-linking to form mature collagen fibers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Where is elastin found, and describe its structure and assembly

A

Elastin found in the lungs, large arterial walls, and elastic ligaments.
The elastin polypeptide is comprised predominantly of small, nonpolar amino acids and is also rich in proline and lysine

Synthesis and Assembly: Elastin is synthesized from a precursor, tropoelastin, which is
secreted into the extracellular space where it interacts with specific glycoprotein microfibrils. Some of the lysyl side chains are oxidized to form allysine residues which cross-link with lysine amino groups of neighboring polypeptides to produce an extensively interconnected, rubbery network that can stretch and bend in any direction when stressed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Explain the mechanism of emphysema.

A

Elastase degrades elastin. Alpha1 antitrypsin (alpha1-AT) is a small protein that inhibits elastase, which is released by neutrophils. Elastase can destroy the alveolar epithelium if unregulated by alpha1-AT.

Causes:
Genetic: Inheritance of an allele with a E342K substitution that causes alpha1-AT to be retained inside the cell.
Environmental: Elements in cigarette smoke can oxidize a methionene residue in alpha1-AT that is essential for binding to elastase. This renders alpha1-AT unable to bind and inactivate elastase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Explain the cooperative binding of oxygen to hemoglobin.

A

Binding of each O2 molecule to the hemoglobin tetramer increases the affinity with which the next O2 binds. Similarly, dissociation of each O2 decreases affinity for those remaining and they dissociate more easily. This cooperativity results in a sigmoidal O2 binding curve and that greatly increases the ability of Hb to release O2 in tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What four things decrease hemoglobin’s affinity for oxygen?
Which of these is important in allowing hemoglobin to release oxygen in peripheral tissues?
Which is important in adapting to hypoxia?

A

H+ (lower pH), CO2, 2,3-DPG and increasing temperature (fever).

H+ and CO2 play an important role in allowing oxygenated hemoglobin to release O2 in tissues. 2,3-DPG allows hemoglobin to adapt to hypoxia by more easily releasing O2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Explain the mechanism behind sickle cell disease.

A

Sickle cell disease is caused by an inherited homozygous mutation of the β-globin gene causing a change from glutamate (Glu) to valine (Val) at position 6 (E6V). This results in patients’ RBCs containing mainly hemoglobin S (HbS), which is comprised of two normal adult alpha-globin subunits and two sickle adult β-globin subunits. Valine is hydrophobic and its presence creates a sticky patch on deoxyHb that leads to polymerization of HbS tetramers into long chains. Those intracellular fibers cause the sickle cell shape of the RBCs that leads to problems in their passage through the microcirculation. These structural changes also lead to a shorter erythrocyte half-life and chronic hemolytic anemia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Describe the structure of the hemoglobin genes.

A

Two alpha genes on chromosome 16, one beta gene on chromosome 11. The hemoglobin tetramer is comprised of 2 subunits coded by the α-globin gene family and 2 subunits coded by the β-globin gene family.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Why does oxygen need to be transported by hemoglobin in RBCs?

A

Because oxygen has low solubility in plasma (the non-cellular part of blood)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What happens to carbon dioxide in a RBC?

A

RBCs contain carbonic anhydrase which catalyzes the rapid reversible hydration of CO2 to carbonic acid (H2CO3). Carbonic acid then rapidly and spontaneously dissociates to bicarbonate (HCO3-) and a H+.

CO2 and, especially, bicarbonate are soluble in plasma and RBC cytosol and most of the CO2 made in tissues returns to the lungs as those species. About 14% of the CO2 made is carried bound to Hb (bound to α-amino groups of Hb).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Which form of iron can bind oxygen?

A

The ferrous form (Fe2+); Fe3+ is the ferric form of iron that cannot bind O2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What is myoglobin? How is it related to hemoglobin?

A

A monomeric protein abundant in muscle that is designed to store oxygen.

The alpha and beta subunits of hemoglobin are evolutionarily related to myoglobin, both proteins contain a Fe2+-protoporphyrin IX (Heme) prosthethic group that is responsible for binding O2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Explain the differences between the oxygen binding curves of myoglobin and hemoglobin.

A

Myoglobin gives a normal binding curve which is hyperbolic in shape. Hemoglobin shows sigmoidal cooperative binding of oxygen. The cooperativity of hemoglobin allows it to release a much larger fraction of its O2 load at the pO2 levels found in the blood of working and even resting muscle.

For both proteins, the partial pressure of oxygen yielding 50% saturation of binding is termed the P50

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the oxygenated and deoxygenated forms of the hemoglobin subunits called?

A

deoxygenated: T form (tense)
oxygenated: R form (relaxed)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What does it mean when a hemoglobin oxygen binding curve shifts to the right?

A

When the binding curve shifts right, hemoglobin loses its affinity for oxygen, making it more apt to release it into the surrounding tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is the difference between the oxygen binding curves for adult and fetal hemoglobin?

A

The fetal hemoglobin binding curve is shifted to the left, because it has a higher affinity for oxygen. This means the fetus’ circulation can draw oxygen from maternal blood at the pO2 present in placenta.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q
Clinical Correlation:
Carbon monoxide (CO) has a much higher affinity for Hb than does O2.  When bound to the heme group of one subunit, it causes all four subunits to “lock” in the R conformation.  What effect would this have on O2 delivery?
A

Hemoglobin would be locked in the conformation that has very high affinity for oxygen (R conformation), causing it to not release oxygen into the surrounding tissues. This would also tie up binding sites preventing any more oxygen to be taken up and transported.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

How does O2 binding change the conformation of a Hb subunit?

A

Without oxygen bound, the heme Fe2+ is pulled away from the plane of the porphyrin ring by a His residue of the hemoglobin polypeptide chain (a His ring N is bound to the Fe2+). When oxygen binds, it pulls the Fe2+ back into the plane of the ring and that moves the His residue and causes the hemoglobin subunits to shift to an arrangement that favors the R-conformation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Describe the allosteric regulation of hemoglobin.

A

H+, CO2, and 2,3-DPG ALL can bind to hemoglobin and reduce its affinity for O2. Correspondingly, if oxygen is high, then the equilibrium is driven to the right and H+ and CO2 and DPG will dissociate from hemoglobin as oxygen binds.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What kind of effectors are H+ and CO2 to hemoglobin?

A

Heterotropic negative allosteric effectors

They are heterotropic because they are not O2; negative because they decrease affinity for O2; and allosteric because they bind to a site other than the O2 site(s) affected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the Bohr effect/isohydric shift?

A

Hemoglobin’s oxygen binding affinity is inversely related to both acidity and concetration of carbon dioxide. Changes in H+ binding result from a shift in the pKa of specific residues (mostly histidines) due to microenvironment effects triggered by conformational changes in the hemoglobin molecule.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Where does 2,3-DPG bind to the hemoglobin tetramer?

A

Between the beta subunits

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What is the relationship between 2,3-DPG and high altitudes?

A

Because there is less O2 at high altitudes, tissues tend to become somewhat hypoxic. RBCs adapt to this by increasing the concentration of 2,3-DPG, making it easier for O2 to dissociate from Hb.
Other causes of tissue hypoxia such as anemia and smoking also cause RBCs to increase 2,3-DPG.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What type of genetic disease is sickle cell?

A

Homozygous recessive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What causes HbS not to polymerize in sickle cell disease?

A

The presence of HbF (fetal hemoglobin).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is the effect of hydroxyurea on sickle cell disease patients?

A

It stimulates HbF production. More HbF means less HbS in a sickle erythrocyte, which reduces sickling and the associated clinical complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Which amino acids have more than one chiral center?

A

Threonine and Isoleucine have 2 chiral centers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Name four factors that contribute to stabilizing the native structure of a protein?

A

Covalent (peptide, disulfide) bonds, hydrogen bonds, hydrophobic (van der Waals) interactions, ionic interactions (+ to – attraction).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Which amino acids contain sulfur?

A

Methionene and cysteine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Why is the R group of histidine particularly well suited to both accept and to donate a proton during enzyme catalysis?

A

It can either be protonated (+) or deprotonated (uncharged) at physiological pH without needing significant microenvironment effects. pKa ~ 7

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What is the three letter code for aspartic acid?

A

Asp

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is the three letter code for asparagine?

A

Asn

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is the three letter code for glutamic acid?

A

Glu

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is the three letter code for glutamine?

A

Gln

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What is the single letter code for asparagine?

A

N

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is the single letter code for glutamine?

A

Q

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the single letter code for glutamic acid?

A

E

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

How does a perm work?

A

A thiol is used to reduce the disulfide bonds that stabilize α-keratin and then after creating the desired shape, hydrogen peroxide is used to reform new disulfides.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What hemoglobin are present during embryonic stages of development?

A

2 epsilon and either 2 alpha or 2 zeta chains; declining in first 10 weeks of gestation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What would the dissociation of hemoglobin into its subunits do to the oxygen dissociation curve?

A

It would shift the curve substantially to the left because they would all have myoglobin-like affinities to oxygen, very high affinity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Individual nucleotides are joined to each other at the __ and __ carbons through a ___________ bridge

A

3’, 5’, phosphodiester

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How many angstroms are there in the major and minor grooves of B-form DNA?

A

12 angstroms and 6 angstroms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

DNA structure: ______ are on the inside of the helix and ______ is on outside. The planes of the bases are “stacked” ______ to the helix axis. Stacking resonance helps stabilize double helix.

A

Bases, phosphate sugar backbone, perpendicular

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q
DNA structure: Relaxed DNA has \_\_ bp per turn of the helix at intervals of \_\_ angstroms.
Two chains of helix are held together by \_\_\_\_\_\_\_\_\_\_\_\_\_\_\_\_\_\_\_\_.
Spacing constant (space between bases): \_\_ angstroms.
A

10.5, 36, hydrogen base pairing, 2.9

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

The two strands of a double helix separate when _________ are disrupted by ______ or _______. The _______ composition (______ concentration) of the solution will effect denaturation and annealing rates.

A

hydrogen bonds, changes in pH, heating, ionic, salt

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What property of DNA does a FISH assay take advantage of?

A

Denaturation and rehybridization. FISH uses small fluorescently labeled probes that bind to specific sequences on the denatured DNA to measure the presence of specific genes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Define hyperchromic shift

A

Hyperchromic shift allows for the monitoring of DNA melting spectrophotometrically, because denatured DNA strands absorb much more UV light than double stranded DNA at 260 nm. This is because in double stranded DNA, bases are stacked on top of each other, whereas in denatured DNA the bases are exposed.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Define the melting temperature (Tm) of DNA and what two factors it is affected by.

A

The temperature at which 50% of the helical structure is lost. It is affected by the GC content of the DNA (higher GC content = higher Tm). It is also affected by ionic strength–high saltfavors duplex, low salt favors denatured.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

In cells, DNA is usually found in a somewhat _____ supercoiled state, which means it is twisted in the _____ direction as the helix, turning it ____ward, which ______ the DNA.

A

negatively, opposite, left, unwinds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is the function of a topoisomerase?

A

To change the topological state of circular or supercoiled DNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What kind of enzyme is a type I topoisomerase? What is it’s main function? How is this accomplished?

A

They are nicking-closing enzymes. They create transient single-stranded breaks to relax supercoiled DNA.

The enzyme acts by:

1) cleaving one strand of DNA
2) passing a segment of DNA through the break
3) resealing the break
4) DOES NOT require ATP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What residue in the active site of type I topoisomerase attacks the DNA strand to be cleaved, and what part of the DNA does it target?

A

Tyrosine residue, attacks the phosphodiester bond

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What is the main function of type II topoisomerase? How is this accomplished?

A

They create transient double-stranded breaks in DNA.

They REQUIRE ATP to complete a reaction cycle in which:

1) two strands are cleaved
2) the DNA is passed through the break
3) the break is resealed

86
Q

How are topoisomerases clinically relevant?

A

Prokaryotic topoisomerases are targets for many antibiotics, target cells will not be able to relieve torsional stress and they die. Topoisomerase function is also targeted in cancer, either to stop topoisomerase function altogether or by locking them in a nickase state, leaving all the DNA in the tumor cells cut up.

87
Q

Which bases are purines?

A

Adenine, guanine, xanthine, hypoxanthine

88
Q

Which bases are pyrimidines?

A

Cytosine, thymine, uracil

89
Q

What is the structural difference between purines and pyrimidines?

A

Purines have two rings and pyrimidines have one.

90
Q

What is a structural difference between adenine and guanine?

A

Guanine has a C=O on its ring.

91
Q

What is a structural difference between cytosine and thymine?

A

Thymine has two C=O’s and a methyl group on its ring.

92
Q

DNA structure: DNA in solution typically has a twisted ladder of ______ ______ _______ structure. The most abundant form of DNA is __ form which is an antiparallel, _____ handed double helix.

A

alpha double helical, B, right

93
Q

What is the first order of chromatin structure?

A

DNA wrapped around nucleosomes

94
Q

Nucleosomes package __ bp of DNA and are made up of __ histone proteins

A

200, 8

95
Q

Name the 5 families of histone proteins and their function.

A

H1, H2A, H2B, H3, H4
H1: linker, between beads
H2A, H2B, H3, H4: core histones, the beads

96
Q

What histone modifications cause inhibition of transcription (via condensation of the chromatin)?

A

Phosphorylation and methylation

97
Q

What histone modification causes activation of transcription?

A

Acetylation

98
Q

What residues are histone proteins highly enriched in?

A

Lysine (K) and arginine (R)

99
Q

Where do histone modifications usually occur?

A

On the amino terminal tails

100
Q

Histone modifying enzymes are recruited by ________ and ________.

A

transcription activator, repressor proteins

101
Q

Acetylation of histone proteins alter the ________ of core histones from ______ to ______, causing _______.

A

electrostatic charge, positive, neutral, DNA to repel from histones

102
Q

What happens when histone H3 is methylated?

A

HP1 binds and blocks transcription.

103
Q

_________ of all histones is necessary for chromatin to condense into mitotic chromosomes.

A

Phosphorylation

104
Q

Epigenetic modifications produce _______ changes in gene function without _________.

A

heritable, changing the DNA sequence

105
Q

Describe how DNA methylation can repress gene expression.

A

Cytosine in CpG sequences gets methylated, which then becomes bound to MBPs. MBPs recruit histone deacetylases which deacetylate histones so that DNA wraps more tightly around them, and histone methyltransferases which cause HP1 to bind and block transcription.

106
Q

How does DNMT inhibitor 5-azacytidine (Vidaza) work as an anti-cancer drug?

A

The drug incorporates into DNA in the place of cytosine so CpGs can’t be methylated, so tumor suppressor genes are hopefully re-activated.

107
Q

____ bp DNA (__ loops of ___ bp each) wrap around each histone octamer and ____ bp between each nucleosome.

A

140, 2, 70, 60

108
Q

How does a nucleosome form in a test tube?

A

2A and 2B form two dimers, and 3 and 4 form a tetramer. These components bind to each other and form four grooves for the two 70 bp loops of DNA to wrap around.

109
Q

Which histone proteins must be acetylated in euchromatin? Which histone protein must be absent?

A

H3 and H4 must be acetylated and H1 must be absent.

110
Q

Explain the role of histone acetyltransferases (HATs) in astrocyte differentiation.

A

Two growth factors bind to transcription factors which become activated and go into the nucleus. These transcription factors recruit a HAT, which acetylate the histones to neutralize their positive charge and loosens the DNA.

111
Q

What is the second order of chromatin structure?

A

Nucleosomes aggregating into solenoids or 30 nm fibers

112
Q

When seen in cross-section, solenoids have __ nucleosomes x ___ bp DNA = ___ bp per turn.

A

6, 200, 1200

113
Q

What role does histone H1 have in solenoid structure?

A

H1 is needed not only to bind linker DNA, but also to bind to DNA that is wrapped around the nucleosomes, in order to condense DNA and nucleosomes tightly together.

114
Q

What is the third order of chromatin structure?

A

50 solenoids form a 60,000 bp loop

115
Q

How does the structure of loops facilitate gene expression?

A

The bases of the loops are attached to the nuclear matrix which contains machinery for DNA replication & transcription. This also enables interaction of DNA domains between loops (e.g. enhancers with promoters), which also facilitates gene expression.

116
Q

What is the fourth order of chromatin structure?

A

The miniband

117
Q

How many bp of DNA are in one miniband?

A

If a miniband contains 18 loops (60,000 bp/loop), there are ~1 million bp DNA in each miniband.

118
Q

What is the fifth order of chromatin structure?

A

The chromosome

119
Q

How many minibands are in one chromosome? How many bp are there in one chromosome?

A

75, 75 million

120
Q

Describe the location and function of centromeres

A

Centromeres are loci of repetitive DNA in the middle of the chromosome. They serve as anchor points that control separation of
chromosomes when cells divide.

121
Q

Where are telomeres located and what is their function?

A

Telomeres are repeat sequences that cap chromosomal ends. They function in protection, replication and stabilization of chromosome ends. In normal cells, telomeres get shorter after each cell division. When they are short enough, the cells become senescent.

In germline cells, telomerase maintains chromosome length = immortality.

122
Q

How many chromosomes are in a human diploid cell, and approximately how many bp does one cell contain?

A

46 chromosomes, 75 million bp per chromosome, >3 billion bp

123
Q

Proteins <__ kDa can diffuse through the nuclear pore complex. Others must be transported via ___________.

A

40, active transport

124
Q

Describe the nuclear pore complex

A

There are three stacked octomeric rings of nucleoporin proteins.

125
Q

How are nuclear (cargo) proteins recognized by nucleoporins?

A

Via a nuclear location sequence (made up of basic lysine and arginine residues), which binds to an NLS receptor protein which escorts the cargo to the nuclear pore

126
Q

How does importin-αβ bring proteins into the nucleus?

A

1) Protein binds NLS receptor (importin-α)
2) Importin-β docks to nucleoporin
3) Protein is translocated into the pore
4) Protein is released by importin inside the nucleus

THIS IS A ENERGY DEPENDENT PROCESS– Ran-GTP!!

127
Q

_____ status of NLSs in nuclear cargo protein regulates entry of proteins into the nucleus.

A

Phosphorylation

128
Q

How does nuclear export work?

A

Exportin proteins bind nuclear export sequences (NES) that are rich in leucine and transport whatever is attached to the outside of the nucleus.

THIS IS ANOTHER RAN-GTP-DEPENDENT PROCESS!!

129
Q

Energy for nuclear import/export is provided by a _________ gradient, high in the ________, and low in the _______.

A

Ran-GTP, in the nucleus, the cytoplasm

130
Q

The nuclear lamina is found between the ___________ and ___________.

A

inner nuclear envelope, marginal heterochromatin

131
Q

Describe the function of nuclear lamin proteins.

A

There are three lamin proteins, A, B, and C. Lamin B is bound by an LBR (Lamin B Receptor), which is an integral membrane protein in the inner nuclear membrane. Lamins A and C interact with the marginal heterochromatin.

132
Q

What happens to the nuclear lamin proteins during mitosis?

A

Lamins A and C get hyperphosphorylated which makes the nuclear envelope completely soluble (there is no nuclear envelope in mitotic cells). Lamin B stays bound the LBRs, which mediates re-formationof the nuclear envelope at telophase.

133
Q

Nuclear lamins A and C are _____ of the _____ gene.

A

splice variant, LMNA

134
Q

The only function of the nucleolus is _________. They have no _________.

A

to make ribosomes, membrane

135
Q

There are __ ribosomal RNA genes (all identical) in the nucleoli of human cells. There are __ genes on each of the __ chromosomes that make up the nucleolus.

A

400, 40, 10

136
Q

The internal matrix confers _______________ of chromatin.

A

non-random order

137
Q

What are the three lines of evidence for non-random order in the Internal Nuclear Matrix?

A

a. Some matrix proteins are tissue-specific.
b. Chromosomes occupy specific locations.
c. The internal nuclear matrix organizes orderly replication & transcription of DNA.

138
Q

mRNA is transcribed in the ______.

A

nuclear matrix

139
Q

Sites of DNA replication–replicons–are in the _________.

A

nuclear matrix

140
Q

What kind of proteins are the nuclear lamin proteins?

A

Intermediate filament proteins.

141
Q

Selected cells from ten trillion cells of the human body proliferate once every __ years.

A

7

142
Q

What are the three stages of interphase?

A

G1
S: synthesis of chromatin (2N DNA -> 4N DNA; histone proteins are replicated)
G2: preparation for mitosis (M-phase), centrosome duplicates

143
Q

What phase of the cell cycle are senescent and differentiating cells in?

A

G0

144
Q

What variation of duration of the phases of the cell cycle exists across different cell types?

A

S, G2 & M ~ constant (12-24 hrs)
G1 is HIGHLY VARIABLE! (Could be forever if in G0!!)

Therefore, the variation of generation time across cell types is due to differences in lengths of G1.

145
Q

What are the delayed response factors of cell cycle regulation?

A

Cyclins and CDKs (which make up the cyclin-dependent protein kinase heterodimers)

146
Q

How do cyclins work?

A

Cyclins are the regulatory subunit of the cyclin-dependent protein kinase heterodimer and the content of a single type of cyclin increases at different points in the cell cycle (D–>E–>A–>B)

147
Q

What is the function of CDKs?

A

CDKs are the catalytic subunits of cyclin-dependent protein kinase heterodimers and they phosphorylate target proteins. Their content does not change during the cell cycle.

148
Q

What happens when the cell cycle is induced in cells either in G0 or G1?

A

1) p27, a CDK inhibitor, decreases and stays low
2) Cyclin D increases and stays high
3) Cyclins E, A, and B sequentially & transiently increase/peak

149
Q

When and what is the Restriction Point in the cell cycle?

A

At the interface between G1 and S (checkpoint #2). After passing this point, a cell must replicate all of its DNA. If not, the cell will die.

150
Q

What happens during the first checkpoint of the cell cycle?

A

1) a growth factor binds and activates C-myc, which activates transcription of the cyclin D gene
2) Cyclin D binds to CDK4 in cytoplasm
3) Cyclin D/CDK-4 phosphorylates Rb (a tumor suppressor gene that inhibits the cell cycle by binding E2F, a transcription factor)
4) P-Rb releases E2F
5) E2F goes into the nucleus and activates genes for cyclin E and A

151
Q

What happens in the second checkpoint of the cell cycle?

A

1) E2F activates the transcription of cyclin E
2) Cyclin E binds CDK2
3) Cyclin E/CDK2 complex goes back into nucleus and phosphorylates target proteins, leading to entry into S phase

152
Q

What cyclin/CDK complex sustains cells in the S phase? How is this accomplished?

A

Cyclin A/CDK 2. This complex phosphorylates protein in the nucleus that keep replication going.

153
Q

How does p53 work?

A

p53 induces the transcription of p21, which inhibits CDK2, preventing the cell cycle from moving forward.
p53 also activates apoptosis in cells that do not replicate their DNA correctly.

p53 is therefore a TUMOR SUPPRESSOR GENE.

154
Q

What happens at the fourth checkpoint of the cell cycle (G2/M interface)?

A

CDK1, attached to cyclin B, is DE-phosphorylated by cdc25, the heterodimer enters the nucleus, and activates genes involved in:

  • nuclear envelope breakdown
  • assembly of the mitotic spindle
  • arrest in mitotic phase
155
Q

Cancer is characterized by a decrease in cell _____, and an increase in cell _____.

A

differentiation, proliferation

156
Q

What is a proto-oncogene? Name the three families.

A

A proto-oncogene is a gene that when mutated, becomes over-activated –> abnormal cell cycle stimulation –> cancer.

1) cell membrane receptors for growth factors
2) cytoplasmic and transcription factors
3) cyclins/CDKs

157
Q

Name some tumor suppressor genes.

A

p21, p53, Rb, BRCA-1, BRCA-2

158
Q

An agent in cigarette smoke directly _________________ in lung cells.

A

mutates the p53 gene

159
Q

Explain the mitochondrial pathway for apoptosis.

A

1) Macrophages secrete TNF (tumor necrosis factor), which binds to TNF receptors
2) TNF receptor balances pro- and anti-apoptotic factors
3) pro-apoptotic factors cause mitochondira to leak cytochrome C into the cytoplasm
4) caspases are activated
5) chromatin is chewed up and cell dies

160
Q

Basic Properties of DNA replication:

  1. DNA synthesis is ________.
  2. DNA is synthesized in the _______ direction.
  3. A ______ and a ______ are required.
  4. DNA replication is ________.
A

semiconservative, 5’–>3’, 3’-OH primer, template, semi-discontinuous

161
Q

Explain how a DNA chain is elongated.

A

Elongation is catalyzed by DNA polymerase.
There is a 3’-OH primer (bound to the template strand) with a free hydroxyl group. This hydroxyl group attacks the alpha phosphate on the incoming dNTP.

162
Q

How does semidiscontinuous DNA replication work?

A

The lagging strand is synthesized by small Okazaki fragments. This is accomplished by short RNA primers (generated by primase) that provide free hydroxyl groups.

163
Q

Name the 3 catalytic activities of DNA polymerase I.

A

1) 5’ –> 3’ polymerase
2) 5’ –> 3’ exonuclease
3) 3’ –> 5’ exonuclease

164
Q

Which catalytic function of DNA Polymerase I is the proofreading mechanism?

A

3’ –> 5’ exonuclease

165
Q

Which DNA polymerase does the bulk of DNA synthesis?

A

DNA Polymerase III

166
Q

What are the major functions of DNA Polyermase I?

A

Extending Okazaki fragments, replacing RNA primers on lagging strand with DNA, repair

167
Q

What are telomeres?

A

Telomeres are tandem hexanucleotide sequences that are found at the ends of eukaryotic chromosomes.

168
Q

The lifespan of somatic cells is due in some part to what?

A

The length of their telomeres.

169
Q

What type of enzyme is a telomerase?

A

An RNA-dependent DNA polymerase.

170
Q

How does telomerase maintain the chromosome length?

A

Telomerase has an RNA component that is complementary to the telomeric ends. It binds to these ends and provides a template to allow the end to be extended.

171
Q

Why does a polymerase sometimes mistakingly put in the wrong nucleotide?

A

Because there are tautomeric forms of certain bases that can mis-basepair with the corresponding base in the growing strand (adenine can undergo tautomeric shift to imino form, which can base pair to cytosine, so you get an A-C basepair)

172
Q

When cytosine gets deaminated, what does it become?

A

Uracil

173
Q

Differences between heterochromatin and euchromatin:
Heterochromatin is more likely to contain _______ed DNA and histones. Euchromatin is more likely to contain ______ed histones.

A

methyl, acetylat

174
Q

Are actively transcribed regions of the genome devoid of nucleosomes?

A

No. Actively transcribed regions of the genome contain nucleosomes.

175
Q

What do some nitrous acid precursors do to DNA?

A

They are chemical mutagens; they can deaminate cytosine –> uracil, guanine –> X

176
Q

How do alkylating groups cause mutations in DNA?

A

When guanine is methylated, it becomes o-methyl guanine and binds to thymine instead of cytosine.

177
Q

In purine synthesis, what molecule is the first to have a completed purine ring?

A

IMP

178
Q

In purine synthesis, which step is the rate-limiting step?

A

PRPP to phosphoribosylamine, catalyzed by ATase.

179
Q

For purine de novo synthesis, what is/are the starting materials?

A

PRPP

180
Q

In de novo pyrimidine synthesis, what is/are the starting materials?

A

Glutamine, HCO3- (bicarbonate), 2 ATP

181
Q

What reaction does PRPP synthetase catalyze? What causes feedback regulation of this enzyme’s activity?

A

Ribose 5-phosphate to PRPP. AMP/ADP/ATP and GMP/GDP/GTP

182
Q

In purine synthesis, what allosterically regulates the enzyme catalyzing the rate-limiting step, and in what ways?

A

ATase is allosterically negatively regulated by end products (AMP/ADP/ATP and GMP/GDP/GTP), and positively regulated by PRPP, the substrate for ATase.

183
Q

What catalyzes the reaction: AMP + ATP <–> 2ADP

A

Adenylate kinase

184
Q

What catalyzes the reaction: GMP + ATP <–> GDP + ADP

A

Guanylate kinase

185
Q

What catalyzes the reaction: GDP + ATP <–> GTP + ADP

A

Nucleoside diphosphate kinase

186
Q

Under steady-state conditions, adenylate kinase is in equilibrium and maintains the various phosphorylated forms of adenine nucleotides at the following ratios:

___X ATP, ___X ADP, and ___X AMP

A

100, 10, 1

187
Q

XDH can be converted to XO under ________ conditions.

A

low oxygen

188
Q

Uric acid is the least _______ purine base

A

soluble

189
Q

Recycling a purine base requires ______.

A

PRPP

190
Q

Deficiency of _____ can lead to Lesch-Nyhan Syndrome

  1. The disease is ______ linked (affects ______).
  2. High levels of ________ is produced in association with _____.
  3. Inadequate feedback inhibition of _______ and _______ leads to increased rates of ___________.
A

HPRT, X chromosome linked, males, uric acid, gout, PRPP synthase, ATase, de novo purine synthesis

191
Q

In ischemic or anoxic heart: ________ is a potent coronary vasodilator and facilitate oxygen delivery back to the damaged tissue.

A

adenosine

192
Q

In vigorously exercising skeletal muscle: ____ facilitates net resynthesis of ATP following exercise.

A

IMP

193
Q

Primary gout (inherited metabolic abnormalities)

  1. Overexpression of __________
  2. Deficiency of ________
  3. Defects in a family of ________ proteins (80% of primary gout patients)
A

PPRP synthetase, HPRT, renal urate transport

194
Q

PRPP induces the ____________ and therefore activation of ATase

A

monomerization

195
Q

Allopurinal affects what parts of the purine synthesis pathway?

A

It inhibits ATase and XDH

196
Q

Xanthinuria is a deficiency of __________ and causes high levels of ________.

A

XDH, xanthine

197
Q

A deficiency of __________ is associated with a severe combined immunodeficiency (SCID) involving T-cell and B-cell dysfunction.

A deficiency of ____________ is associated with impairment of T-cell function.

A

adenosine deaminase, purine nucleoside phosphorylase

198
Q

CPS II is _________ and uses __________ as the source of the amino group,whereas CPS I is __________ and uses _______.

A

cytosolic, glutamine, mitochondrial, NH4+

199
Q

XDH uses ______ as electron acceptor in the oxidation reaction and XO uses _______ as the electron acceptor.

A

NAD+, O2

200
Q

Explain the glucose effect.

A

When glucose is low, cAMP goes up. cAMP binds to CAP/CRP protein, cAMP-CAP complex binds to promoter. IF there is lactose present, allolactase acts as an inducer, which binds to the repressor and makes it fall off, so then RNA polymerase binds and transcribes beta galactosidase from lac operon.

201
Q

Describe the helix-turn-helix motif

A

This motif mediates DNA binding of certain proteins. They bind as dimers and the helix interacts with the major groove of the DNA.

202
Q

What does the lac repressor binding site look like?

A

It is a palindrome, because the repressor binds as a dimer.

203
Q

What is the function of a zinc finger motif?

A

Mediates DNA binding and also serves as a protein-protein interface

204
Q

Describe the leucine zipper motif.

A

They are a dimer of amphipatic alpha helices that bring DNA binding domains (usually basic) of transcription factors together.

205
Q

Describe helix loop helix motifs

A

They are dimers of two amphipatic helices separated by an intervening loop, they are often heterodimers, and the helices are not responsible for DNA binding.

206
Q

ADP ribosylation is caused by what toxins?

A

Diptheria, cholera, and pertussis

207
Q

How does cholera toxin work?

A

It modifies a G protein that renders it permanently active in signaling adenylyl cyclas, resulting in high cAMP levels –> massive water loss in intestinal epithelial cells –> severe dehydration and electrolyte loss

208
Q

____ core particle of proteasome has proteolytic activity.

A

20S

209
Q

The linkage of ubiquitin is through an isopeptide bond between the _____ group of ______ residues of the target protein and the _______-terminal ________ residue of ubiquitin

A

ε-amino, lysine, carboxyl, glycine

210
Q

What is BRCA1?

A

It is a gene encoding an E3 ubiquitin ligase; heterozygous mutations are found in 10% of ovarian and breast cancers

211
Q

Why is it beneficial for a protein to undergo hydrophobic modifications? What are examples?

A

They promote the interactions with the lipid bilayer of the membrane; palmitoylation, myristoylation, farnesylation

212
Q

_______ of Ras is required for its _____________, its functioning in _____________, and is essential for the transforming activity of oncogenic variants of Ras.

A

farnesylation, association with the plasma membrane, signal transduction