Final Exam Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

What are some common causes of metabolic alkalosis?

A

Acute alkali administration, vomiting, gastric aspiration, diuretics

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

What are the functions of prostaglandins/thromboxanes?

A
  • inflammatory mediators (vasodilation) - stimulation of contraction of uterus - platelet aggregation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
1
Q

How is the PDH complex similar or dissimilar to the alpha-ketoglutarate DH complex?

A
  • Each complex has 3 enzyme subunits. Each produces CO2. Substrate for PDH is 3C alpha-keto acid (pyruvate), while substrate for aKGDH is 5C alpha-keto acid (alpha-ketoglutarate). Product for PDH is 2C (acetyl-CoA), while produce for aKGHD is 4C (succinyl-CoA). Regulation of PDH occurs via phosphorylation, while aKGDH is not regulated via phosphorylation. Reactions catalyzed by both are irreversible as a lot of free energy is lost.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
1
Q

What are the degradation products of Tryptophan?

A
  • Acetoacetyl-CoA - Acetyl-CoA - Fumarate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
1
Q

Discuss roles of the CREB, CBP and PKA in transcriptional control

A
  • PKA phosphorylates and regulates many cytosolic proteins, but is also present in activated form in nucleus under high levels of cAMP - Many genes regulated by PKA contain CRE (cAMP responsive element) in their promoter region - PKA influences transcription by phosphorylation of CREB, which then binds CRE - CREB bound to CRE recruits CBP - CBP then recruits EP300 - CBP/EP300 interacts physically with GTFs. Together CBP/EP300 function as HATs, relaxing chromatin and stimulating transcription
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
1
Q

Explain how the Philadelphia chromosome affect the health of a carrier

A
  • Philadelphia chromosome = translocation bw c/s 9 & 22- ABL tyrosine kinase is moved from 9 to BCR region on 22.- BCR-ABL protein functions as dominant oncogene causing CML
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How do you convert the rectangular hyperbolic V vs. [S] curve into a straight line using the Lineweaver-Burk (double-reciprocal) method. What are the X- and Y-intercepts and the slopes of this plot? How could the Vmax and Km be determined using this plot?

A
  • 1/v = (Km/[S]).(1/Vmax) + 1/Vmax (y = mx + b form) - The x intercept = -1/Km – the negative reciprocal gives you Km - The y-intercept = 1/Vmax – the reciprocal gives you Vmax - Slope = Km/Vmax
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Explain how F26BP affects gluconeogenesis. Include details about the enzymes involve, pathways activated and substrates affected.

A
  • Take home message: high concentrations of F26BP inhibit gluconeogenesis, low concentrations of F26BP stimulate gluconeogenesis - F26BP is produced by enzyme PFK2 (produces F26BP from F6P) - Insulin stimulates PFK2 via cAMP leading to increased concentration of F26BP, which causes inhibition of gluconeogenesis via pyruvate carboxylase, PEP carboxykinase and glucose-6-phosphatase - Glucagon inhibits PFK2 via cAMP leading to decreased concentration of F26BP, which causes stimulation of gluconeogenesis via PEP carboxykinase and glucose-6-phosphatase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is Li-Fraumeni syndrome?

A
  • Dominant heritable condition conferring increased susceptibility to many forms of cancer as a result of dominant negative effect of mutation to TP53 allele - Cells with damaged DNA are free to continue to divide and tumors develop
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe degradation of GMP vs degradation of AMP to their constituent parts

A
  • General theme is to remove phosphate, then ribose, leaving you with base a.) GMP: - GMP = Guanosine + Pi (ez: 5-nucleotidase) - Guanosine + Pi = Guanine + Ribose-1-P (ez: purine nucleoside phosphorylase) b.) AMP pathway 1: - AMP = IMP + NH4+ (ez: AMP deaminase) - IMP = Inosine + Pi (ez: 5-nucleotidase) - Insoine + Pi = hypoxanthine + Ribose-1-P (ez: purine nucleoside phosphorylase) c.) AMP pathway 2: - AMP = Adenosine + Pi (ez: 5-nucleotidase) - Adenosine = inosine + NH4+ (ez: adenosine deaminase) - Insoine + Pi = hypoxanthine + Ribose-1-P (ez: purine nucleoside phosphorylase)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Explain how epigenetic changes play a role in the development of cancer

A
  • Global hypomethylation of DNA outside of CpG islands is seen in most cancers- Hypomethylation causes genomic instability and therefore elevated transposon activity, resulting in chromosomal abnormalities. This causes cancer.- Hypermethylation of CpG islands in promoter can shut down tumor suppressor genes – sometimes seen in non-familial breast cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Explain how ethanol is metabolized? What is its impact on CHO metabolism?

A
  • Alcohol metabolism occurs in the liver by two mechanisms: 1.) two DH reactions (main pathway) and 2.) MEOS: microsomal ethanol oxidizing system: induced by chronic alcohol abuse. - Both DH reactions reduce NAD to NADH, which interferes with CHO metabolism and utilization. - When NADH is high in the cytoplasm, pyruvate and oxaloacetate are converted into lactate and malate respectively via lactate dehydrogenase and malate dehydrogenase respectively. Pyruvate and oxaloacetate are both intermediates in gluconeogenesis and glycolysis. - Lactate levels rise (more than normally) and cannot be processed in the Cori cycle, causing accumulation in blood, leading to metabolic acidosis. Body responds by increasing respiration in order to attempt restoral of acid/base balance. - Failure to complete Cori cycle also leads to epinephrine and glucagon release, which induces a stress response. - NADH inhibits key enzymes of TCA and shunts acetyl-CoA into FA/ketone body biosynthesis leading to a fatty liver over time.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Explain how nitroglycerin can reduce blood pressure.

A
  • Nitroglycerin is converted to NO in the body. When binding to guanylate cyclase in smooth muscle lining blood vessels, it causes them to relax and volume within the vasculature to expand. This causes a reduction in blood pressure.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Explain what reaction(s) the cell can perform when there is a need for NADPH and ATP. Ribose is not required.

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

How do high levels of NADH prevent gluconeogenesis from proceeding?

A
  • Malate-oxaloacetate shuttle. When levels of NADH are high, it will donate its electrons to oxaloacetate to form malate, which moves into mitochondria and reconverts to oxaloacetate and NADH in the matrix. This ensures low oxaloacetate in the cytosol and inhibition of gluconeogenesis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the committed step in TAG synthesis? What are TAG synthesis precursors used for?

A
  • Committed step is conversion of phosphatidic acid to 1,2 diacylglycerol via phosphatidate phosphatase. - Phosphatidic acid can be shunted into lipid membrane synthesis pathways instead of going down TAG synthesis pathway.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the function of lipoxygenase inhibitors? What downstream molecule is being targeted with these inhibitors? What disorder/illness can these alleviate? How?

A
  • These target synthesis of leukotrienes from AA via inhibition of the lipoxygenase enzyme. - Alleviates asthma by reducing smooth muscle contraction.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Which AAs can be used to synthesize succinyl-CoA? How?

A
  • Mnemonic: VOMIT - Valine, odd-chain fatty acids, methionine, isoleucine, threonine - These are converted into propionyl-CoA, which undergoes intermediates to become succinyl-CoA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the mechanism of action of tamoxifen?

A
  • Tamoxifen inhibits estrogen action - It is a competitive inhibitor, blocking estrogen binding to its receptor, however ERE dimer still remains dimerized and bound to DNA - Growth of breast cancer is reduced by disrupting this estrogen signaling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Examples of exogenous uncouplers to ETC/ox-phos. Symptoms? Treatment?

A
  • DNP (2,4 dinitrophenol) is a pesticide and poison. It causes sweating, flushing, nausea, inc RR, tachycardia, fever, coma, death in 1-2 days. Treatment with ice baths, oxygen and fluid/electrolyte replacement. - Aspirin in very high doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Explain the biochemistry of CO (carbon monoxide poisoning).

A
  • CO binds to heme iron at ~ 240 higher affinity than o2 (known as carboxyhemoglobin) - When 2 x CO molecules bind to Hb, R-form stabilizing, meaning T-form unfavored and o2 wants to remain bound and not deliver to tissue. - Treatment is removing individual from CO and administering 100% o2 at high pressure.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Explain how lipids are transported into the mitochondria. Is it in the form of TAGs or FAs?

A
  • FAs are transported into the mitochondria.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Discuss regulation of FA (TAG) synthesis, degradation and mobilization/release

A
  1. ) FA synthesis
    - Well-fed state:
    a. ) insulin: induces expression of acetyl-CoA carboxylase, FA synthase, malic enzyme and G6PD long-term basis
    b. ) insulin: activates acetyl-CoA carboxylase and glycolysis (both via phosphoprotein phosphatase, which removes phosphates) – short term basis
    c. ) citrate: activates FA synthase
    - Starvation
    a. ) absence of insulin: levels of acetyl-CoA carboxylase, FA synthase, malic enzyme and G6PD fall
    b. ) glucagon: caused cAMP and PKA activity to rise.
    c. ) PKA: inactivates acetyl-CoA carboxylase by phosphorylation, inhibits glycolysis
  2. ) FA degradation
    - Limiting step = bringing FA into mitochondria via carnitine shuttle / CPTI/II. This is inhibited by malonyl-CoA in well-fed state when FA synthesis is occurring.
  3. ) FA mobilization
    - High glucagon/low insulin stimulates mobilization: In fasting state, glucagon raises cAMP levels and activates PKA. PKA activates perilipin and hormone sensitive lipase both by phosphorylation (remember, glucagon acts via phosphorylation). This results in FAs being clipped off glycerols allowing them to become FFAs diffuse out of adipose tissue and move to other tissues. In other tissues they move into mitochondria via CPT I shuttle mechanism which are active as malonyl-CoA is low (this should only be occurring in starved state) and proceed through beta-oxidation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Describe orotic aciduria

A
  • Rare hereditary condition resulting in UMP synthase mutation - Crystalluria, hypochromic megaloblastic anemia, growth retardation, neurologic abnormalities - Anemia is unresponsive to B12 and folic acid - Treatment with uridine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Common alpha-ketoacid/aminoacid pairs in reactions with alpha-ketoglutarate/glutamate?

A
  • pyruvate/alanine - oxaloacetate/aspartate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Explain patterning. Discuss role of signaling molecules that are necessary for this to occur

A
  • Following specification of axes, patterning takes place. Patterning defines which end of undevelopmed mass of cells is head, which is tail. It segments cells to define which parts become head, thorax, abdomen etc.- Patterning along ant/post axis is determined by homeobox (HOX) genes, which are a family of transcription factors containing a special DNA binding domain called homeodomain. There are 4 HOX clusters on 4 chromosomes- Expression of each of the genes belonging to a cluster correlates with position of respective cell in embryo and with timing of expression. Each cell along axis experiences a different ratio of expression of different HOX genes. Ultimately, the pattern of HOX gene expression determines fate of a cell in segment.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are modifier genes?

A
  • The individual genetic background modifies the phenotype
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Regarding the molecular structure of collagen, describe: a. the importance of amino acid composition and sequence. b. unusual amino acids and their molecular function. c. the importance of vitamin-C and iron in collagen formation.

A

a. the importance of amino acid composition and sequence. - Collage contains 35% glycine, 11% alanine and modified amino acids known as 4-hydroxyproline and 5-hydroxylysine (previously proline and lysine, but post-translationally modified). - Collage contains the repeating sequence Gly-Pro-Hydroxyproline. Since three polypeptides (in helices) are to come together, it would be important for a smaller residue to be present at the contact points. This is the case, glycine is found here, which has R group of H. b. unusual amino acids and their molecular function. - Collagen has two unusual/modified amino acids known as 4-hydroxyproline and 5-hydroxylysine. Without proline-peptide and peptide-hydroxyproline bonds, helix and triple helix formation would not be possible. Also, strength and rigidity of the molecule would also be compromised. Therefore functional would be lost. Stability is also present in collage as a result of hydrogen bonds formed by hydroxyproline. Lastly, hydroxylysine allows for covalent linkages between helices to form via reactive aldehyde functional groups. c. the importance of vitamin-C and iron in collagen formation. - Ascorbate (vit C) is necessary to catalyze the reaction of proline to hydroxyproline via the prolyl hydroxylase enzyme. This occurs in conjunction with Fe2+ and alpha-ketoglutarate. Ascorbate is required to reduce Fe3+ to Fe2+ in the reaction and allow the enzyme to remain active. Without vit C, enzyme is inactivated and scurvy develops. - Remember hydroxylation in collage allows for normal collagen fibril formation. Without, you have collagen that lacks strength, rigidity and stability.

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

Effect of lead on heme synthesis

A
  • Lead inhibits porphobilinogen synthase, causing accumulation of ALA - Lead inhibits ferrochelatase, causing accumulation of protoporphyrinogen IX - Causes symptoms similar to the porphyrias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How is the pentose phosphate pathway regulated?

A
  • Glucose-6-Phosphate Dehydrogenase is inhibited by NAPDH. When NADPH is high, G6P can be used elsewhere.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Explain what a competitive inhibitor is?

A
  • A competitive inhibitor is a reversible inhibitor. These compete with substrate to bind at the active site on the enzyme. Increasing substrate concentration will prevent inhibitor binding. Infinite concentration of substrate will abolish inhibition. Inhibitor once removed will not affect enzyme functionality. - These are the most common type of drug
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Achondroplasia. Inheritance, defect, frequency

A
  • AD- Caused by defect in FGFR3, gain of function mutation where receptor is constitutively active, exhibits dominant negative effect. Most frequent form of dwarfirsm. Initiation of bone growth leads to short stature. New mutations occur frequently in this disease.- Mutation hotspot
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Explain the tumor progenitor cell model

A
  • Tumor progenitor cells arise during development due to epigenetic changes and the action of tumor progenitor genes (TPG)- Then follows a Gatekeeper Mutation (GKM) in a tumor suppressor gene (TSM) or oncogene (ONC) to generate a benign tumor- Finally, epigenetic and genetic plasticity help evolve the benign growth into a metastatic, invasive and drug resistant tumor- Crux of matter: errors in epigenetic programming of stem cells in development lays seed(s) for cancer later in life
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Describe the role of HLA haplotypes in the development of type 1 diabetes and other autoimmune disorders

A
  • Think of haplotypes as blocks of genetic information that are inherited together. HLA haplotypes are expressed in a codominant manner. Each parent only transmits one haplotype to the child. Certain HLA haplotypes favor or protect against diseases.- T1D: Contribution of HLA haplotypes to risk for T1D is strong, but not exclusive – only accounting for ~40% of genetic risk. Genetic variation in DR-DQ haplotypes affects risk for T1D. Some DR-DQ haplotypes increases risk and are known as susceptibility alleles. Others decrease risk and are designated protective alleles- Other immune disorders: HLA-B haplotypes determine risk for spondyloarthropathy. HLA-C haplotypes predict risk for psoriatic arthritis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is heterozygote advantage? Describe in respect to mutation affecting CFTR, beta-globin and HFE.

A
  • Heterozygote advantage refers to positive selection of heterozygotes as a result of their genotype conferring increased fitness in a particular environment.- CFTR: protects against typhoid fever (population that benefits = European)- Beta-globin/sickle cell: protects against malaria (population that benefits = Mediterranean and African)- Beta-globin/beta-thalassemia: protects against malaria (population that benefits = Meditteranean)- HFE/hemochromatosis: protects against plague (population that benefits = European)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What types of acid-base imbalances are caused by changes in level of CO2?

A

Respiratory acidosis is caused by higher levels of CO2 resulting in a decrease in blood pH. Respiratory alkalosis is caused by lower levels of CO2 resulting in an increase in blood pH.

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

Discuss rate of DNA transcription in context of basal transcription complex

A
  • Basal transcription complex = RNA pol II with GTFs - This complex initiates transcription at low rates - High rate requires NF1 binding to CAAT box and SP-1 binding to GC-rich sequences
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Define tumor suppressor. How does mutation in these genes contribute to development of cancer?

A
  • Tumor suppressor: inhibit cell growth - Loss of function relieves growth inhibition = non-functioning brake pedal, typically recessive mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Explain how to the Robertsonian translocations affect the health of a carrier

A
  • Robertsonion translocation is movement of long and short arms of two chromosomes. Result = one chromosome composed of both the long arms the other composed of both of the short arms. The shorter derivative chromosome does not contain essential genetic information and is typically lost during cell division. Common Robertsonion translocation involves c/s 13 q and 14 q – happens at frequency of 1/1300.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Explain what occurs to the Hb binding curve as pH, 2,3-BPG, temp, CO2 change? Draw it.

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

Which AAs can be used to synthesize Oxaloacetate?

A
  • Every AAs used to generate pyruvate, then pyruvate into OAA via pyruvate carboxylase - In addition, aspartate and asparagine – Ox with a big ASP (ass)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Cofactor requirement(s) for aminotransferases.

A
  • Vitamin B6 (pyridoxal 5’-phosphate)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How do you determine if R-group on amino acid will be in protonated, deprotonated form and will act as proton donor, acceptor?

A
  • If pH < pKa : acid form predominates : will act as proton donor
  • If pH > pKa: basic form predominates : will act as proton acceptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Action of tetracycline

A
  • 70 S ribosome inhibitor - Specifically blocks A site and prevents tRNA binding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

The formation of native protein structure is said to be cooperative. What does this mean? Outline the ‘seeding and nucleation’ model for the progression of prion diseases, Parkinson disease, and Alzheimer disease. What proteins are associated with these disease states?

A

Cooperative native protein structure indicates that the protein folding cannot be a random process, but instead occurs through favored pathways. This involves the rapid formation of short stable segments of secondary structure, which generate structural motifs which continue to fold into a thermodynamically stable protein structure. The seeding/nucleation model is where a misshaped protein (typically beta-sheets) induces other proteins (during folding) to change confirmation, which has a trickle down/domino effect on other proteins. Parkinson’s: alpha-synuclein is implicated, affect NT release (?) Alzheimer’s: tau and amyloid-beta are implicated, affecting microtubule (?) and neuronal plasticity (?) respectively

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

How can epigenetic changes contribute to the development of cancers? Example

A
  • Increased methylation silences transcription of a gene, reducing tumor suppressor expression - Decreased methylation activates transcription of a gene, increasing production of oncoprotein - Example: INK4 promoter methylation suppresses transcription, preventing p16ink4 synthesis, allowing for continued phosphorylation of Rb by cyclin-D:CDK4/6 and cell proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are disorders that prevent proper use of fructose? Explain each

A
  • Essential fructosuria: absence of fructokinase (normally in liver), prevents uptake of fructose. Benign. Fructose can still be used by muscle or become excreted. - Hereditary fructose intolerance: deficiency in aldolase B enzyme. F1P accumulates in liver causing depletion of liver phosphate pools, preventing liver from breaking down glycogen, causing liver to be damaged by accumulation of glycogen, lack of Pi and synthesis of ATP. Pts are required to avoid fructose and sucrose (disaccharide of fructose and glucose)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is galactosemia?

A
  • There are 3 kinds depending on the enzyme deficiency: Galact-1-P uridyl transferase, galactokinase or UDP-galactose epimerase. - Results in accumulation of galactose1-phosphate in liver and other tissues (CNS, kidney). Newborns present with milk intolerance and signs of liver failure, cataracts and intellectual disability. Also present with jaundice, lethargy and hepatomegaly. Diagnosis is by detection of galactose or galactose-phosphate in urine. Pts must receive a galactose-free diet (no lactose either, which is a glucose, galactose disaccharide).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Name two main sources for mutations affecting chromosome structure

A

1.) Nonhomologous End Joining during double-strand break repair2.) Unbalanced recombination between non-homologous sequences

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

Define gain of function.

A
  • Gain of function: Mutated protein may have functions different from its wildtype variant. In this case, the few proteins with novel functions will have an effect no matter how many wildtype versions are present. This mechanism is frequently observed in signal transduction proteins.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is HIF-1alphabeta and how is it regulated? What is the importance of HIF-1alphabeta in the growth of tumors?

A
  • In normoxic environment, HIF-1alpha (hypoxia-inducible-factor) is hydroxylated by proline hydroxylase, leading to accumulation and destruction of itself - In hypoxic environment, HIF-1alpha associates with HIF-1beta, which regulates expression of VEGF and many other genes, allowing for angiogenesis to proceed. - Result = new blood supply delivers nutrients/oxygen, removes waste to/from tumors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Describe/draw the reactions of glycogen formation (glycogenesis)

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

What is Burkitt’s Lymphoma? Describe biochemistry that causes it.

A
  • Translocation event moves c-myc gene from normal position on c/s #8 over to c/s #14 - Result = myc gene is now near control elements of antibody HC and continuous production of high levels of Myc occurs in B-cells. B-cells become cancerous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are population bottlenecks and founder effect and how does it affect the gene pool of a population? Example

A
  • If a large part of a population is wiped out by a catastrophic event, population has to recover from a small founder population of survivors. This bottleneck leads to amplification of rare alleles in the founder’s genotype. - Example: Samuel King immigrated to Eastern PA and founded a large family. He carried a rare recessive mutation in EVC, which cause Ellis Van Creveld syndrome (skeletal dysplasia disorder). This was propagated through founder effect. Consanguineous matings were inevitable in this genetically isolated community and children homozygous for EVC mutation were born. Carrier frequency for EVC is 12.3% in Old Order Amish community in PA, which is only 0.8% in general population
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Why are non-competitive inhibitors potentially better drugs than competitive inhibitors? What is the difficulty in manufacturing them?

A
  • Non-competitive inhibitors would perhaps make better drugs since they can inhibit enzyme activity irrespective of the substrate concentration. Would not have to worry about dosing based on substrate concentration. They are difficult to manufacture from the design perspective. Since non-competitive inhibitors bind in a location other than the active site, yet upon binding cause active site to be unable to bind substrate, how can you determine where this other site is?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Synthesis of alanine

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

From what are Ala and Ser made from?

A
  • Pyruvate and 3-PG
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What GTFs are required by RNA Pol II?

A
  • TFIID binds to TATA box, which distorts the DNA helix, acts as signpost and recruits remainder of factors and pol II - Transcriptional initiation complex forms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the Pierre Robin Sequence?

A
  • Collagen disorder, generalized growth retardation, neurogenic hypotonia, oligohydramnios leads to ….- Mandibular hypoplasia leads to ….- U-shaped cleft palate and micrognathia (small jaw/chin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Describe the clinical importance of LDH isozyme profiles.

A
  • There are 5 isozymes of LDH and they are distributed throughout the body in different abundances. - The normal LDH isozyme profile looks at relative abundances of each LDH isozyme and indicates that LDH2 is highest. - After an MI; however, this profile changes and LDH1 (found mostly in the heart and RBCs) becomes elevated. This used to be an important diagnostic tool to determine heart attacks. If LDH5 is evelated, it is diagnostic for acute hepatitis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is Crigler-Najjar/Gilbert syndrome? Symptoms.

A
  • Glucuronic acid is a prerequisite for bilirubin excretion. - UGT, which is an enzyme that catalyzes the conjugation of bilirubin with glucuronic acid leads to buildup of bilirubin and syndromes known as Crigler-Najjar/Gilbert. - Jaundice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Discuss checkpoints of cell

A
  • G1/S = restriction point – checking if conditions are favorable for division - G2/M = has DNA been replicated and are conditions still favorable for division - Metaphase/anaphase transition point = are c/s all attached to mitotic spindles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the target of the drug trastuzumab (Herceptin) used in the treatment of some breast cancers?

A
  • Binds to EC domain of Her2/Neu and is important inhibitor of tumor growth. - Mechanism? Inhibition of receptor dimerization and activation, induction of apoptosis, stimulation of antibody or complement-mediated cytotoxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Types of phospholipases and their functions?

A
  • Phospholipase A1: removal of FAs from C1 of phospholipid - Phospholipase A2: removal of FAs from C2 of phospholipid, usually arachidonic acid - Phospholipase C: removal of phosphate head group from C3 of PIP2 phospholipid creating DAG and IP3
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What pyrimidine bases cannot be salvaged?

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

What enzyme catalyzes the committed step of purine synthesis and what is its product?

A
  • Formation of PRA is committed step of purine synthesis - PRPP + gln = PRA + glu (ez: amidophosphoribosyltransferase)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Draw a substrate saturation curve and indicate Vmax, Km, Vmax/2.

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

List a few mucopolysaccharidoses. Describe defect and symptoms. Genetics?

A
  • Hunter’s: defect in iduronate sulfatase, accumulation of dermatan sulfate and heparan sulfate. Symptoms = skeletal abnormalities, mental retardation. X-linked recessive. - Hurler-Scheie: defect in alpha-iduronase, accumulation of dermatan sulfate and heparan sulfate. Symptoms = skeletal abnormalities, mental retardation. Autosomal recessive. - Sanfilippo’s: defect of heparan sulfate degradation. Symptoms = mild physical defects, severe mental retardation. Autosomal recessive.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

The blood concentration of alanine and glutamine are higher than the concentration of other AAs. How do these AAs help rid the body of toxic ammonia?

A
  • Alanine and glutamine are released in greatest quantity from muscle - BCAAs transfer their nitrogens to alpha-ketoacids (forming alanine and glutamine) and they travel to the liver where urea is formed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is acute pancreatitis? Symptoms? Causes? Triggers? Treatment?

A
  • Inflammatory disease of pancreas caused by premature activation of pancreatic digestive zymogens - Symptoms = abdominal pain, vomiting - Causes = pancreatic necrosis - Triggers = alcohol, infections, gallstones - Treatment = supportive via analgesics, fasting, elemental jejunal tube or TPN
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Regulation of ribonucleotide reductase

A
  • Allosteric: NTPs and dNTPs activate and inhibit (dATP inhibits) - Transcriptional
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is phenylketonuria. Causes? Pathology? Symptoms? Treatment? Why avoid aspartame if phenylketonuric?

A
  • Mutation in phenylalanine hydroxylase (more common) or DHBtn reductase leads to a elevation of phenylpyruvate, phenylalanine and other metabolites. - Pathology: not fully understood, but: accumulation of phenylalanine competitively inhibits the transport of other AAs across the BB barrier, which might interfere with NT synthesis. Also leads to reduced synthesis and increased degradation of myeline. Phe is also a competitive inhibitor of tyrosinase and intereferes with melanin synthesis - Symptoms: intellectual disability, recurrent seizures, hypopigmentation, eczematous skin rashes - Treatment: limiting dietary Phe (not eliminating, as it is essential), supplementation with tyrosine, treatment before child is 3 weeks of age, lifelong monitoring of plasma phenylalanine - Aspartame is a dipeptide of aspartic acid and methylated Phe derivative. Eating will cause formation of phenylalanine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How can one experimentally distinguish between noncompetitive and irreversible inhibition?

A
  • removal of irreversible inhibitor does not restore enzyme activity; however, removal of non-competitive inhibitor will restore enzyme activity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is problematic about unsaturated and branched FAs in beta-oxidation? How are these handled?

A
  • Unsaturated FAs: dbl bonds have to be moved around so enzymes used in beta-oxidation can recognize molecules and process them. a.) If dbl bond bw C3,4, isomerizes to trans-delta2-enoyl CoA b.) If dbl bond bw C4,5, reduced to trans-delta3-enoyl CoA, then isomerized to trans-delta2-enoyl CoA - Branched FAs: result from degradation of chlorophylls a.) requires peroxisomal alpha-oxidation, which starts with hydroxylation and ends with release of CO2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How do problems in gluconeogenesis cause fasting hypoglycemia and metabolic acidosis?

A
  • Pyruvate can be oxidized to acetyl-CoA and moved into TCA or can be oxidized to lactate by lactate dehydrogenase. - When issues occur with gluconeogenic enzymes, much pyruvate can be oxidized to lactate as opposed to taking gluconeogenic pathways, leading to high levels of serum lactate and metabolic acidosis. - For example, when pyruvate carboxylase functions correctly, pyruvate is converted to oxaloacetate and is prevented from forming lactate and is now dedicate to moving through the gluconeogenesis pathway.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What molecules (besides ketogenic/glucogenic) are derived from tyrosine?

A
  • Catecholamines (dopamine, NE, epi) - Thyroid hormones – T3/4 - Melanin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How is ATM activated? Implication

A
  • ATM is activated by replication forks: when RFs are present, DNA replication is present and cell-cycle needs to be halted until this completes - ATM also activated by double-stranded breaks
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is myasthenia gravis and cystic fibrosis. Name the defective ion channels in each.

A
  • Myasthenia gravis: autoimmune disease characterized by muscle weakness and fatigue to the muscles involved with talking, swallowing, facial expression, chewing and eyelid movement. It is the result of self-antibodies targeting nicotinic ACh receptors at the NMJ - Cystic fibrosis: genetic disorder (pop in Caucasians) characterized by multi-organ disease, main manifestation is the accumulation of thick mucous secretions in lungs. Leading to recurrent infections. It is the result of a mutation to a chloride channel, leading to defects in fluid and electrolyte transport.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

How do serum lactic acidoses occur as a result of defects to complexes I-IV, administration of DNP, administration of cyanide and lack of oxygen?

A
  • Defects in complexes I-IV would result in “back up” of intermediate substrates / electron carrier molecules, including NADH. This would result in lack of NAD+, which is limited in supply. End result is that pyruvate builds up in the cell and is reduced to lactate via LDH enzyme. Lactate is an acid and therefore a lactic acidosis is occurring here. - DNP, cyanide and lack of oxygen cause the exact same thing to occur.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

List endogenous uncoupling proteins. What are their functions?

A
  • UCP1-4. These are found in brown fat, skeletal muscle, brain and other parts of the body and function to increase body temperature.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the two hit model? Describe this model in relation to the development of retinoblastoma. What is the normal function of Rb?

A
  • Tumor suppressor genes inhibit proliferation - Damage to both alleles must occur to allow unregulated proliferation as the genes are recessive - This is why it is called two hit model - Individuals inheriting one mutant allele of tumor suppressor gene are at increased risk for certain cancers. If somatic mutation in second allele, tumor growth will occur Retinoblastoma mutation: - Rb protein is phosphorylated by cyclin D-CDK4/6 or cyclin E-CDK2. When phosphorylated, drives cell cycle. - Inherited form: one mutant allele from parent, somatic mutation of second allele in retinal cell leads to retinoblastoma. Tumor tends to develop in infancy - Sporadic form: two separate somatic mutations in a retinal cell or its progeny produces retinal cell in which both copies of functional Rb are lost - Symptoms: Leukocoria = white pupil, represents calcified intraorbital masses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Describe biochemistry behind HPV-induced cervical cancer

A
  • HPV virus produces E6 and E7 proteins that target p53 and Rb respectively leading to ubiqutination
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is Wernicke-Korsakoff syndrome? What are the symptoms?

A
  • It is an advanced thiamin (B1) deficiency that typically occurs in alcoholics. Symptoms include mental confusion, ataxia and loss of eye coordination.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

How can oxaloacetate initiate gluconeogenesis?

A
  • Malate-oxaloacetate shuttle. Oxaloacetate can be moved from the mitochondria to the cytoplasm via this shuttle and gluconeogenesis can occur.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

LHON

A
  • Most prevalent mitochondrial disorder. Rare at 1/50000- Caused by mutation by ND1 gene (oxphos)- LHON leads to a rapid deterioration of the optic nerve- Exhibits heteroplasmy as mitochondrial disorder
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Describe the properties of HbS, the variant of hemoglobin found in sickle cell anemia.

A
  • HbS is an altered Hb structure where a glutamate residue in the beta chain has been substituted for valine. Glu is a acidic hydrophilic AA, which valine is a non-polar, aliphatic AA. As a result, a “sticky hydrophobic pathy” is generated in the betal chains. When in the R-form, it is shielded from water. When in the T-form; however, it is exposed to surface. When in this form, it interacts with other Hb molecules to avoid interaction in polar environment and polymerization of poly Hb is catalyzed. This distores the shape of RBCs. Cells block blood vessels and damage organs.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Why aren’t ketone bodies used in liver or kidney?

A
  • These tissues don’t express acetoacetate:succinyl-CoA transferase to generate the intermediate: acetoacetyl-CoA that generates acetyl-CoA.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the products of pyrimidine degradation? How are these secreted?

A
  • Beta-alanine and beta-aminoisobutyrate - Water soluble and eliminated in urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Explain the effect of hypomethylation on genome stability

A
  • Hypomethylation reactivates transposable elements (jumping genes) that lead to somatic recombination and genomic instability.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is Hartnup disease? Symptoms? Treatment?

A
  • Rare genetic disorder affecting transporter of large, neutral amino acid resulting in no absorption of these amino acids across intestinal epithelial cells, also in prox tubule of kidney causing elimination issue - Symptoms? Similar to pellagra, which is niacin deficiency – 4 D’s – diarrhea, dermatitis, dementia, death. - Treatment? Dietary supplementation with niacin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe the progression of ribosome assembly

A

1.) GTP binds eIF2a 2.) GTP:eIF2a becomes bound to Met-tRNA to form ternary complex 3.) 40S:eIF3 binds ternary complex (with eIF1 and eIF1alpha) 4.) mRNA now binds small subunit and pre-initiation complex is formed (with aid of eIF-4a, eIF-4b, eIF-4f, eIF-5 and PAB) 5.) eIF-5b:GTP are added to this complex displacing hydrolyzed GDP:eIF2a and 60 S subunit is recruited and positioned with met-tRNA in P site. Elongation can now ensue

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

What do protons bind to on Hb?

A

-His. Causes formation of salt bridge, which stabilizes T-form.

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

What is allele heterogeneity?

A
  • Different mutations in the same gene cause different phenotypes. Mutations can be gain of function or loss of function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What molecule does FA synthesis begin with? What are the second, third… molecules used?

A
  • FA synthesis begins with acetyl-CoA and then joins with malonyl CoA during round 1 - Round 2-X begins with malonyl-CoA.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Why do T1 diabetic pts often present with DKA whereas type 2 don’t?

A
  • Function of insulin in addition to glucose metabolism is to repress ketone body production and inhibit lipolysis. - In absence as is case with T1 diabetics, large amounts of FFAs are released, ketogenesis ensues and ketoacidosis results. - Lipolysis is seen in T2 diabetics, but not as much as insulin is still available in large amounts and does have somewhat of an effect of repressing lipolysis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the two causes of megaloblastic anemia?

A
  • B12 deficiency - Folate deficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Describe problems associated with refeeding and causes of refeeding syndrome. Consider what starvation leads to? How to refeed?

A
  • Starvation: 1.) leads to degradation of digestive enzymes, 2.) depletes intracellular phosphate pools and 3.) depletes potassium stores - Problems: 1.) digestion is impaired and feeding leads to diarrhea and dehydration, 2.) glycolysis starting soaks up phosphate from serum leading to life-threatening hypophosphatemia and 3.) sudden insulin release (insulin moves K into cells) lowers serum potassium further - Watch electrolytes (esp. phosphate and K)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

List the enzymes involved in glycogen synthesis. Explain how each is regulated. What are the factors that stimulate and inhibit each.

A
  • Hexokinase: +: insulin; -: G6P - Glucokinase: +: insulin - Phosphoglucomutase: not regulated - UDP-glucose pyrophosphorylase: not regulated - Glycogen synthase: +: insulin; -: glucagon, epinephrine - Branching enzyme: not regulated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

In what cells does insulin have a function? What response?

A
  • Muscle: uptake of glucose, production of glycogen - Liver: production of glycogen, production of TAG - Adipose: uptake of glucose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is Gly made from?

A
  • From serine, which is made from 3-PG
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Names of 70S ribosome inhibitors

A
  • Streptomycin - Neomycin - Gentamicin - Tetracycline - Chloramphenicol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is imprinting? Explain the time and effects of imprinting

A
  • Imprinting is a form of DNA silencing that marks a chromosome as having come from the paternal or maternal parent. Why care? Transcriptional activities of paternal and maternal chromosomes are different.- Imprinting takes place during gametogenesis. Chromosomal regions are silenced by DNA methylation and histone deacetylation. This persists throughout the life of the individual.- Parent-of-origin imprint is erased and rewritten during gametogenesis. Female will reprogram both her chromosomes to make them look like maternal chromosomes. Male reprograms both his chromosomes to make them look like paternal chromosomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What enzyme(s) require(s) thiamin (B1)?

A
  • Alpha-ketoglutarate dehydrogenase complex and PDH complex.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Li-Fraumeni

A
  • XR- Defects in p53 causes brain tumors and leukemias- Two-hit model
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What enzyme is defective in tyrosinemia-I? Symptoms? Treatment

A
  • fumarylacetoacetate hydrolase - symptoms: severe condition affecting liver, kidney and peripheral nerves, fatal at young age if untreated (liver failure) - Dietary management and nitisinone (inhibits coversion of p-hydroxyphenylpyruvate to homogentisate)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are two major classes and subclasses of drugs?

A

1.) Reversible inhibitors: removal of inhibitor fully restores enzyme activity a.) competitive: compete with and bind at substrate’s active site b.) non-competitive: binds at site other than active site forming inactive enzyme 2.) Irreversible inhibitors: removal of inhibitor does not restore enzyme activity

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

What is neurofibromatosis I? What is normal function of the protein?

A
  • Symptoms/signs: café au lait spots, multiple non-malignant peripheral nerve tumors (neurofibromas) and iris Lisch nodules - Result of loss of function mutation in NF1 gene (recessive) that encodes neurofibromin that normally accelerates rate of GTP hydrolysis by Ras, causing it to be in inactive form - Lack of NF1 gene function prolongs Ras signaling increases proliferation through MAP kinase pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Outline the changes that must occur before normal cells of the colon can develop into an adenoma and then a carcinoma.

A
  • FAP pts inherit one defective allele from parent - Inactivation/loss of second allele occurs = - Proto-oncogene mutation, typically in Ras and loss of tumor suppressor genes typically TP53 causes adenomas - Additional mutations, gross c/smal changes, increased telomerase leads to carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Glycogen storage disorders

A
  • AR- Hypoglycemia, accumulation of glycogen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the relationship between the magnitude of Km and enzyme/substrate affinity?

A
  • inverse relationship - low Km = high S-E affinity - high Km = low S-E affinity - means some enzyme-substrates interact better than others
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Draw function of insulin in regulation of FA (TAG) synthesis.

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

Name the five most frequent birth defects. Give frequencies

A

1.) Heart defects – 1/100-2002.) Pyloric stenosis – 1/3003.) Neural tube defects – 1/10004.) Orofacial clefts – 1/700-10005.) Clubfoot – 1/1000

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

Retinoblastoma

A
  • Autosomal dominant inheritance, expression at cellular level is autosomal recessive- Defects in Rb protein, leads to predisposition and / or development of cancer- Two-hit model
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Know some examples of α-neurotoxins and how these compounds exert their effects.

A
  • alpha-conotoxin: produced by cone shell - alpha-cobratoxin: produced by cobra - alpha-bungarotoxin: produced by krait - tubocurarine: produced by plant These cause paralysis by blocking nicotinic ACh receptor on muscle.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What AAs are made from pyruvate and 3-PG?

A
  • Ala, Ser
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Why is the formation of cAMP and irreversible reaction?

A
  • Generation of cAMP from ATP via adenylate cyclase produces pyrophosphate (PPi). PPi is hydrolyzed into 2xPi via pyrophosphatase, which drives the formation of cAMP.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Synthesis of cysteine

A
  • s-adenosylhomocysteine is acted on by adenosylhomocysteinase, which generates adenosine and homocysteine - cystathionine beta-synthase and cystathionase are both B6 requiring enzymes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Explain the function of the HOX transcription factors in development

A
  • Following specification of axes, patterning takes place. Patterning defines which end of undevelopmed mass of cells is head, which is tail. It segments cells to define which parts become head, thorax, abdomen etc.- Patterning along ant/post axis is determined by homeobox (HOX) genes, which are a family of transcription factors containing a special DNA binding domain called homeodomain. There are 4 HOX clusters on 4 chromosomes- Expression of each of the genes belonging to a cluster correlates with position of respective cell in embryo and with timing of expression. Each cell along axis experiences a different ratio of expression of different HOX genes. Ultimately, the pattern of HOX gene expression determines fate of a cell in segment.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the precursors for gluconeogenesis?

A
  • Lactate, - Amino acids - Glycerol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Where is cholesterol synthesized, what is the rate-limiting step/enzyme, what is the precursor used, in what other pathway is this precursor seen? What enzyme is used to make this precursor? Is this the same enzyme used in the previous pathway? Explain. During what environmental conditions is the precursor made in both pathways?

A
  • Cholesterol synthesis takes place in the liver, intestine and reproductive tissues - Rate-limiting step = HMG-CoA reductase - Precursor = HMG-CoA – seen during synthesis of ketone bodies - Precursor synthesized by cytosolic HMG-CoA synthase isoform that is active in well-fed state. Mitochondrial HMG-CoA synthase isoform synthesizes the precursor when in starving/fasting state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

How does aspirin function?

A
  • It is an irreversible inhibitor of prostaglandin synthase and binds to the active site of the enzyme forming a covalent complex.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Outline salvage of pyrimidine bases

A
  • Orotate/uracil and thymine + PRPP = XMP + PPi (ez: pyrmidine phosphoribosyltransferase) - No cytosine salvage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What is the substrate for NO synthase? What affect does NO have on the vasculature? What enzyme is affected by NO? How is NO signaling turned off?

A
  • NO produced from arginine by the enzyme nitric oxide synthase. It is a gas and diffuses readily in cell, acting locally due to short half-life and conversion into nitrates, nitrites when reacted with o2 and water. - Typical function of NO is to cause relaxation of smooth muscle. - Typical mechanims: ACh is released from nerve terminal, causes activation of NO synthase in endothelial cells, triggering conversion of arginine into NO. NO diffuses to neighbouring smooth muscle cells, binds guanylate cyclase in the cytoplasm, stimulating production of cGMP, which activates cGMP-dependent protein kinase, leading to smooth muscle relaxation. - Inactivation: NO has a short half-life as it is highly reactive with oxygen and water, produced nitrates and nitrites. In addition, cGMP is degraded by phosphodiesterasterases yielding GMP, which are not able to activate cGMP-dependent protein kinases that lead to smooth muscle relaxation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is the Bohr effect? What causes it to occur?

A
  • It refers to oxygen’s binding affinity of Hb being inversely related to acidity and concentration of CO2.
  • When CO2 concentration is high and acidic environment exists, o2 affinity for Hb is low and therefore Hb is more likely to give up o2 in tissues where this environment exists.
  • Causes: H+ binds to histidines foming stabilized T-form salt bridge; CO2 binds to N-terminus forming negatively charged carbamates increasing stabilization of T-form.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Discuss concept of reduced fitness and new mutations in context of autosomal dominant disorders

A
  • In some autosomal dominant disease, an allele carriers has a reduced chance of reproduction, ie. reduced fitness- Therefore, reduced fitness of the carriers would lead to disappearance of the mutant alleles in populations- However, allele frequencies stay constant as new mutations appear constantly and compensate for the loss of mutant alleles- Low fitness of carriers means high percentage of new mutations- If parents are not affected by dominant disease, affected children could have inherited a new mutation- New mutations are seen in DMD, NF and achondroplasia. Genes implicated in these disorders are large, complex or contain mutation hotspots.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Would a normal cell divide if chromosomes were not correctly attached to the mitotic spindle? Explain

A
  • No - Anaphase of mitosis is delayed until chromosomes attach to mitotic spindle - Correct assembly of mitotic spindle activates anaphase promoting complex (APC) - APC allows degradation of cohesin complexes, which hold sister chromatids together at centromere, sister chromatids can move to opposite poles - APC acquires different subunit and causes degradation of cyclin A or B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Describe heme degradation and excretion

A
  • Heme is cleaved between A and B rings by heme oxygenase, yielding biliverdin - Biliverdin reductase used NADPH to form bilirubin, which is unconjugated (aka indirect) - Becomes bound to albumin and travels to liver where it is conjugated to two UGT molecules and becomes bilirubic diglucuronide. This is form that is excreted in bile - When in gut, BDG is modified into urobiliongen by gut and then spontaneously to urobilins / stercobilins which is found in feces - Some urobilinogen is absorbed back into blood and spontaneously converted into urobilin, which is urinated out.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Biochemical basis for megaloblastic anemia – explain how B12 deficiency leads to functional folate deficiency

A
  • Vitamin B12 deficiency results in deficiency in methionine synthase deficiency and therefore accumulation of N5-methyl THF and concomitant decreased in concentrations of more oxidized forms of THF, therefore Vitamin B12 leads to a functional folate deficiency - N5 N10 methylene is one of these more oxidized forms that would decrease. This is required for DNA synthesis (thymidine) - N10 formyl THF is required for purine synthesis, it is however decreased now too - Therefore lack of THF in oxidation states blocks DNA replication - Result is production of megaloblastic anemia – RBCs with large cytoplasm, but unable to divide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What molecules accumulate with a vitamin B12 deficiency? Explain which reactions these are implicated in?

A
  • 1.) D & L methylmalonyl-CoA (D and L methylmalonate) and 2.) N5-methyl THF - 1.) In process of converting VOMIT AAs to succinyl-CoA - 2.) In converting homocysteine and N5 methyl THF into methionine and THF respectively
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Describe synthesis of prostaglandins, thromboxanes and leukotrienes.

A

1.) Prostaglandins - AA to PGG2 via COX1/2 - PGG2 to PGH2 via PGH synthase - PGH2 is precursor to other prostaglandins via PGD/PGE/PGF synthase enzymes 2.) Thromboxanes - PGH2 to TXA2 via thromboxane synthase - TXA2 to TXB2 via hydrolysis in blood 3.) Leukotrienes - AA to leukotrienes via lipoxygenase and other enzymes

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

Role of glutathione-S-transferase enzyme family?

A
  • catalyze transfer of glutathione to molecules with reactive electrophiles, generating thioether linkage bw compound and cysteine of glutathione, preventing interaction with other molecules
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Describe chromosomal defects in Di George Syndrome

A
  • Deletion in chromosome 22 q. Frequency = 1/4000. Usually a new mutation- Malformations include: congenital heart defect, immunodeficiency, hypoparathyroidism, mental retardation, cleft palate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Function of glutathione

A
  • Intracellular reducing agent: scavenging free radicals and destroying peroxides - Drug detoxification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is penetrance? What is expressivity?

A
  • Penetrance: percentage of people with disease gene who develop symptoms- Expressivity is the severity of the symptoms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Heme synthesis enzymes affected by lead

A
  • Porphobilinogen synthase - Ferrochelatase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Contrast haploinsufficiency and dominant negative effect in the development of osteogenesis imperfecta subtypes

A
  • Osteogenesis imperfecta-1 is caused by mutations in the collagen I genes.- OI-1 causes deformity of skeleton and predisposes bones to breakage. - Frequency = 1/10000, AD- There are 4 different classes of OI depending on the number of proalpha1 and 2 collagen chains and also if the chains are defective or not. Defect in one chain may disturb the larger structure. This is referred to as dominant negative effect.- Allele heterogeneity is also exhibited in this disease in that the severity depends on the AA exchanged. Type I have brittle bones and blue sclerae without bone deformities. Type II is perinatal lethal. Type III is progressively deforming. Type IV has bone deformities with predisposition to bone fractures.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Explain the role of sorbitol in the pathology of diabetes

A
  • In DM pts, there is more free glucose in the cells than in other pts. This is a result of many things, including the failure of phosphorylation by hexokinase/glucokinase. This turns on aldose reductase, which uses NADPH to general sorbitol, which is used by polyol DH to form fructose, consuming NAD. As a result, antioxidant defenses that rely on NADPH are weakened and NADH is high, so glycolytic capacity is reduced. - Sorbitol also is osmotically active and draws water into cells, distorting cellular/tissue structure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is THF? What is its function? Where is it derived from?

A
  • THF = tetrahydrofolate - Serves as a carrier of one carbon groups - Derived from folate, vitamin B9, which is found in foliage, green leafy veg, liver, eggs and beans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are disorders of FA degradation?

A
  • Primary carnitine deficiency - Acyl-CoA dehydrogenase deficiency – eg. VLCAD, LCAD, MCAD and SCAD. MCAD is most common. - Refsum disease – alpha-oxidation deficiency - Peroxisome biogenesis disorders (PBD)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Describe regulation of G1/S transition

A
  • S phase requires active cyclin A-CDK2, which is initially formed as inactive complex with inhibitor called p27kip1 - Late g1: cyclin E-CDK2 is desphosphorylated by phosphatase (Cdc25A) - cyclin E-CDK2 phosphorylates p27kip1 in complex w/ cyclin A-CDK2, cyclin A-CDK2 is activated by being freed from p27KIP2 - It promotes DNA replication by phosphorylating components of ori complexes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Beri-beri. a.) What is it? What is it caused by? b.) What are the symptoms associated with it? c.) What are the two forms of this disorder? d.) What organ/tissue(s) are most affected by this disorder? e.) Do RBCs contribute to this condition? Explain. f.) Which populations are susceptible to this disorder?

A
  • a.) It is a severe thiamin (B1) dietary deficiency. - b.) Neuromuscular symptoms – muscle weakness, muscle atrophy, fatigue, peripheral neuropathy and lactic acidemia. - c.) Dry beri-beri which affects neuromuscular function, does not lead to fluid retention; Wet beri-beri causes peripheral edema and cardiac failure. - d.) It affects the nervous system and musculoskeletal system primarily. - e.) No, as they do not contain mitochondria, where enzymes such as PDH complex and aKGDH that participate in linking glycolysis to TCA cycle and TCA cycle function respectively. - f.) Populations that have high intake of polished rice diets.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Explain why NADH concentration increases while NADPH concentration decreases in alcohol-metabolizing cells.

A
  • The main step to handling ethanol in the liver is through two dehydrogenases (alcohol DH and aldehyde DH), both, which produce NADH. - The other way to handle ethanol is through the higher capacity MEOS system, which as a means to process the alcohol, also oxidizes NADPH.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Give the frequency of chromosomal aberrations, both at conception and at birth

A
  • Structural chromosomal abnormalities (rearrangements, loss and duplications) occur in approximately 0.5 % of pregnancies and 0.2 % of live births
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What enzyme is involved in regulating AA catabolism? How does regulation occur?

A
  • Primarily regulated by liver glutamate dehydrogenase (in mitochondria) - Regulation of glutamate DH is by cellular energy charge - High energy (high GTP, high NADH) = inhibition of enzyme - Low energy (high ADP) = activation of enzyme
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Describe base excision repair process using the example of the removal of a cytosine residue that has been deaminated to uracil.

A
  • DNA glycosylases recognize diff types of altered bases in DNA and catalyze their hydrolytic removal - Cytosine spontaneously deaminated to uracil - Uracil DNA glycosylase removes uracil base creating AP site - AP endonuclease introduces nick - DNA phosphodiesterase removes remnant sugar and phosphate - DNA Pol and ligase fill in correct base
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Explain how LDL receptor production changes in response to cholesterol. Discuss mechanism

A
  • Transcription of LDL receptor gene is upregulated in response to low cellular cholesterol - Result: more LDL receptor production and enhanced cholesterol uptake from blood Low-level transcription - SP1 (zinc-finger protein) binds to GC rich regions in LDL receptor promoter - SP1 requires CRSP protein cofactor to be activated - SP1 and CRSP help pol II and GTFS to assemble at promoter - These are all considered basal transcription factors Up-regulated transcription - Requires basal transcription factors - Also SREBP-1a enters into nucleus as triggered by low cellular cholesterol - SREBP-1a binds to SRE and recruits HAT activity plus some other proteins - Result = enhanced transcription of LDL-receptor gene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Explain the dominant inheritance of familial hypercholesterolemia

A
  • FH is frequent 1/500 person disease, AD- Heterozygotes have 2-fold elevation in LDL levels as there are insufficient receptors to clear LDL from serum. Homozygotes have 4-fold evelated in LDL levels. This is an example of haploinsufficiency, allele heterogeneity- Symptoms: xanthomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What is meant by term ‘salvage pathways’? What are the sources of substrates for these pathways?

A
  • Conversion of free bases and nucleosides to nucleotides - Sources are: diet, purine/pyrmidine nt degradation products
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What is methemoglobin?

A

-Type of hemoglobinopathy. This is Hb where Fe2+ is oxidized to Fe3+ and has reduced ability to deliver o2 to tissues.

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

Which molecules are allosteric effectors? What are their signals and affects?

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

What is Rett Syndrome?

A
  • X-linked dominant disorder mapped to mutations in the gene for methyl-cytosine binding protein MECP2- Mutation leads to loss of transcriptional silencing- Symptoms: autism-like symptoms, repetitive teeth grinding and hand-wringing, motor problems and characteristic gait. Onset age 6-18 months
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What gluconeogenic deficiency(ies) seen in human? What is/are the clinical presentation(s)?

A
  • G6PD deficiency: defect that lowers the activity of PPPathway and cells have lower NADPH levels. These levels impair glutathione reduction and deprive RBCs of antioxidant protection. Affected pts are very sensitive to hydrogen peroxide, which are produced in infections (by macrophages), by drugs (eg. Primaquine that is used to treat malaria and pneumocystis pneumonia) and during consumption of fava beans (known as favism) as these cause oxidatie stress and trigger hemolytic crisis and hemolytic anemia. Most prevalent in African and Mediterranean Americans. It is x-linked recessive.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What is the enzyme that commits acetyl-CoA to FA synthesis? What does this reaction do?

A
  • Acetyl-CoA carboxylase, which converts acetyl-CoA to malonyl-CoA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Treatment of argininosuccinate lyase deficiency

A
  • As a result of this deficiency – argininosuccinicaciduria - Treat with arginine – supports continued citrulline synthesis and continuation of cycle. Argininosuccinate is water soluble and relatively non-toxic, gets eliminated from kidney as waste - Argininosuccinate carries both nitrogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

Describe vitamin B12 absorption into body and storage

A
  • R-binders in salivary enzymes bind B12 and pass it onto intrinsic factor, produced by stomach parietal cells - Absorbed in ileum through receptor mediated endocytosis - Binds to transcobalamin and is transported to tissues - Preferentially distributed to liver as vit-B12:transcobalamin complex and stored there. Kidney has some B12 stores
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

List and describe the glycogen storage diseases (GSD). What is occurring in each and what is the clinical presentation?

A
  • GSD-I: aka Von Gierke’s disease: deficiency in G-6-phosphatase. Leads to hypoglycemia and increased glycogen stores in liver. This ez is only found in liver. - GSD-II: aka Pompe’s disease: deficiency in alpha-glucosidase (aka acid maltase). It is required for the degradation of glycogen, which with the deficiency, slowly accumulates in the lysosome. Over time, this process eventually kills affected cells. Death occurs by cardiac or respiratory failure. - GSD-III: aka Cori’s disease: deficiency in debranching enzyme. Glycogen granules grow large because only the non-branched, outer layers of the deposits can be degraded. Presents in early childhood or infancy with hepatomegaly, hypoglycemia and growth retardation. Can also present with muscle weakness. Some pts develop cardiomyopathy. - GSD-V: aka McArdle’s disease: deficiency in glycogen phosphorylase in muscle. Muscle is unable to use glycogen. Leads to greater than normal glycogen stores in muscles. During periods of high glucose demand, ie. exercise, patients experience muscle cramps. - GSD-VI: aka Hers disease: deficiency in glycogen phosphorylase in liver. Presents with mild to moderate hypoglycemia, mild ketosis, growth retardation and prominent hepatomegaly.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Fuel preferences / energy needs of RBCs, muscles, brain, adipose and liver

A
  • RBCs: glucose, anerobic metabolism - Non-cardiac muscle: glucose-anaerobic/aerobic, FFAs - Cardiac muscle: glucose aerobically only, FFAs - Brain: glucose aerobically, ketone bodies after serious period of starvation - Adipose: glucose, TAG – FAs - Liver: FAs, glucose, AAs and lactate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Describe the effects of 5-azacytidine on genome methylation

A
  • Hypermethylation can be reversed by this drug, which is a DNMT inhibitor. Treatment serves to allow for re-activation of silenced tumor suppressor genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Explain the different types of adrenergic receptors.

A
  • 1.) Alpha receptors a.) alpha-1 subtype: Gq protein coupled = activation of PLC b.) alpha-2 subtype: Gi protein coupled = inhibition of adenylate cyclase - 2.) Beta receptors three subtypes (beta-1,2,3) are Gs protein coupled = activation of adenylate cyclase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What is situs inversus and situs ambiguous, what causes it?

A
  • Both are defects in left/right axis formation- Situs inversus: all organs are complete mirror images in what is normally found (known a situs solitus). Usually asymptomatic- Situs ambiguous describes a more serious condition in which orientation of organs is randomized. This is usually accompanied by heart defects.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Under what conditions will lactic acid fermentation occur in the body? Why? What enzyme is involved? Why is this problematic in the body?

A
  • Concentration of NAD to NADH are tightly monitored. Reason: NAD+ is limited in concentration. NADH must be recycled to NAD+ in order for glycolysis to continue. Lactic acid fermentation takes the electrons from NADH and places them onto pyruvate, generating lactate. This is done by lactate dehydrogenase. - Lactic acid buildup leads to lactic acidosis. Cori cycle tries to circumvent this from occurring.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are other pathologies related to mutations in collagen?

A
  • Ehlers-Danlos: CT disorder characterized by skin fragility, skin hyperextensibility and joint hypermobility resulting from mutations in collage proteins or proteins required for collage processed. Some benign symptoms, others complicated including bowel and arterial rupture. - Osteogenesis imperfecta: brittle bone diseases characterized by fragile bones, abnormalities in teeth, hearing losss. It can be lethal, otherwise individuals can have normal lifespan with slightly reduced bone mass.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

RBC surface antigens. Core? Sugar residues for O, A and B antigen?

A
  • Core: Serine – Gal – GlcNAc – Gal X – Fucose - O: Serine – Gal – GlcNAc – Gal X – Fucose - A: Core – Gal X attached to GalNAc as well as Fucose - B: Core – Gal X attached to Gal as well as Fucose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

Describe the 4 main groups of amino acids. What are their characteristics?

A

a.) non-polar, aliphatic: hydrophobic and chained/branched, found interior aspect of proteins, uncharged Gly, Ala, Val, Leu, Ile, Met, Pro • Gly – no asymmetric carbon (2 Hs) • Pro – ring structure • Met – sulfur-containing AA b.) aromatic: hydrophobic (non-polar rings), often found interior of water-soluble proteins, uncharged Phe, Tyr, Trp • Tyr – has OH group and can be phosphorylated c.) polar, uncharged: hydrogen bond, interior/exterior of proteins Ser, Thr, Cys, Asn, Gln • Ser/Thr – have OH groups that can be phosphorylated • Cys – sulfur-containing AA, forms disulfide bridges with other thiol containing molecules (ie. cystine = 2 cysteines) d.) Ionizable side chained AA – basic/acidic: Acidic: Asp, Glu (net -1 charge at physiological pH) Basic: Lys, Arg, His* (net +1 charge at physiological pH) * His can be basic/uncharged as pKa = 6.0

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What is familial adenomatous polyposis? Outline the role of APC in the WNT signaling pathway. How does loss of APC function lead to cell proliferation?

A
  • Hereditary condition due to mutations in APC (adenomatous polyposis coli) gene that encodes tumor suppressor APC. Develop 100-1000s of adenomatous polyps. 100% of untreated FAP pts developed colorectal adenocarcinomas. Prophylactic colectomy is done - APC usually functions to down-regulate WNT signaling pathway - When mutation exists, WNT signaling is activated and expression of cyclin D, Myc and other growth-promoting genes occur - Additional mutations are implicated: oncogenic forms of Ras, may impact p53 function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Describe utilization of glucose by brain, RBCs, brain, liver, muscle and adipose.

A
  • RBCs: glucose through glycolysis to produce lactate (used in liver), G6P into PPP - Brain: glucose through glycolysis to produce acetyl-CoA (oxphos), G6P into PPP. No production of lactate - Muscle/heart: glucose through glycolysis to produce both lactate (used in liver, ? produced in heart) and acetyl-CoA (oxphos), G6P into PPP, G1P as intermediate into glycogenolysis and glycogen synthesis - Adipose tissue: same as muscle/heart, except no lactate production AND acetyl-CoA used for FA synthesis in addition to oxphos. - Liver: glucose through glycolysis to produce pyruvate (to acetyl-CoA to oxphos or FA synthesis), pyruvate to OAA (to gluconeogenesis), G6P to PPP, G1P in glycogenolysis and glycogen synthesis. * no exit of glucose to serum unless from liver.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Impact of diabetes on glucose and FA metabolism.

A

1.) Glucose: Insulin has no impact on liver, fat and muscles, as a result: - Glucose comes in, remains in serum - Liver receives AA from muscles and performs gluconeogenesis - Fat leaves adipose and enters liver, where ketone bodies are generated 2.) Lipids: insulin does not activate LPL and deactivate HSL, as a result: - Chylomicrons carrying dietary TAG stay in serum and cannot enter adipose via action of LPL - HSL is on and is performing lipolysis, TAGS are degraded into FAs, which enter circulation and travel to liver. FAs are generated in excess of what is needed to generate ketone bodies. As a result, TAGs are synthesized and packaged into VLDL, which become elevated in blood.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

How is CDK4/6 activity inhibited

A
  • INK4 proteins inhibit these proteins. - INK4 transcription is stimulated by growth inhibitory factors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What is purpose of vitamin B12 in THF metabolism and keeping functional folate pool?

A
  • Serves to take the most reduced form of THF = N5 methyl THF back to THF form and in the process converts homocysteine to methionine - THF has no other functions in most reduced form, only when in oxidized forms does it have specific functions, including in DNA synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What are point mutations? How might point mutations arise? Distinguish bw silent, missense, and nonsense mutations.

A
  • Exchange of one nt for another - Caused by replication errors, chemical mutagens, radiation, DNA damage repair - Silent: no changes to AA - Missense: change from one AA to another - Nonsense: change from AA to stop codon
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

How can cholesterol synthesis be inhibited in a clinical setting? Explain how this works.

A
  • Use of statins - Statins inhibit the HMG-CoA reductase enzyme and prevents synthesis of mevalonic acid, a precursor molecule to farnesyl-PP, a precursor to cholesterol.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

ETC. a.) What are the components? What are the enzyme complexes involved? Name them b.) What reaction does each component catalyzed? List reduced substrates and oxidized products. c.) What is the sequence of flow through these carriers? Draw and indicate the reactions of each of the complexes. Include movement of protons. What coenzymes and metals do each of the complexes require? d.) Which components are proton pumps? e.) There are other dehydrogenases besides complexes I through IV contained in the mitochondrion. What are they?

A
  • a.) dehydrogenase enzymes (complexes), heme-iron proteins (aka cytochromes), iron-sulfur proteins, CoQ/ubiquinone Dehydrogenase enzymes are: Complex 1: NADH-CoQ reductase Complex 2: Succinate-CoQ reductase Complex 3: CoQH2-cytochrome c reductase Complex 4: Cytochrome c oxidase - b.) Complex 1: NADH + H+ + CoQ = NAD+ + CoQH2 Complex 2: Succinate + CoQ = Fumarate + CoQH2 Complex 3: CoQH2 + 2 cyt c (Fe3+) = CoQ + 2 cyt c (Fe2+) + 2H+ Complex 4: 4 cyt c (Fe2+) + 4H+ + o2 = 4 cyt c (Fe3+) + 2H2o - c.) see picture - d.) Protons are moved to the intermembrane space via complex I, II and IV - e.) Fatty acyl-CoA DH and glycerol-3-P DH. These both reduce CoQ and are not part of the std I-IV complexes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

How do the oxygen-binding properties of HbF and HbA1 differ? Why is this important?

A
  • HbA1 refers to the alpha2beta2 Hb tetramer, which is the most common form in adults, account for over 90% of total in blood - HbF is the predominant Hb tetramer present in the 2nd/3rd trimester of development. In contrast to adult Hb, HbF curve is shifted to the left indicating that at a specific o2 pressure, HbF is more saturated than adult Hb. Has higher affinity for o2 than adult Hb. This ensures that the fetus’ high metabolic o2 requirements are met. - This is due to the replacement of a histidine residue with a serine residue in the gamma chain of HbF. As a result, HbF has reduced affinity for 2,3-BPG and is left shifted.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Define primary structure, secondary structure, tertiary structure, and quaternary protein structure.

A

a. Primary structure: the amino acid sequence of the polypeptide chain b. Secondary structure: a segment of polypeptide chain that has regular repeating structure. Results from H-bonding between the C=O and H-N groups of the polypeptide backbone. Alpha-helices and beta-pleated sheets. c. Tertiary structure: the 3D structure resulting from the interaction of various secondary structures and non-ordered regions of the polypeptide chain, results from disulfide bridges (covalent) and hydrogen bonding, ionic interactions (salt bridges) an hydrophobic forces (all non-covalent) d. Quaternary structure: protein molecules that have more than one subunit, each with tertiary structure, have overall quaternary structure. The subunits associate through same forces found in tertiary structures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

How does beta-oxidation differ in peroxisomes from other tissues?

A
  • Beta-oxidation in peroxisomes involves preferential catabolism of LCFAs and produces hydrogen peroxide instead of FADH as seen in beta-oxidation in other tissue. Peroxisomes use oxidase enzyme
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Explain how F26BP affects glycolysis. Include details about the enzymes involve, pathways activated and substrates affected.

A
  • Take home message: high concentrations of F26BP stimulate glycolysis; low concentrations of F26BP inhibit glycolysis - F26BP is produced by enzyme PFK2 (produces F26BP from F6P) - F26BP activates PFK1 leading to increased production of F16BP - F26BP inhibits fructose bisphosphatase leading to increased production of F16BP - Insulin stimulates PFK2 via cAMP leading to increased concentration of F26BP, which stimulates PFK1, inhibits bisphosphatase and stimulating glycolysis - Glucagon inhibits PFK2 via cAMP leading to decreased concentration of F26BP, which means that PFK1 is inhibited, bisphosphatase is stimulated and glycolysis is inhibited - AMP stimulates PFK2, causing increased concentrations of F26BP, which stimulates PFK1, inhibits bisphosphatase and stimulated glycolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What molecules are derived from tryptophan?

A
  • Serotonin - Melatonin - Niacin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Describe and explain graphically with 1/V vs. 1/[S] plots and pictorially with drawings: • irreversible inhibition

A
  • Graphically indistinguishable from a non-competitive inhibition graph.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Why do some porphyrias cause photosensitivity?

A
  • Porphyrins are conjugated systems, which absorb UV and visible light. - The light energy must be discharged upon being absorbed, so it is discharged into the tissues and generates ROS. As a result photosensitivity of skin occurs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Explain process of beta-oxidation. Include enzymes, substrates, cofactors and products.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Describe reactions that form triacylglycerols.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is the role of phospholipids in prevention of respiratory distress syndrome in premature infants?

A
  • Surface of lung epithelium is covered with secretion that prevents alveolar membranes from sticking together (surfactant) - This surfactant is rich in phospholipids, particularly dipalmitoyl phosphatidylcholine (sometimes called dipalmitoyl lecithin). - This surfactant is produced at about 31 weeks gestation. - Infants born prematurely have a lecithin/sphingomyelin ratio below 2, indicating they have insufficient amounts of surfactant, which puts them at risk for respiratory distress syndrome (RDS). - Artificial surfactant mixtures treat premature infants. - Also they are given corticosteroid growth hormones to stimulate lung maturation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

How does methotrexate function to inhibit thymidylate synthase?

A
  • Methotrexate is an antifolate medication - Remember, N5-N10-methylene THF is needed to convert dUMP to dTMP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

How does methylation silence transcription?

A
  • Methylation of DNA occurs on cytosine residues in CpG islands- After methylation occurs, methylcytosine binding proteins bind methylcytosine.- MBPs interact with repressors of transcription leading to transcriptional block and HDACs that lead to chromatin condensation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

CF. Inheritance, defect, frequency

A
  • AR- Defect that impairs trafficking of CFTR chloride channel to cell surface causing pulmonary problems and pancreatic malfunction. CF pts have 2-5 times amount of NaCl in sweat. Severity varies (allele heterogeneity, modifier loci): pancreatic sufficient or insufficient- 1/2000
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Describe how the receptor tyrosine kinases HER2 and ErbB1 can be converted into constitutively active forms that function as oncoproteins

A
  • HER2 (aka Neu): point mutation allows activation of kinase in the absence of ligand - ErbB1: deletion of ligand bind domain results in constitutive activation of kinase domain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What is the malate-aspartate shuttle? Where does it occur? Draw it.

A
  • It is a shuttle mechanism to move electrons from NADH in cytoplasm to the mitochondrion. It occurs in liver and heart.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

Why is gluconeogenesis not a reversal of glycolysis? Explain. Explain what reactions occur to circumvent this issue.

A
  • Glycolysis has 3 irreversible reactions: hexokinase, phosphofructokinase 1 and pyruvate kinase. In order for glucose to be synthesized, ie. gluconeogenesis, 4 new enzymes not seen in glycolysis are seen: Pyruvate carboxylase, PEP carboxykinase, Fructose-1,6-bisphosphatase and glucose-6-phosphatase.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Name 3 antineoplastic agents that are inhibitors of DNA replication and repair

A
  • Cytarabine - Cyclophosphamide - Doxorubicin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Outline salvage of pyrimidine nucleosides.

A
  • Uridine / cytidine + ATP = UMP / CMP + ADP (ez: uridine-cytidine kinase)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

Fructose 1,6 bisphosphatase deficiency

A
  • AR- Fasting hypoglycemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

Describe how a mutation in the amino acid sequence could affect protein structure and/or function. Give several examples.

A

A change in amino acid sequence will lead to different interactions and therefore different folding (see 14). Different structure will confer different function. The original purpose of the protein can no longer be served. Examples of diseases where misfolded proteins are implicated: • CJD: inherited or spontaneous • Kuru: infectious • vCJD: infectious Above diseases lead to spongiform encephalopathies (neurodegeneration, brain becomes spongy) • Parkinson’s: loss of dopaminergic neurons, formation of Lewy bodies (aggregates of alpha-synuclei protein seeds more) leads to tremor, bradykinesia, postural instability • Alzheimer’s: neurological condition leading to dementia from plaque/fibrillary structures in brain containing amyloid-beta and tau proteins that seeds more

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Describe homocystinuria. Causes? Symptoms? Treatment?

A
  • Grossly evelated homocysteine (plus other metabolites of this) accumulat in body and are seen in urine - Most commonly due to cystathionine beta-synthase mutation - Symptoms: dislocation of optic lens, osteoporosis, lengthening and thinning of long bones, thromboembolism, intellectual disability - Treatment: low methionine diet with cysteine supplements, pyridoxine or betaine supplements
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

Describe ways by which ATM/ATR kinases can prevent cell cycle progression in the face of DNA damage.

A

1.) ATM is activated by double-stranded breaks 2.) ATR is activated by UV light and certain DNA-damaging drugs - Both kinases stabilize p53 - P53 upregulates p21cip1 - p21 inhibits activates a kinase that prevents cdc25a and cdc25C from activating cyclin A/E-CDK2 (S phase) and cyclin A/B-CDK1 (M phase) respectively. - ATM and ATR also activate a checkpoint kinase that prevents cdc25a and cdc25C from activating cdc25a and cdc25C from activating cyclin A/E-CDK2 (S phase) and cyclin A/B-CDK1 (M phase) respectively

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What roles do chromatin remodelling complexes, histone acetyltransferases (HATs), histone deacetylases (HDACs) and DNA methylation play in controlling the availability of genes for transcription?

A

1.) chromatin remodeling complexes: use ATP to change nucleosome structure temporarily to allow or inhibit transcription 2.) HATs: acetylate lysine residues in histones, reduce net pos charge of protein, weakens histone:DNA interaction, facilitates transcription 3.) HDACs: remove acetyl groups from histone, increasing net pos charge of protein, strengthens histone:DNA interaction, inhibits transcription 4.) DNA methylation: methyl groups added to cytosine, found in silent regions of genome, binds proteins that recruit HDACs to DNA, promoting condensation and inhibiting transcription

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Molecular action of insulin

A
  • Binds to IR in PM of target cells - IR = kinase, phosphorylates itself then IRS - Phosphorylation cascade ensues, but at level of enzymes, dephosphorylation occurs via phosphatase - Cell undergoes dramatic changes in protein activation, protein localization and gene transcription
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Which enzymes of gluconeogenesis are regulated? What factors inhibit and stimulate these enzymes? Which is the main regulated step? Which are reversible/irreversible?

A
  • Pyruvate carboxylase: +: acetyl-CoA; -: insulin - PEP carboxykinase: +: glucagon via cAMP; -: insulin, AMP - Fructose-1,6-bisphosphatase: +: citrate; -: F26BP, AMP (main regulated step) - Glucose-6-phosphatase: +: glucagon; -: insulin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What are the 2 sources of cytosolic NADPH?

A
  • PPP via G6PD - Conversion of malate to pyruvate via malic enzyme
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What is Smith-Lemli-Opitz Syndrome?

A
  • Impairment of Shh signaling secondly to defect in cholesterol biosynthesis. - This is an autosomal recessive disorder affecting 1/20-50K- Leads to multiple severe congenital malformations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Describe and explain graphically with 1/V vs. 1/[S] plots and pictorially with drawings: • noncompetitive inhibition

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What molecule is the temporary ammonium ion carrier in the body?

A
  • glutamine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What inhibits thymidylate synthase?

A
  • 5-fluorouracil - Methotrexate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

Defect in Shh mutation

A
  • Midline defects such as holoprosencephaly = failed or incomplete separation of forebrain early in gestation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

Describe de novo synthesis of pyrimidines

A

1.) Synthesis of uridine nucleotides (three* enzymes below are part of CAD multifunctional protein) - Gln + bicarb + ATP = carbomyl phosphate + ADP + glu (ez: *carbomyl phosphate synthetase II) - Carbomyl phosphate + Asp = N-carbamoyl aspartate + Pi (ez: *asp transcarbamoylase) - N-carbamoyl aspartate = dihydroorotate (ez: *dihydroorotase) - Dihydroorotate + NAD = orotate + NADH (ez: dihydroorotate DH) - Orotate + PRPP = OMP + Pi (ez: ^orotate phosphoribosyltransferase) - OMP = UMP + Co2 (ez: ^OMP decarboxylase) Notes: three beginning ezs are part of *CAD multifunctional enzyme, last two are part of ^UMP synthase multifunctional enzyme 2.) Synthesis of cytosine nucleotides - UMP is converted to UTP via nucleoside-5-monophosphate kinases and nucleoside diphosphate kinases - UTP + glut + ATP = CTP + gln + ADP + Pi (ez: CTP synthase) 3.) Synthesis thymine nucleotides - Note: TMP, TDP or TTP are never synthesized (never get ribose forms), only make dTMP, dTDP and dTTP - dUMP + N5,N10-methylene THF = dTMP + THF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What is the name of the molecule that synthesizes rRNA?

A
  • RNA pol I, some Pol III
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

Distinguish between carbomyl phosphate synthetase I and carbomyl phosphate synthetase II? Clarify what their substrates are and where each activity is found in the cell?

A

1.) CPS I: - Found in mitochondria of liver, synthesizes carbomyl phosphate for urea cycle - Substrates: bicarb, 2ATP, ammonium; Products: carbomyl phosphate, ADP, phosphate 2.) CPS II: - Found in cytosol of liver, synthesizes UMP - Substrates: bicarb, 2ATP, gln; Products: carbomyl phosphate, ADP, glu

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

Write the Michaelis-Menten equation. Define what each of the variables are.

A
  • v = (Vmax [S])/(Km + [S]) - v = rate of ez catalyzed reaction - Vmax = maximum reaction rate (reached when every ez molecule is bound to substrate). Depends only on ez concention. - [S] = substrate concentration - Km = Michaelis-Menten constant (k2+k3)/k1 – it is the substrate concentration at which v=Vmax / 2 . Km is independent of enzyme concentration, reflects physical properties of enzyme.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

An inability to generate THBtn would impact synthesis of which of the following molecules? A.) histamine B.) NE C.) Thyroid hormone

A
  • B.) NE (remember, tyrosine in thyroglobulin molecule, not free tyrosine)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Explain what a non-competitive inhibitor is?

A
  • A non-competitive inhibitor is a reversible inhibitor. These bind at sites on the enzyme other than the active site, forming inactive enzymes (E-I form or E-S-I). Increasing the concentration of the substrate does not influence inhibitor binding. It is reversible if the inhibitor can be removed. Enzyme will work after inhibitor removal.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Describe and explain graphically with 1/V vs. 1/[S] plots and pictorially with drawings: • competitive inhibition

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

Which reactions in glycolysis use ATP, produce ATP and produce NADH? What is the net production of ATP from 1 molecule glucose?

A
  • use of ATP: hexokinase (produce G6P from glucose) and phosphofructokinase (produce F16BP from F6P) - produce ATP: phosphoglycerate kinase (produce 3PG from 13BPG) and pyruvate kinase (produce pyruvate from phosphoenolpyruvate) - produce NADH: glyceraldehyde phosphate DH (produce 13BPG from dihydroxyacetonephosphate) - net ATP: 2 - net NADH: 1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What is the function of farnesyl PP?

A
  • converted into dolichol-PP for protein glycosylation - converted into coQ for electron transport - used for prenylation (post-translational) of proteins which influence water solubility of proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Pyruvate. a.) What is the fate of pyruvate? b.) What enzyme converts pyruvate to acetyl-CoA? c.) What enzyme converts pyruvate to oxaloacetate? d.) What determines whether pyruvate is used to form acetyl-CoA or oxaloacetate?

A
  • a.) lactate, oxaloacetate, acetyl-CoA and alanine - b.) PDH complex - c.) pyruvate carboxylase - d.) pyruvate carboxylase is allosterically activated by acetyl-CoA. When there is sufficient amount of acetyl-CoA and therefore little need to divert more pyruvate into acetyl-CoA, pyruvate carboxylase converts pyruvate to oxaloacetate.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

A defect in tyrosine hydroxylase would impact synthesis of all except? A.) dopamine B.) epinephrine C.) melanin

A
  • C.) melanin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Describe how Phe, Tyr are degraded

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

Describe metabolism of glucose, AAs and FAs in well-fed state

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

Explain how the genetic and the environmental influences on complex disease can be estimated

A
  • Twin and adoption studies (MZ vs DZ twins)- Concordant trait = trait shared by both twins- Discordant trait = trait not shared by both twins- Diseases with significant genetic component will show higher concordance rate in MZ twins than in DZ twins using crude measure known as heritability H2- H2 = (concordance MZ – concordance DZ) x 2- High heritability = trait is determined predominantly by genetic factors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

Synthesis of aspartate

A
146
Q

Which AAs are degraded to acetyl-CoA and fumarate?

A
  • To acetyl-CoA: Leucine, Lysine, Phe, Tyr, Trp, Iso - To fumarate: Trp, Tyr, Phe
147
Q

Where are O-glycosylated proteins seen?

A
  • Proteoglycans of the ECM - H-antigen on surface of RBCs (predictable)
148
Q

List the enzymes involved in glycogenolysis. Explain what factors regulate each, ie. what inhibit and stimulate each?

A
  • Glycogen phosphorylase: +: glucagon, epinephrine; -: insulin - Glucose-6-phosphatase: +: glucagon, epinephrine; -: insulin - Debranching enzyme: not regulated
149
Q

Types of glycosylation. Describe

A
  • N-linked: starts in ER before protein folding is complete, adds sugars to Asn residues in protein in predictable fashion, modification of this can occur in Golgi - O-linked: starts in Golgi after protein folding is complete, adds sugars to serine or threonine residues, but not in predictable fashion
150
Q

What is Treacher-Collins Syndrome?

A
  • Pts present with cleft palate, micrognathia (small jaw), down slanting palpebral fissures and malar hypoplasia (underdeveloped zygoma). - This is an AD disorder. Individuals have high risk for complications relating to this disorder – eg. Cardiovascular.
151
Q

Give figures for the genetic and environmental contributions of the burden of congenital malformations

A
  • In over 50% of birth defects, no cause can be identified, remaining are:- Genetics: 50% have complex inheritance, 25% caused by chromosomal defects, 20% caused by single-gene mutations- Non-genetic factors such as maternal medication and infections = 5%
153
Q

Why does the drug hydroxyurea block cell proliferation?

A
  • Inhibits ribonucleotide reductase and prevents dNTP synthesis, blocking cell division
155
Q

Describe the synthesis of pyruvate from AAs.

A
156
Q

Thiamin deficiency. a.) What vitamin is deficient? Where is this required? b.) Who does it affect? c.) What are the early symptoms? d.) What are the advanced / severe deficiencies of thiamin? Which groups are affected? What are the symptoms?

A
  • a.) Vitamin B1. Required for PDH and alpha-KGDH enzymes to function. - b.) Primarily affects alcoholics, elderly and low income groups who are more likely to have poor diets. - c.) loss of appetite, constipation, nausea, fatigue, depression, peripheral neuropathy - d.) Wernicke-Korsakoff = advanced deficiency frequently seen in alcoholics: mental confusion, ataxia, loss of eye coordination Beri-Beri = severe deficiency seen in populations that heavily rely on polished rice diets: neuromuscular symptoms – muscle weakness, muscle atrophy, fatigue, peripheral neuropathy, lactic acidemia
157
Q

Describe familial hyperinsulinemic hypoglycemia type 6. Causes? Result?

A
  • Mutations to glutamate dehydrogenase enzyme rendering it insensitive to inhibition by GTP. - As a result, increased AA catabolism occurs in environment that is high energy - Leads to elevated levels of ATP and hyperammonemia - Increased ATP in beta-cell promotes insulin releases, resulting in hypoglycemia - Less active urea cycle with more ammonia production = hyperammonemia
159
Q

Describe the clinical presentation of pyruvate kinase deficiency.

A
  • In this disorder, RBCs are deprived of ATP (only get 1 ATP instead of 2 from glycolysis), which leads to lysis of the cells (chronic hemolytic anemia) because the membrane potential cannot be maintained. These patients present at pale, jaundiced, fatigued, SOB, tachycardic. They have splenomegaly, excess of iron in blood and have gallstones. Severe cases require pts to have regular blood transfusions.
159
Q

Which is the only purine nucleoside that can be salvaged to a nucleotide?

A
  • Adenosine
159
Q

Synthesis of serotonin

A
160
Q

Synthesis of glutamine

A
161
Q

How are proteins directed to lysosomes

A
  • Proteins directed to lysosome require phosphomannose
162
Q

Describe how ammonium ions are transported and excreted within the blood

A
  • Glutamine produced in peripheral tissues enters the bloodstream and is absorbed by the kidneys, liver and gut - Amide group is hydrolyzed by glutaminase and produces glutamate and ammonium ion - Kidney excretes NH4+ directly and liver produces urea in urea cycle. Glutamine is nutrient in gut, ammonium enters hepatic portal and metabolized by liver
163
Q

Discuss how proteins can return to their interphase states after the M phase

A
  • APC causes degradation of cyclin A and B, which shuts off CDK1 activity and stops mitotic events from occurring - Desphosphorylation of cyclin-CDK targets by Cdc14 returns proteins to their interphase state: promotes decondensation of chromatin, disassembly of mitotic spindle, reassembly of nuclear membrane, cytokinesis) - APC becomes inactivated and Rb is dephosphorylated
164
Q

Pyruvate carboxylase. a.) What is the function of this reaction? b.) Write out the reaction is catalyzes. Include all energy components and coenzymes. c.) What is an allosteric activator of this enzyme? Why is that important?

A
  • It is an anaplerotic reaction to regenerate oxaloacetate when levels are low. - Pyruvate + ATP + HCO3 = oxaloacetate + ADP + Pi + H+ (ez: pyruvate carboxylase, coenzyme: biotin) - Acetyl-CoA. When there is sufficient amount of acetyl-CoA and therefore little need to divert more pyruvate into acetyl-CoA, pyruvate carboxylase converts pyruvate to oxaloacetate.
166
Q

What is meant by term essential AAs. List them. Special case

A
  • AAs that cannot be synthesized - Pvt Tim Hall = mnemonic - Phe, Val, Thr, Try, Iso, Met, His, Arg, Leu, Lyc - Special case = Arg – can be synthesized, not sufficiently to keep up with needs
167
Q

Describe elongation and termination of translation

A

1.) EF-1-GTP charges tRNA molecule (EF1-GDP = product). AA-tRNA moves into A site 2.) Peptide bond formation occurs 3.) EF-2-GTP hydrolysis allows ribosomal complex to move one codon down with mRNA-peptidyl-complex now occupying the P site and the A site is empty. Uncharged tRNA leaves through E site 4.) Ribosome is now ready to repeat the cycle 5.) Once the stop codon is moved into the A site, eRF bound to GTP is hydrolyzed and the ribosomal complex dissociates

168
Q

Three types of jaundice: Causes of each? Explain? What happens to color of urine and feces in each case?

A
  • Pre-hepatic jaundice: hemolysis. More unconjugated bili in blood than liver can deal with. Moves into tissue membranes. No change to urine or feces color - Hepatic/hepatocellular jaundice: liver disease prevents conversion of unconjugatd bilirubin into bilirubin diglucuronide. Unconjugated bilirubin moves into other tissue membranes. Urine is pale, feces is pale - Post-hepatic/cholestatic jaundice: obstruction of liver exits allows for some processing of unconjugated to conjugated bilirubin, but prevents conjugated form from moving out (into gut). Conjugated form can move into blood and embed into tissue membranes with unconjugated. Also moves to kidney where it causes orange color in urine. Feces are pale
169
Q

What influences cause deviations from HW equilibrium?

A
  • Genetic drift (non-small populations)- Selection (fitness is unequal in offspring)- Assortative mating (mating is non-random)- Population bottlenecks/founder effect
170
Q

Action of neomycin

A
  • 70 S ribosome inhibitor - Specifically causes mistranslation of codons
171
Q

Ehlers-Danlos Syndrome

A
  • AR and AD collagen disorder- Dominant forms of EDS are caused by mutations in the collage genes. Misfolded collagen molecules exert a dominant negative effect- Other forms of EDS are caused by mutations in enzymes required or the processing of collage molecules. As typical for enzyme defects, these mutations show a recessive mode of inheritance.
172
Q

What is the name of the molecule that synthesizes tRNA?

A
  • RNA Pol III
173
Q

In what cells does glucagon have a function? What response?

A
  • Liver: stimulates degradation of glycogen, stimulates gluconeogenesis from AA sources, stimulates energy production from AA sources - Adipose: stimulates lipolysis
174
Q

Regulation of purine salvage?

A
  • PRPP consumed by HGPRtase and APRtase - Less available for amidophosphoribosyltransfer, therefore less PRA formed therefore less de novo synthesis
175
Q

What is pernicious anemia?

A
  • Lack of intrinsic factor secreted by parietal cells of stomach
177
Q

What is the Bohr effect? What causes it to occur?

A
  • It refers to oxygen’s binding affinity of Hb being inversely related to acidity and concentration of CO2.
  • When CO2 concentration is high and acidic environment exists, o2 affinity for Hb is low and therefore Hb is more likely to give up o2 in tissues where this environment exists.
  • Causes: H+ binds to histidines foming stabilized T-form salt bridge; CO2 binds to N-terminus forming negatively charged carbamates increasing stabilization of T-form.
178
Q

Describe three processes that can lead to small insertions or deletions.

A

1.) Alignment out of registration, unequal crossover 2.) Strand slippage in replication as a result of repetitive sequences 3.) Intercalating agents causes slip bw stacked base pairs of DNA that distort helix and cause insertions or deletions

179
Q

Describe synthesis and sulfation of glycosaminoglycans.

A
  • Core proteins are manufactured in ER - Transferred to Golgi where glycosyltransferases add disaccharides to them. - Sulfotransferases add sulfate groups to them (can be unpredictable) - Exported in membrane-bound vesicle, finds linker and attached to polysaccharide backbone
180
Q

Examples of caretaker genes. Cancers resulting in defects from these genes

A
  • MSH2 or MLH1 code for DNA mismatch repair. Mutations lead to HNPCC: hereditary nonpolyposis colorectal carcinoma - BRCA1 or BRCA2 gene code for machinery involved in homologous recombination. Mutations lead to increased risk of breast/ovarian and cancer development. Unable to repair of double-stranded DNA breaks.
181
Q

Outline the steps involved in replication of DNA

A
  1. Origin recognition complex proteins bind to origins – ORC activated s phase, then inactivated until after mitosis 2. DNA helicase unwind the double helix 3. SSBPs bind DNA and prevent reformation of double helix 4. Tension in DNA acted on by topoisomerase I, which cuts phosphodiester bond of one strand and allows strand to rotate, religation occurs 5. Primase activity of DNA pol alpha uses exposes SS DNA region as template to synthesize a short complimentary stretch of RNA, provides 3’ OH 6. DNA pol alpha is replaced by delta or epsilon 7. Protein complex known as sliding clamp is loaded onto DNA, facilitates pol activity 8. DNA pol replicates continuously on leading strand, discontinuously (delta) on lagging strand 9. RNase activity removes primer, DNA pol possibly delta fills in gap left by primer removal 10. DNA ligase joins Okazaki fragments
182
Q

What changes occur that result in metHb?

A

-Fe3+ is the predominant form and there are no enzymes to oxidize back to Fe2+

184
Q

What are the effects of insulin, glucagon and epinephrine on glycolysis, glycogenolysis and glycogenesis in liver and muscle? Tabulate.

A
185
Q

From what are Asp, Asn, Arg, Glu, Gln and Pro made from?

A
  • alpha-ketoglutarate and oxaloacetate
187
Q

Contrast de-novo and maintenance methylation

A

De-novo DNA methylation- Methylation is introduced into an unmethylated strand of DNA by DNA methyltransferases DNMT3a and bMaintenance- Pattern of DNA methylation is maintained through mitosis by DNA methyltransferase DNMT1- During S-phase, DNA pol synthesizes a non-methylated strand from the methylated template strand- New double-strand consists of a methylated and non-methylated strands transiently. DNMT1 methylates the non-methylated strand based on what is seen on the template

189
Q

Most oxidized form of THF and most reduced form of THF? Which form accumulates within the body? In context of THF, what is the one carbon pool and what are the major contributors? Which THF form accumulates in body?

A
  • oxidized form: N10-formyl THF - reduced form: N5-methyl THF - THF can carry and move single carbon molecules around the body. - Major one carbon pool source = serine via 3-PG - Minor sources = formaldehyde (from methanol), formate (from tryptophan), histidine - Accumulated THF form: N5-methyl THF
190
Q

What apoproteins do HDL associate with? Where?

A
  • Nascent HDL associates with apoE,C and A in hepatocytes.
192
Q

How does one determine the value of pKa to predict what form(s) a weak acid will predominate at, at a given pH?

A

The value of the pKa can help determine which forms of the acid will be dominant at a certain pH. When the pH=pKa the concentrations of acid and base are equal. When pHpKa the base form dominates.

193
Q

Identify the first visible axis in the developing embryo

A
  • Anterior/posterior axis. This is defined by the primitive streak, which is defined by the position of the entry of the sperm into the egg. Node is the anterior end of the streak and the groove is the primitive streak and serves as the anterior/posterior axis.
194
Q

Explain the effect of insulin-induced dephosphorylation on glycogen phosphorylase and glycogen synthase.

A
  • Glycogen phosphorylase is inactivated - Glycogen synthase is activated
196
Q

Strategies used to lower ammonia levels in individuals with urea cycle defects

A

Protein restriction (not elimination d/t essential AA req or supply AA via alpha-ketoacids)
Hemodialysis (removes excess ammonia)
Phenylbutyrate (metabolite binds glutamine in water soluble form, which is excreted by kidneys) and benzoate (metabolite binds glycine in water soluble form, which is excreted by kidneys)
Mannitol (promotes diuresis)

198
Q

Sucrase-Isomaltase Deficiency

A
  • AR- Sucrose/glucose polymer intolerance
199
Q

Give epidemiology figures for congenital malformations

A
  • 2-3% of children are born with a recognizable birth defect- 20% of total mortality, birth defects are most common cause of infant death in US- Another 20% of infant deaths are caused by prematurity, which also can be considered a failure of normal development
201
Q

What is Zellweger syndrome?

A
  • Zellweger syndrome is a disorder of failure to produce functional peroxisomes and therefore failure to produce plasmalogens. Results in defective myelination, which leads to hypotonia, severe intellectual disability and is generally fatal in few few months of life. Infants present with poor weight gain, weak suck, hypotonia, high foreheads, flat broad nasal bridges and underdeveloped jaws.
202
Q

What signaling cascades to Gs, Gi and Gq affect?

A
  • Gs – stimulation of cAMP via adenylate cyclase
  • Gi – inhibition of cAMP via adenylate cyclase
  • Gq – stimulation of IP3/DAG via PLC
203
Q

What is the function of cyanide? What are treatments?

A
  • Cyanide inhibits cytochrome oxidase (complex IV) by binding Fe3+ and preventing reduction to Fe2+. It is fast acting and lethal. Causes lactic acidosis. Treatment: nitrites (oxidize Hb so cyanide binds to Hb instead of cytochrome oxidase) and Na thiosulfate (converts cyanide to non toxic SCN- form).
204
Q

How is gluconeogenesis in liver connected with muscle glycolysis?

A
  • Glucose enters glycolysis with pyruvate as end product - Pyruvate is converted into alanine via ALT and sent into bloodstream - Alanine is taken up by liver and converted back into pyruvate by ALT - Pyruvate is converted into glucose via gluconeogenesis
205
Q

What enzyme is defective in tyrosinemia-II? Symptoms?

A
  • tyrosine aminotransferase - Symptoms: keratitis, photophobia, skin lesions on palms and soles, intellectual disability
206
Q

Explain the process of FA desaturation. Why is this necessary? Where does it occur?

A
  • Many lipids in body are not saturated. - Desaturation of FAs occur in endoplasmic reticulum as shown here:
208
Q

Briefly explain the role of F26BP in gluconeogenesis and glycolysis.

A
  • High F26BP concentrations: glycolysis is stimulated, gluconeogenesis is inhibited - Low F26BP concentrations: glycolysis is inhibited; gluconeogenesis is stimulated
209
Q

What are branched-chain amino acids? Metabolism / Pathway of BCAA degradation? Products?

A
  • BCAA = LIV mnemonic = leu, iso, val - Metabolism: muscle has high levels of BCAT (branched-chain AA aminotransferase), which forms branched-chain alpha-ketoacids that are released into blood. BCKDH (branched-chain alpha-ketoacid DH) complex decarboxylates these BCAA in liver - Valine (see picture) - Isoleucine produces acetyl-CoA and propionyl-CoA, it is ketogenic and glucogenic AA - Leucine produces acetyl-CoA and acetoacetate, it is ketogenic AA only
210
Q

What is SCID? Causes?

A
  • Group of immune disorders impacting B and T cell proliferation - ~15% of cases due to adenosine deaminase (in degradation of AMP) where adenine metabolites build up - Model: accumulation of dATP inhibits ribonucleotide reductase blocking DNA replication and preventing proliferation OR adenine metabolites are toxic to lymphocytes
211
Q

Draw the glycolytic pathway. Include all the enzymes, where ATP is made or used and where reducing equivalents are produced.

A
213
Q

How does thymine nucleotide synthesis differ from the other pyrimidines?

A
  • Only make deoxyribose nucleotide forms
214
Q

Where are the following glucose transporters found? What are their functions?

A
  • SGLT1: found in small intestine, responsible for actively transporting glucose from lumen into intestinal epithelia. - GLUT1: found in all tissues, responsible for basal glucose uptake - GLUT2: found in liver, intestine and beta-cells of pancreas. In liver: removes glucose from blood. In intestine: releases glucose from epithelia into blood. In pancreas: regulates secretion of insulin. Also able to move fructose. - GLUT3: found in all tissues, responsible for basal glucose uptake - GLUT4: found in muscle and adipose tissue. Increases with endurance training, induced by insulin. - GLUT5: found in small intestine, responsible for uptake of fructose into epithelial cells and movement into blood serum.
215
Q

Describe degradation of bases?

A
  • Hypoxanthine + o2 = xanthine + H2o2 (ez: xanthine oxidase) - Guanine + H2o = xanthine + NH4+ (ez: guanine deaminase) - Xanthine + o2 = uric acid + H2o2 (ez: xanthine oxidase) - Uric acid eliminated in urine
216
Q

How can rifampicin kill certain bacteria yet have low toxicity towards human cells?

A
  • Used to treat mycobacterial infections such as TB and leprosy - It is a specific inhibitor of RNA pol found in many types of bacteria, but euk Pol II is not sensitive to it
218
Q

Describe role of chylomicron, where synthesized, association with apoproteins, source of apoproteins, destination of contents and formation / fate of chylomicron remnant.

A
220
Q

Glucose 6-phosphate dehydrogenase deficiency

A
  • XR- Sensitivity to H2o2 generating agents and fava beans
222
Q

Implication of epigenetic changes in T-Cells as implicated in SLE

A
  • SLE is caused by epigenetic changes in T-cells- This is an autoimmune disease with incidence = 1/2000 affecting females 8-10 times more than males- In this condition, antibodies against nuclear components are produced.- Global hypomethylation of the T-cell genome is seen in these patients- DNMT inhibitor treatment on T-cells causes SLE-like phenotypes
222
Q

Sickle Cell Anemia

A
  • AR- Hemolysis
223
Q

Describe the quaternary structure of hemoglobin. Compare and contrast the structure and oxygen binding properties of hemoglobin and myoglobin.

A

Hemoglobin is comprised of two alpha-globulin subunits and two beta-globulin subunits. Each has an iron containing heme prosthetic group. The hemoglobin forms by association of two alpha-beta-dimers which interact through H-bonds and salt bridges. Each subunit resembles that of myoglobin. Myoglobin has high affinity for o2 at low partial pressures of oxygen (hyperbolic o2 binding curve), whereas hemoglobin has a lower affinity for oxygen at lower partial pressures and its affinity increases as partial pressures increase and a conformational change takes place (sigmoid binding curve), changing affinity of each heme group that is to be “loaded.”

224
Q

What are Tyr and Cys made from?

A
  • From Phe and Met, which are both essential AAs
226
Q

What are the targets of nalidixic acid, cirprofloxacin and doxorubicin?

A
  • Nalidixic acid and ciprofloxacin act on bacterial topoisomerase II enzymes and function as antibiotics - Doxorubicin acts on human topoisomerase II and is used as an anticancer agent
227
Q

What are the products of the non-oxidative phase of the pentose phosphate pathway? Why are these important?

A
  • 2 F6P and 1 glyceraldehyde-3-phosphate - These are intermediates in the glycolytic / gluconeogenic pathways and can be synthesized from pentoses depending on the need of the cell.
228
Q

Explain the function of boundary elements

A
  • Boundary elements aka chromatin barriers serve to separate active and inactive genomic regions
230
Q

Contrast the effects of gain-of-function and loss-of-function mutations in the RET gene

A
  • RET gene encodes tyrosine kinase receptor located in the plasma membrane. Both mutations below are autosomal dominant.- Loss of function mutation in RET gene causes Hirschprung disease. Receptor has inability to respond to stimulus. In this disease, there is impaired development of neurons that populate the colon giving rise to aganglionic colon.- Gain of function mutation in RET gene causes Multiple Endocrine Neoplasia (MEN). Receptor renders signaling molecule constitutively active. Proliferation of neuroendocrine cells occur.
231
Q

Ketogenic AAs?

A

L starting AAs – leucine, lysine

232
Q

T2D patients present with high serum LDL even when serum glucose is well controlled. Where does this lipid abnormality originate? Why does it occur?

A
  • Type 2 diabetics are insulin insensitive - As a result of insulin signaling stating to the body that glucose is in abundance and should be taken up, fatty acids are still being released from adipose tissue and the liver has to repackage large amounts of these, which in turn means that LDL levels are high.
233
Q

Three amino acids used to synthesize glutathione? Where synthesized? Why is it such a stable molecule?

A
  • Glu, Cys and Gly - Made in liver (not on ribosomes) - Contains non-regular peptide bond called gamma-glutamyl linkage that is stable and resistant against proteases
235
Q

Charcot Marie Tooth Disease

A
  • Congenital chaperone defects cause protein folding disorder
237
Q

The Km for a substrate often corresponds closely to metabolic concentrations of the substrate. Why is this advantageous?

A
  • When changes to substrate concentration occur, enzyme is sensitive enough to decrease reaction rate or increase reaction rate.
238
Q

What is ALT?

A
  • alanine aminotransferase - synthesizes alanine and alpha-ketoglutarate from pyruvate and glutamate
239
Q

Explain how cell migration is necessary for the development of the cerebral cortex

A
  • Development of CNS begins from neural tube. Neuronal stem cells on ventricular side of neural tube divide and generate neuronal precursor cells- These precursor cells have to migrate outward from the ventricle along a scaffold of glial cells, which occurs in waves- Mutation / deletion of LIS1 gene interferes with orderly pattern of migration. Result = cerebral cortex is thickened and lacks defined cell layers. Lissencephaly = smooth brain. Individuals suffer from severe mental retardation.
240
Q

Describe and identify the reactions catalyzed by the following classes of enzymes: oxidoreductases, transferases, hydrolases, lyases, isomerases and ligases.

A
  • a.) oxidoreductases: class I: catalyze redox reactions, involve electron acceptors / donors incl. oxidases, oxygenases, reductases, dehydrogenases - b.) transferases: class II: transfer groups (carboxyl, amino, glucosyl, phosphoryl, methyl etc.) from one substrate to another without the input of energy incl. kinases, aminotransferases, carboxylases, methyltransferases - c.) hydrolases: class III: cleave bonds by addition of water incl. glucosidases, ATPases, phosphatases, peptidases, lipases - d.) lyases: class IV: break bonds without addition of water or oxidative cleavage - e.) isomerases: class V: change steoreochemistry of optical or geometric isomers incl. epimerases, mutases, racemases - f.) ligases: class VI: catalyze formation of bonds via input of energy (usually via hydrolysis of XTP), not via transfer to bond incl. ligases, synthases, synthetases, carboxylases
242
Q

What is an irreversible inhibitor? Give a common example.

A
  • Irreversible inhibitors chemically modify and inactivate enzymes by forming covalent complexes at the active site residues. Aspirin is an example and it is a prostaglandin synthase inhibitor.
243
Q

What is Maple Syrup urine disease? Causes? Symptoms? Result? Treatment?

A
  • Genetic defect in BCKDH that leads to accumulation of BC alpha-ketoacids in body - Symptoms: poor feeding, vomiting, slow/irregular breathing, ketoacidosis, hypoglycemia and neurological dysfunction - High incidence in old order menonite - Result: fatal in one week unless treatment - Treatment: reduced BCAA in diet and monitoring of serum BCAA levels
243
Q

Describe regulation of transcription in response to thyroid hormone

A
  • Thyroid hormone becomes bound to receptors that are constitutively in the nucleus and are bound to DNA elements of genes that respond to it - THR (receptor) when not bound to thyroid hormone is dimerized with RXR (retinoid X receptor) - This dimer recruits HDAC, which condenses chromatin and inhibits transcription - Binding of thyroid hormone triggers conformation change in THR, HDAC is released and HAT binds in its place, leads to relaxation of chromatin and enhancement of transcription - Response: stimulated metabolic rate in most tissues
245
Q

What is pellagra?

A
  • Deficiency in niacin, leading to diarrhea, dementia, dermatitis and death.
246
Q

What is the function of diacylglycerol acyltransferase (DGAT) inhibitors?

A
  • Inhibits synthesis of triacylglycerol
247
Q

What is scurvy?

A

-Scurvy is the result of vit C deficiency. Blood vessels, tendons and skin become fragile. Symptoms include malaise, lethargy, easy bruising, dry skin, loosening of teeth, sleep disturbances and leg pain.

249
Q

Describe the synthesis of FAs.

A
250
Q

What are examples of hormones that function through the cAMP signal transduction pathway? Function of hormone?

A
  • Epinephrine (function: glycogenolysis in muscle, fat hydrolysis in adipose, increased HR and FOC) - Glucagon (function: glycogenolysis in liver, fat hydrolysis in adipose) - AVP/ADH/vasopressin (function: water re-absorption in kidney)
252
Q

How do gallstones form?

A
  • Bile contains cholesterol, bile salts and phospholipids. These aid in emulsification of dietary fats. Cholesterol is prone to precipitating in gallbladder and duct works as its water solubility is low. When in high concentration, it particularly precipitates out causing formation of cholesterol stones known as gallstones, which can obstruct the gallbladder and ducts causing painful backup of bile of pancreatic enzymes.
254
Q

To what AA residue does 2,3-BPG bind on Hb? Result.

A

-His. Causes stabilization of T-form.

255
Q

Action of diphtheria toxin

A
  • protein produced by C. diphtheriae that inactivates EF-2 by ADP ribosylation, preventing elongation
257
Q

Roles of nucleoside 5 monophosphate and nucleoside disphosphate kinases?

A
  • Nucleoside 5 monophophate kinases: XMP + XTP = 2 XDP - Nucleoside disphosphate kinases: XDP + XTP = XTP + XDP
258
Q

Action of streptomycin

A
  • 70 S ribosome inhibitor - Specifically binds to small subunit (30 S) and inhibits initiation and causes mistranslation of codons
259
Q

Which pyrimidine degradation product is unique to pyrimidine metabolism? How is this important?

A
  • Beta-aminoisobutyrate - Found in urine of cancer pts undergoing radiation or chemotherapy
260
Q

Synthesis of proline

A
262
Q

Explain the gender-specific differences in recurrence risk for multifactorial disorders such as pyloric stenosis

A
  • Pyloric stenosis is more common in male birth 1/200 than in female birth 1/1000. Accordingly, in pyloric stenosis, there is a lower threshold for males (males need few contributing alleles) and a higher threshold for females (females need more contributing alleles).- When assessing recurrence risk then: affected female proband has more contributing alleles than affected male proband and therefore has higher risk of having an affected sibling than does a male proband. Also her risk of having an affected brother is higher than her having an affected sister because a brother would only need fewer contributing alleles to develop a disease.- Affected male proband has higher recurrence risk for having a brother than a sister with pyloric stenosis.
263
Q

What are two important B vitamins and what are their roles in metabolism?

A
  • Niacin (aka B3): part of structure of NAD+ (oxidized and reduced) and NADP+ (oxidized and reduced). - Riboflavin (aka B2): part of structure of FAD and FMN (reduced and oxidized forms).
264
Q

Explain how Viagra works.

A
  • Viagra is a phosphodiesterase inhibitor and prevents deactivation of cGMP, allowing their levels to remain elevated for longer. cGMP is what binds to cGMP-dependent protein kinases that cause smooth muscle to relaxed. As a result, increase in blood flow to the penis is prolonged, treating erectile dysfunction.
265
Q

Give examples of diseases that affect genetically isolated populations

A

1.) Ellis van Creveld Syndrome: Old Order Amish of Lancaster County2.) Tay-Sachs Disease (ganglioside storage disease): Ashkenazi Jews3.) Tyrosinema: French Canadians in Quebec

267
Q

Describe metabolism of early fast.

A
267
Q

Action of gentamicin

A
  • 70 S ribosome inhibitor - Specifically causes mistranslation of codons
268
Q

Compare the impact of problems during the first 4 weeks, weeks 5 to 8 and in later stages of development

A

1.) 1-4 weeks (blastogenesis): abnormalities during this period produces multiple major abnormaities in entire embryonic regions. Example = VACTERL2.) 5-8 weeks (organogenesis): abnormalities during this period affects specific organs and produces single major anomalies. Example = congenital heart defects3.) 9+ weeks: major organs have formed, abnormalities here will have mild effects on the individual. Example = single palmar crease

269
Q

Outline salvage of purine nucleosides

A
  • Adenosine + ATP = AMP + ADP (ez: adenosine kinase) - No salvage of guanosine
271
Q

What does CO2 bind to on Hb?

A

-N-terminus. Causes formation of carbamate, forms salt bridge which stabilizes T-form.

271
Q

Asparagine synthesis

A
272
Q

Give general prevalence figures for common multifactorial birth defects

A
  • Risk for general population = 0.5% risk- If second degree relative affected = 0.7-2% risk- If first degree relative affected = 3-4% risk- If two first degree relatives affected = 5-8% risk- Three first degree relatives affected = 9-12% risk- Identical twin = 20-30% risk
274
Q

PKU. Inheritance, defect, frequency

A
  • AR- IEM, Defect in Phe hydroxylase, Phe accumulates and damages the CNS. With many other IEM, screened for at birth- 1/2900
275
Q

What are the function of leukotrienes?

A
  • promotion of SM contraction - anaphylaxis
276
Q

Precursor to GABA, how is it derived? What are cofactor requirements? Use?

A
  • Glutamate - Decarboxylation of glutamate forms GABA - Requires pyridoxal phosphate - Neurotransmitter
277
Q

Outline salvage of purine bases

A
  • Hypoxanthine + guanine + PRPP = IMP / GMP (ez: HGPRtase) - Adenine + PRPP = AMP (ez: APRtase)
278
Q

What contributes to color of feces? Urine? Why?

A
  • BDG (conjugated bilirubin) moves into gut in bile. It is converted to bilirubin and urobilinogen by bacteria. - Urobilinogen can be further converted into urobilins and stercobilins in gut and gives color to feces - Urobilinogen can also be absorbed into blood and converted to urobilin in blood stream, being moved to and excreted from kidneys.
280
Q

Describe de novo synthesis of purines

A
  • PRPP = 5 phosphoribosyl 1 pyrophosphate - Ribose 5 phosphate + ATP = PRPP (ez: PRPP synthetase) - PRPP + gln = PRA (phosphoribosylamine) + PPi (ez: amidophosphoribosyltransferase) - 9 step process to convert PRA to IMP (4 reactions require ATP, C and N donated from gly, gln, asp, CO2 and N10-formyl THF). IMP = inosine monophosphate - IMP precursor for AMP and GMP via following rxns: - IMP + Asp + GTP = adenylosuccinate + GDP + Pi (ez: adenylosuccinate synthetase) - Adenylosuccinate = AMP + fumarate (ez: adenylosuccinase) - IMP + NAD = xanthosine5monophosphate (ez: IMP dehydrogenase) - XMP + Gln + ATP = GMP + glu + AMP (ez: GMP synthase) - Specific Nucleoside monophosphate kinases (convert XMP to XDP) and diphosphate kinases (XDP to XTP)
281
Q

What enzyme is defective in phenylketonuria? Symptoms?

A
  • phenylalanine hydroxylase - Symptoms: intellectual disability, recurrent seizures, hypopigmentation, eczematous skin rashes
283
Q

Describe N-linked glycosylation mechanism

A
  • Dolichol phosphate in ER membrane acts as site for oligosaccharide in ER - Glycosyltransferase adds two GlcNAcs to dolichol - Glycosyltransferase adds five mannose - Dolichol phosphate linked to above CHOs reorientates from cytoplasm into ER lumen - 4 more mannoses are added onto oligosaccharide using dolicholphosphomannose - 3 glucoses are added to mannose forming universal oligosaccharide containing 14 sugars - Highly specific modification of universal oligosaccharide occurs in Golgi apparatus by addition or removal of CHOs, yielding high mannose type or complex type (sialic acid, fucose, N-acetyl glucosamine, N-acetyl-galactosamine, galactose)
284
Q

Explain how masculinization of female babies (male pseudohermaphroditism) and feminization of male babies (female pseudohermaphroditism) occurs

A

1.) Masculinization of female babies: normal ovaries, but ambiguous or male genitalia- Congenital adrenal hyperplasia (most common, defect in 21-hydroxylase involved in cortisol biosynthesis results in block in cortisol synthesis with intermediates being shunted into androgen synthesis pathway). As result: females have high levels of androgens and develop ambiguous or male genitalia2.) Feminization of male babies: failure to develop unambiguous male genitalia- Defect in testes development during embryogenesis- Problem in androgen biosynthesis by testes (eg. Issue with steroid 5 alpha reductase) - Deficiency in androgren receptor production or signaling by target cells (androgen insensitivity)

285
Q

How is entry into mitosis coupled to completion of DNA replication?

A
  • Completion of DNA replication activates protein phosphatase (cdc25C) - Protein phosphatase dephosphorylates a particular tyrosine residue in CDK1, which activates cyclin A or B-CDK1 complexes - Cyclin A or B-CDK1 complexes phosphorylate many proteins that participate in early mitotic events including chromosome condensation, assembly of mitotic spindle and breakdown of nuclear membrane
287
Q

Outline the process of strand-directed mismatch repair.

A
  • Newly synthesized DNA contains nicks, which distinguishes parental and daughter strands - Base mismatch distort DNA helical structure - Repair enzymes recognize distortion and remove mismatched base, excise nucleotides back to nearest nick - Gap fill by DNA pol using parental DNA strand as template
288
Q

Characteristics of dominant and recessive inheritance patterns

A

1.) Recessive inheritance: one normal allele prevents disease, recessive inheritance is mostly observed in defects of enzymes/proteins involved in transport and storage, loss of one functional allele can be compromised2.) Dominant inheritance: mostly observed in defects of structural proteins, proteins involved in growth, differentiation and development and receptor/signaling proteins

289
Q

What is DNP? What is its effect? What symptoms does it cause?

A
  • It is a pesticide and poison that uncouples ETC/ox-phos. It causes sweating, flushing, nausea, inc RR, tachycardia, fever, coma, death in 1-2 days. Treatment with ice baths, oxygen and fluid/electrolyte replacement.
290
Q

How are ribonucleotides converted into deoxyribonucleotides?

A
  • Ribonucleotides must be in diphosphate form - Enzymes needed : ribonucleotide reductase and thioredoxin reductase. - Cofactors: thioredoxin - Thioredoxin reductase uses NADPH to generate reduced thioredoxin for another rxn to occur
291
Q

What pentose phosphate pathway deficiency(ies) seen in human? What is/are the clinical presentation(s)?

A
  • Beriberi: result of thiamine deficiency that in addition to affecting PDH and alphaKGDH, also affects the transketolase reactions of the non-oxidative phase of the PPPathway preventing rearrangement of carbon structures and leading to an imbalance in the CHO pool. Prevalent in low income communities, but most especially in alcoholic patients. Alcoholism reduces thiamine uptake and storage.
293
Q

What is polyadenylation? Describe. What types of RNA molecule are polyadenylated?

A
  • Polyadenylation = addition of 100s of adenine residues to end of mRNA moledule - Stop pt for transcription is specified by a consensus seq in DNA known as polyadenylation signal, which is transcribed into pre-mRNA - This signal is recognized by specific protein factors - This tail is important for RNA stability
295
Q

Describe repair of a pyrimidine dimer by process of nucleotide excision repair.

A
  • Helicases and nucleases work together to peel off region containing thymidine dimer lesion - DNA Pol and DNA ligase fill in gap
296
Q

Outline mechanism of action of the chemotherapeutic agents cytarabine, cyclophosphamide, and doxorubicin.

A

1.) Cytarabine is an analogue of cytidine, contains arabinose in place of ribose. Converted to cytarabine triphosphate. Competes with dNTPs for binding to DNA polymerase 2.) Cyclophosphamide is converted to phosphoramide mustard by liver. It is a bi-functional alkylating agent that forms inter and intra strand DNA crosslinks that block replication and can trigger apoptosis 3.) Doxorubicin form tripartite complex with DNA and topo II. Topo II introduces D-S breaks in DNA. Binding doxorubicin prevents re-ligation of D-S breaks. Breaks accumulate and trigger apoptosis

297
Q

Gluco-/ketogenic AAs?

A

TIPhe = try, trp, tyr, iso and phe

297
Q

Why are double-strand DNA breaks particularly troublesome? What two avenues of repair are available? Explain what each does.

A
  • No intact template / parental strand to direct repair - Nonhomologous end-joining: broken ends processed, loss of nucleotides at breakpoint, brought together, ligated, ends with permanent change in DNA sequence - Homologous end-joining: repair of damaged chromosome using information contained in undamaged, homologous chromosomes, allows repair of d-s breaks without loss of information
299
Q

Explain mechanism that brings acetyl-CoA to cytoplasm from mitochondrion that preceeds FA synthesis?

A
300
Q

What is assortative mating and how does it affect the gene pool of a population?

A
  • Assortative mating is the selection of partners based on specific genetic traits.- It disturbs the equilibrium distribution of alleles, specifically, the mating of genetically similar individuals increases the degree of homozygosity in a population. The allele frequencies in the gene pool don’t change, only change is increase in homozygote frequency.
302
Q

Describe role of VLDL, IDL and LDL. Include where each are synthesized/formed, association with apoproteins, source of apoproteins, destination of contents and fate of lipoprotein.

A
302
Q

What is I-cell disease? Describe the defect

A
  • Surface mannose residues are not phosphorylated and therefore lysosomal proteins don’t reach the correct compartment and appear in serum - Result: lysosomal degradation of proteins and CHOs is impaired, inclusion bodies are seen in lysosomes making them appear dense
303
Q

Explain the function and regulation of HSL.

A
  • HSL=hormone sensitive lipase - In adipose tissue, HSL releases FFAs into blood by cleaving TAGs into glycerol and FAs - Inhibited by insulin
305
Q

Synthesis of serine

A
305
Q

Why is phosphorylation of Rb such as crucial event in cell cycle progression? Which kinases phosphorylate Rb?

A
  • Phosphorylation of Rb in Rb:E2F by cyclin D:CDK4/6 causes dissociation of Rb from E2F - E2F is a transcriptional activator of cyclin E:CDK2 - In the absence of growth factors, Rb phosphorylation can still occur independent of cyclin D:CDK4/6 and cell cycle can progress
306
Q

Contrast isolated anomalies, sequences and syndromes

A
  • Isolated anomalies: single problem that can be sporadic or multifactorial in order that just affects a single body region. Ex: cleft palate. Major anomalies = anomalies requiring surgical or cosmetic consequences. Minor anomalies = little impact on well-being of pt – can give diagnostic clues about presence of a syndrome.- Sequences: cascade of events, starting from isolated anomaly and leading to multiple malformations- Syndromes: affect several body regions and often display chromosomal or Mendelian inheritance.
308
Q

Describe the factors that determine protein half-life

A
  • Conformation: eg. Misfolding results in hydrophobic domains being placed on surface and leads to degradation - N-terminus: Ser/Met proteins are more stable than Arg/Lys proteins - Other sequences: eg. PEST sequences shorten lifespan
309
Q

What is the glycerol phosphate shuttle system? Where does it occur? Draw it.

A
  • It is a shuttle mechanism to move electrons from NADH in cytoplasm into the mitochondrion. It occurs in brain and muscle.
310
Q

Explain transportation from cytoplasm to nucleus

A
  • Ran-GTP bound to importin = inactivated Importin - Ran hydrolyzes GTP to GDP and importin dissociates, Ran-GDP moves into nucleus - Importin binds cargo and moves through nuclear pore complexes into nucleus - Ran-GDP in nucleus binds GTP and dislocates GDP - Importin dissociates from cargo in nucleus and associates with Ran-GTP moves out to cytoplasm
312
Q

What does the formation of hydroxyproline and hydroxylysine require?

A

-Fe2+, ascorbate and alpha-ketoglutarate in addition to enzyme.

314
Q

What are some common causes of metabolic acidosis?

A

Renal failure, methanol poisoning, DKA

316
Q

What reaction does cholera toxin catalyze? What reaction does pertussis toxin catalyze? What g-protein subtypes are implicated? What is the result?

A
  • Both toxins catalyze ADP-ribosylation, which is the attachment of ADP-ribose to specific residues. - Cholera toxin catalyzes this reaction in Gs alpha subunit proteins, causing subunit to remain in active state, leading to high adenylate cyclase and cAMP levels. - Pertussis toxin catalyzes this reaction in Gi alpha subunit proteins, causing subunit to remain in inactive state, leading to high adenylate cyclase and cAMP levels.
318
Q

Describe the typical layout of a gene that is transcribed by RNA polymerase II

A
  • Enhancer (way upstream) - Gene-specific elements: DNA sequences unique to particular genes or groups of genes - CAAT box or GC-rich region: may be present - TATA box: almost invariably found in genes transcribed by Pol II - Gene
319
Q

Describe the function of HP1 in transcriptional control

A
  • De-acetylated histones are methylated and bind HP1 proteins- HP1 proteins bind histone methylase- This binding results in methylation of histones spreadings along chromosomes until boundary elements are reached.
320
Q

Explain the regulation of translation

A
  • Points of regulation are at 1.) recognition of start codon and 2.) activity of initiation factors 1.) Recognition of start codon: bind of regulatory protein in 5’ UTR can mask start codon 2.) eIF-2a can be inactivated by phosphorylation
321
Q

Explain the difference between the T and R form of Hb.

A
  • T-form refers to the tight form of Hb and occurs when Hb is deoxygenated. In this form, Hb does not have high affinity for o2. At the tissue level where low pH, high CO2 and high 2,3-BPG are present, the tight form is favored and o2 is offloaded. - R-form refers to relaxed form of Hb when it has a high affinity for o2 molecules. This occurs in alveoli where high pH, low CO2 and low 2,3-BPG are present.
322
Q

What signal transduction mechanism is most commonly utilized by the receptor for glucagon?

A
  • cAMP signaling pathway, which leads to glycogenolysis.
324
Q

Describe chromosomal defects in cri-du-chat

A
  • Deletion in chromosome 5 p. Usually a new mutation- Facial changes: microcephaly, hypertelorism (wide set eyes), micrognathia (undersized jaw). Brain/CNS changes: severe mental retardation. Cardiovascular: heart defects. Characteristic cat-like cry.
325
Q

Treatment of argininosuccinate synthetase deficiency

A
  • As a result of this deficiency – citrullinemia - Treat with arginine and phenylbutyrate supplements - Cirtruline can be cleared directly, but only carries single nitrogen
327
Q

How is adenylate cyclase activated? Deactivated? Inactivated?

A
  • Adenylate cyclase is activated by Gs proteins. Deactivated when Gs proteins are inactivated by their GTPase or when Gi proteins bind adenylate cyclase.
327
Q

Explain what reaction(s) the cell can perform when there is a need for ribose, but not for NADPH. ATP is not required.

A
328
Q

Action of chloramphenicol

A
  • 70 S ribosome inhibitor - Specifically prevents peptidyl bond formation
330
Q

Describe defects in heme synthesis

A
  • AIP: acute intermittent porphyria: deficiency in PBG deaminase, causes build up of PBG and ALA. Give urine a dark red color. Life threatening and causes episodes of confusion and sharp abdominal pain - PCT: porphyria cutanea tarda: deficiency in uroporphyrinogen decarboxylase, leads to buildup of porphyrins (detected in urine). Porphyrins able to absorb visible and UV light. As a result, light energy is builtup and discharged into tissue and generates ROS. As a consequence, PCT causes photosensitivity of skin and leads to blistering of sun-exposed areas.
331
Q

What is genetic drift and how does it affect the gene pool of a population? Example

A
  • Genetic drift is the change in allele frequency within a population due to random sampling/statistical variation. This occurs in small populations. - Example: 10 couples stranded on deserted island. One of males is heterozygous carrier of CFTR mutation. Allele frequency is 1/40 (2.5%). Time goes by and couples have average of 2 children. By chance, none of children have inherited mutation from father. By genetic drift, CFTR allele is now completely lost from this island’s population.
333
Q

Main source of glutamate is the diet. How else is it synthesized in the body?

A
  • Transamination via aminotransferase enzyme (ALT / AST) - Deamidation of glutamine via glutaminase
334
Q

Describe how arachidonic acid is synthesized and inserted into phospholipids.

A
335
Q

Describe how xenobiotics are detoxified.

A
  • Example: acetaminophen. - UGT catalyzes conjugation of UDP-glucuronate with acetaminophen, making it water soluble, allowing it to be excreted in urine
337
Q

Describe protein digestion in the digestive tract

A
  • Acidic pH of stomach denatures proteins - Pepsinogen is secreted by the stomach - It autoinhibits it’s active site, becomes altered in acidic environment and cleaves itself into pepsin - Pepsin’s active site contains aspartic acid residues. - It is a endopeptidase and cleaves internal peptide bonds, prefers bonds formed between AA groups of aromatic and hydrophobic amino acids - Chyme enters duodenum, stimulates release of secretin and CCK by intestinal mucosa - Secretin: stimulates acinar cells in pancreas = alkaline fluid released (bicarb rich), which neutralizes chym - CCK: stimulates release of bile and release of pancreatic digestive zymogens (trypsinogen, chymotrypsinogen, proelastase, procarboxypeptidases) - Enteropeptidase present on wall of intestinal mucosa cleaves trypsinogen to trypsin - Trypsin cleaves remaining pancreatic zymogens into active forms (chymotrypsin, elastase, carboxypeptidases)
338
Q

What is meant by the capping of pre-mRNA? Is the cap unique to pre-mRNA? Function?

A
  • Methylguanosine is added to the 5’ end of pre-RNA - This distinguishes mRNA from other types of RNA - Required for subsequent processing steps - Protects against degradation - Important for initiation of translation
340
Q

Implication of pyrimidine salvage pathway?

A
  • PRPP consumptions means less PRPP available to stimulate CPS II, orotate phosphoribosyltransferase, therefore de novo pyrmidine synthesis is decreased
342
Q

What is the committed step in ketone body formation?

A
  • Conversion of HMG-CoA to Acetoacetate via HMG-CoA lyase, which produces and acetyl-CoA. Procession through this step will commit the pathway to forming ketone bodies.
344
Q

Describe the synthesis of cholesterol from farnesyl PP.

A
345
Q

What are the products of the oxidative phase of the pentose phosphate pathway?

A
  • Ribose-5-phosphate, 2 NADPH and CO2
347
Q

Which AAs are converted into pyruvate?

A
  • Mnemonic: Some Good Children are Pyrates - Serine, Glycine, Cysteine, Alanine = Pyruvate
348
Q

What is locus heterogeneity?

A
  • Mutations in different genes cause the same phenotype
349
Q

Describe what happens at a molecular level when oxygen binds to a heme in deoxygenated hemoglobin. How do these events contribute to allosterism? How does the oxygen-binding behavior of hemoglobin translate into an efficient oxygen carrier?

A

Deoxygenated Hb is in a T-form (tight) with low o2 affinity. A valine reside partially blocks the o2 binding site in the beta subunits. When o2 concentration increases (ie. in lungs), o2 is forced by mere concentration onto alpha subunits. A conformational change occurs as a result of this binding and the valine residues are rotated out of the o2 binding site in the beta subunits. The Hb molecule is now in a high o2 affinity form known as the R-form (relaxed). The importance of this is considered when looking at the physiological range when o2 is loaded / unloaded from Hb.

350
Q

Explain what an irreversible inhibitor is?

A
  • An irreversible inhibitor is one that forms covalent complexes with active site residues in enzymes, chemically modifying and therefore inactivating an enzyme. No change occurs with increased substrate concentration, nor removal of inhibitor.
351
Q

Which molecules are inducers of covalent modifications (aka phosphorylations)? Signals and affects?

A
352
Q

Describe important of asymmetric distribution of lipids between lipid bilayers. What is typical distribution? Implication in RBCs

A
  • Outer leaflet is rich in PC and sphingomyelin (both have choline) - Inner leaflet is rich in PE and PS - Stress activates enzymes known as flippases that flips lipids between leaflets - Eg. When PS appears in outer leaflet of RBCs, RBCs are destroyed by macrophages.
353
Q

What is selection and how does it affect the gene pool of a population?

A
  • Selection works against or in favor of certain genotypes. Over time, selection will reduce number of detrimental mutant alleles in a population. However, frequency of mutant alleles will stabilize at a low level. In this state of equilibrium, loss of mutant alleles by negative selection will be equal to spontaneous occurrence of new mutant alleles
354
Q

Describe molecule basis for PKU

A
  • Inability to convert phenylalanine to tyrosine due to mutation often at 5’ splice donor site in phenylalanine hydroxylase gene - Results in defective splicing, protein produced lacks one exon and is rapidly degraded
355
Q

Synthesis of tyrosine

A
357
Q

Describe pathology of familial hypercholesterolemia. What is the presentation?

A
  • Autosomal dominant disorder resulting from defects in LDL receptor. As a result, serum lipid concentrations are elevated and result in high levels of LDL in the blood serum. As a result of the overflow pathway into macrophages, atherosclerotic changes in blood vessels occur leading to higher incidences of CVD in the 3rd and 4th decades of life. - These individuals present with xanthomas (deposition of lipis) to joints and tendons. Lab studies indicate elevelated cholesterol and triglyceride levels.
358
Q

What are the products, substrates, cofactors of phenylalanine hydroxylase?

A
  • substrates: o2, THBtn, phenylalanine - products: tyrosine, H2o and DHBtn - cofactors: DHBtn reductase, which requires NADH
359
Q

Explain how a cell changes shape and polarity. Provide example using developing kidney

A
  • In development, cells must respond to environmental clues by changing their shape and polarity, which involves rearranging cytoskeleton and polarizing secretion of proteins to apical or basal surfaces of cell- Kidney: Epithelial cells need to sense fluid flow in kidney tubule. Detection of fluid stream leads to stopping of cell proliferation and correct polarization of cell. This occurs by relocating erb-b2/EGFR to basal surface. In polycystic ovary disease, mutations to two genes polycystin 1 and 2 render a cell incapable of sensing fluid flow. As a result, cell proliferation doesn’t stop, development of polarity doesn’t occur (erb-b2/EGFR) remains on the apical surface and kidney develop cysts.
361
Q

What reaction does galactokinase catalyze?

A
  • Galactose + ATP =(ez: galactokinase) Galactose-1-P
362
Q

What is ataxia telangiectasia?

A
  • Rare inherited disorder resulting from mutations in gene that encodes ATM kinase - Symptoms: difficulty in coordinating movement (ataxia) and appearance in small, widened blood vessels on skin (telangiectasia) - Pts susceptible to infections and at increased risk of leukemias and lymphomas. They are highly sensitive to radiation exposure.
364
Q

What is the purpose of the pentose phosphate pathway? What are the products from this pathway used for?

A
  • Way for generating pentoses and NADPH - Pentoses are used for synthesis of: DNA, RNA, ATP, NADH, FADH, Coenzyme A - NADPH are used for synthesis of: FAs, cholesterol, NTs, nucleotides and reduction of oxidized glutathione and P450 monooxygenases
366
Q

Define haploinsufficiency

A
  • Haploinsufficiency: half of gene dosage might not be sufficient for a cell to carry out its function. Many structural proteins are needed in quantities too large to be supplied by just one allele
368
Q

Describe the composition of lipid droplets?

A
  • Lipid droplets are packages containing TAGs. These are surrounding by a protein coat composed of many proteins including perilipin. Perilipin acts as protector of the TAGs stores and prevents lipases, such as hormone sensitive lipases from degrading TAGs into FFA and glycerol. When perilipin is phosphorylated, it allows lipases to do this, but not when dephosphorylated.
370
Q

What is CML and what is the biochemistry behind it? What gene fusion is generated? What is the biochemical activity associated with the fusion protein? Treatment?

A
  • CML = chronic myelogenous leukemia - Result of translocation bw c/s 9 and 22, fuses the ABL gene usually on 9 to the BCR gene on 22, creating Philadelphia chromosome BCR-ABL on 22 - Result = constitutively active Bcr-Abl kinase can phosphorylate many targes. In hematopoietic cells, stimulates expansion of white blood cells - Treatment = imatinib mesylate (Gleevec) is specific inhibitor of this kinase
371
Q

List disorders of phenylalanine and tyrosine degradation. Name enzymes.

A
  • Phenylketonuria: deficiency in phenylalanine hydroxylase - Tyrosinemia-II: deficiency in tyrosine aminotransferase - Alcaptonuria: deficiency in homogentisate oxidase - Tyrosinemia-I: deficiency in fumarylacetoacetate hydrolase
373
Q

What is xeroderma pigmentosum and what cause it?

A
  • Extreme sensitivity of the skin to sunlight - Pigmentation changes, skin cancers, develop on areas exposed to sun - Result from any of several mutations that impair or abolish the nucleotide excision repair
374
Q

How is glycerol (or G3P) synthesized? What is this a precursor to?

A
  • Glycerol-phosphate-dehydrogenase takes DHAP and converts it into glycerol-3-phosphate, which is precursor to TAGs.
375
Q

What molecules are regulators of gene expression? Signals? Affects?

A
377
Q

Describe the effect of arsenic on glycolysis.

A
  • Arsenic mimics phosphate. In its presence, Glyceraldehyde-3-phosphate DH reaction yields arsenic product instead of 13BPG. The arsenic product is unstable and hydrolyzes to 3PG. No ATP is gained from the process, where previously the conversion to 3PG yields a single ATP. Net ATP from glycolysis with one molecule of glucose is 0. This is problematic for RBCs who rely solely on glycolysis for their energy production. - Pts present with SOB and dizziness. CBC reveals hemolytic anemia and elevated urine arsenic levels. Treatment includes discontinuation of arsenic and therapeutic red-cell exchange.
378
Q

What are some common causes of respiratory acidosis?

A

Asthma, bronchitis, obesity, sedatives / anesthetics

379
Q

Symptoms of imborn errors of creatine metabolism. Treatment?

A
  • Children present with hypotonia, little to no skeletal or cardiac muscle pathology - Neurological symptoms: epileptic seizures, intellectual disability - Treated by creatine administration
380
Q

Describe synthesis of succinyl-CoA from AAs.

A
381
Q

What is familial nonpolyposis colorectal carcinoma (HNPCC; Lynch syndrome)? Which genes are generally mutated in HNPCC? Which DNA repair pathway is impacted?

A
  • Hereditary susceptibility to colon cancer, 80% lifetime risk of colorectal carcinoma - Arises from defect in DNA mismatch repair (mutations to MSH2 or MLH1) genes - Pt inherits one defective allele from parent, loss or inactivation of second copy results in accumulation of mutations at accelerated rate typically in microsatellite repeats, which means microsatellite instability occurs - Microsatellite instability can reduce Bax protein function, which suppresses apoptosis and abnormal cells survive
382
Q

Discuss pre-mRNA splicing.

A
  • Catalyzed by complex known as spliceosome, which has protein and snRNA compoments involved in catalysis and recognizing exon/intron boundaries - Consensus nt seqs are required for splicing: 5’ splice site, 3’ splice site and branch point within intron - Adenine at branch point attacks 5’ splice site, cutting sugar-phosphate backbone of RNA - 5’ end becomes linked to adenine, forms a loop - Free 3’OH reacts with 5’ end of next exon, joins. Intron released as lariat
383
Q

Describe Duschenne/Becker Muscular Dystrophies

A
  • Both common x-linked recessive diseases - Affects 1/3000 - Defect in dystrophin leads to muscle damage. Gene is large target for new mutations. Large target - Pts are wheel-chair bound by age 12, death occurs before repro age
385
Q

How are x-chromosomes inactivated?

A
  • XIC (x-chromosome inactivating center) on x-chromosomes has a gene known as XIST (inactive x-specific transcript).- XIST is transcribed- XIST RNA associates closely with x-chromosome and mediate inactivation of most of the chromosome- DNA and histones on x-chromosome become methylated, transcription is inactivated and chromosome condenses
386
Q

Explain the parent-of-origin effect observed in Prader-Willi and Angelman syndromes

A
  • Prader-Willi: deletion exists from paternal chromosome 15, mothers is imprinted and therefore silenced. Incidence = 1/10-50K. Characterized by obesity, excessive food seeking behavior, hypogonadism, mental retardation, small hands and feet- Angelman: deletion exists on maternal chromosome 15, fathers is imprinted and therefore silenced. Incidence = 1/15K. Characterized by unusual facial features – large mandible and open mouth, seizures, movement and gait disorders, mental retardation, excessive laughter and absence of speech.
387
Q

What amino acid change occurs resulting in: (Explain each)

a. )HbS
b. )HbF
c. )Hbsaki/Hbgenova

A
  • a.) Glu to Val. Valine residue is sticky when in T-form, polymerizes with other sticky residues.
  • b.) His to Ser. Serine has decreased affinity for 2,3-BPG.
  • c.) Val to Pro. Proline breaks alpha-helix resulting in unstable Hb molecule, unable to bind o2 (or decreased affinity?)
388
Q

What are suicide substrates?

A
  • Suicide substrates, aka Trojan Horse substrates, are special classes of irreversible inhibitors that only become inhibitors through the catalytic action of the target enzyme. They bind covalently at active site residues.
389
Q

Explain why balanced chromosomal alteration often cause infertility of otherwise health carriers. Explain the inheritance of balanced alterations.

A
  • Carriers of balanced alterations ~ 0.2% of live births are not aware of their condition as there is typically no impact on the carrier - Their genetic abnormality does; however, shows at reproduction when they lead to the production of aberrant gametes. See image below
391
Q

How to decipher folate or vitamin B12 or combined deficiency in clinical setting?

A
  • Megaloblastic anemia can occur when deficiency in both and can be treated with folate in either cases. Folate given to B12 pt with allow sufficient oxidized THF to be available for DNA synthesis and resolving of megaloblastic anemia - Folate alone will not resolve the neurologic symptoms that go along with vit B12 deficient patients – demyelination leading to brain and nerve damage. Methylmalonyl acidemia seen in vitamin B12 deficient pt, not in folate deficient pt - Combined deficiency seen in chronic malnourished (eg. Alcoholic pts)
392
Q

What is VACTERL association? What is an association?

A
  • vertebral defects, anal atresia, cardiac abnormalities, tracheo-esophageal fistula, renal and limb abnormalities- defects are through to occur in blastogenesis phase of development- risk factor: maternal diabetes- association = group of birth defects that for unknown reasons often occur together
393
Q

Which AAs can be used to synthesize alpha-ketoglutarate?

A
  • Mnemonic: Greg’s Hot Girlfriends Are Pregnant Ovals - Glutamine, Histidine, Glutamate, Arginine and Proline (and ornithine)
395
Q

Describe role of HDL. Include where synthesized, association with apoproteins, source of apoproteins, destination of contents and fate of this lipoprotein.

A
396
Q

Describe how the determination of the 3-D structures of the isozymes of prostaglandin synthase aided in the design of drugs that selectively inhibit COX-2.

A
  • NSAIDs act by inhibiting prostaglandin synthase, which are responsible for pain and inflammation. This enzyme has two enzymatic activities, one of which is cyclooxygenase activity and the other, hydroperoxidase. NSIADs inhibit the cyclooxygenase. Turns out there are two isozymes for this enzyme, COX-1 and 2. - COX-1 protects gastrointestinal mucosa, while COX-2 is associated with inflammation of arthritis. - As a result of this discovery, drugs have been designed specifically as inhibitors to COX-2. Eg. Celebrex, bextra and vioxx.
397
Q

Neurofibromatosis type I. Inheritance, defect, frequency

A
  • AD- Neurological defect in neurofibromin gene. Causes multiple benign tumors (neurofibromas) in skin, benign tumors on iris of eye called Lisch nodules, pigmented skin lesions (café-au-lait spots), tumors of CNS and mental retardation. New mutations, complete penetrance and variable expressivity- 1/3500
398
Q

Regulation of GDH

A
  • High energy (high GTP, high NADH) = inhibition of enzyme = decreased formation of alpha-KG - Low energy (high ADP) = activation of enzyme = increased formation of alpha-KG
399
Q

Function of vitamin B12 – what reactions is it involved in?

A
  • Only needed by two enzymes: 1.) methymalonyl-CoA mutase and 2.) methionine synthase - 1.) Converts L-methylmalony-CoA (intermediate in taking VOMIT AAs from propionyl-CoA) to succinyl-CoA - 2.) Converts N5 methyl THF to THF while converting homocysteine to methionine
400
Q

Outline synthesis of melanin

A
  • Tyrosinase converts tyrosine to DOPA and then to dopaquinone, which eventuall yields black/brown (eumelanin) or red/yellow (pheomelanin)
402
Q

What characteristics does an ideal population follow in terms of HW?

A
  • Large population size- Equal fitness among offspring- Random mating- No influx of new alleles by migration or mutation
403
Q

How are primers removed and Okazaki fragments joined together?

A
  • Specific RNase activity removes primer - DNA pol (possibly delta) fills in gap left by primer removal - DNA ligase uses ATP to seal Okazaki fragments
405
Q

Describe the synthesis of farnesyl PP.

A
406
Q

What do defects in Shh cause?

A
  • Left/right asymmetrical defects – situs inversus
407
Q

Describe synthesis of OAA from AAs

A
408
Q

Which amino acid residue is different in fetal Hb?

A

-Histidine residue in beta chain (adult) is replaced by serine in gamma chain (fetal). This is where 2,3-BPG usually binds in adults. Reduced affinity in HbF.

410
Q

What does a niacin deficiency lead to?

A
  • Pellagra, which has symptoms of 4 Ds (diarrhea, dementia, dermatitis, death)
411
Q

What are G-proteins? Distinguish between Gs, Gi and Gq. Explain how G-proteins function.

A
  • G-proteins are heterotrimeric proteins (with alpha, beta and gamma subunits) that are associated to the plasma membrane. When bound to GDP, they are in their inactive form. When bound to GTP, they are in their active form. They are GTPases and can self hydrolyze GTP to GDP and inactivate themselves. - There are many different types of G-proteins, including: a.) Gs, b.) Gi and c.) Gq. a.) Gs typically stimulate the activity of downstream enzymes b.) Gi typically inhibit the activity of downstream enzymes c.) Gq typically stimulate the activity of downstream enzymes - When receptors become activated, GDP-bound G-proteins bind them. This binding triggers and exchange of GDP for GTP, which activates the G-protein and causes dissociation of the alpha subunit from the beta-gamma dimer. Activated G-protein (alpha with GTP bound) associates with effector enzymes and alters their activity (inhibition or activation, depending on what type of G-protein it is). Intrinsic GTPase activity of the alpha subunit hydrolyses GTP to GDP and the G-protein is deactivated, re-associates with the beta-gamma dimer, able to participate in further signal transduction.
412
Q

Describe/draw the synthesis of glucose from non-CHO precursors (gluconeogenesis).

A
413
Q

Mechanism by which allopurinol reduces uric acid levels

A
  • Allopurinol and its metabolite oxypurinol (made by xanthine oxidase) are analogs of hypoxanthine - They inhibit xanthine oxidase and reduce uric acid formation - Intermediates such as hypoxanthine and xanthine are more soluble than uric acid and are eliminated in urine
414
Q

What is Rubinstein-Taybi syndrome?

A
  • Inherited condition characterized by hypoplastic maxillar (insufficient growth of midface), prominent nose, intellectual disability, polydactyly, growth retardation - Results from mutation in gene encoding CREB binding protein (CBP) - This gene is implicated in processes related to memory, cognition and developmental processes
415
Q

Explain the meiotic problems arising from an inversion

A
  • Inversion results when a c/s suffers two breaks and the broken off fragment is re-inserted in the wrong orientation.- As this is a type of balanced alteration, carriers are asymptomatic- Problems arise in offspring as chromosome with inverted region forms inversion loop during pairing with normal homolog. If crossing over occurs, material is translocated and some gametes are inviable.
417
Q

What are the regulatory points in glycolysis? What factors are stimulating (+) and inhibiting (-)? What is the rate-limiting step in glycolysis?

A
  • Regulatory points: a.) Hexokinase (+: insulin; -: G6P, acetyl-CoA, glucagon) b.) phosphofructokinase 1 (+: hormones via F26BP, ADP, AMP; -: ATP and citrate) c.) pyruvate kinase (+: F16BP and insulin; -: ATP) - Rate-limited step = phosphofructokinase 1
418
Q

What residues cover the o2 binding site in the beta subunits of Hb prior to existing in R-form?

A

-Valine. When rotated out, R-form is stabilized.

420
Q

The cell cycle is driven by the activity of cyclin-dependent kinases. Which cyclin-CDK complexes are most important at: a.) M-phase b.) Mid G1 phase c.) Late G1 phase d.) S phase

A

1.) M-phase: cyclin A/CDK1 and cyclin B/CDK1 2.) Mid G1 phase: cyclin D/CDK4 and cyclin D/CDK6 3.) Late G1 phase: cyclin E/CDK2 4.) S phase: cyclin A/CDK2

421
Q

Explain what reaction(s) the cell can perform when there is a need for NADPH and glucose. ATP is not required.

A
423
Q

What is uniparental disomy? What are the effects of this?

A
  • Zygote with only maternally or paternally imprinted homologues of a chromosome- This leads to problems with gene dosage (overabundance of one gene, lack of another for example).
424
Q

Which three AAs contribute to creatine synthesis? Where does the nitrogen in creatine come from? Where does the synthesis take place? Where does creatine come from in diet?

A
  • glycine, arginine and methionine (s-adenosylmethionine) - nitrogen comes from arginine - liver (some in kidney) - meat and fish
425
Q

Describe role of ATM in G2/M checkpoint

A

1.) ATM kinase senses replication forks, which indicates ongoing replication, and prevents checkpoint kinase from allowing activation of Cdc25C, which normally dephosphorylates CDK1. Therefore CDK1 remains inactive. ATM remains active as long as replication forks active

426
Q

What is an anaplerotic reaction? What is the importance of these reactions for the TCA cycle? What is an important anaplerotic reaction for the TCA cycle? Include enzyme and coenzymes required. Are there other reactions?

A
  • Anaplerotic reactions are those that replenish substrates. The TCA cycle is not an end-product producing reaction. It re-generates oxaloacetate in order for the cycle to occur again. However; TCA cycle intermediates, including oxaloacetate, are constantly being drawn away from the cycle as they serve as intermediates for biosynthetic pathways and therefore the oxaloacetate concentrations decrease preventing expanding rates of the TCA cycle should the need occur. - Pyruvate + ATP + HCO3 = oxaloacetate + ADP + Pi + H+ (ez: pyruvate carboxylase, coenzyme: biotin) - Yes, any reaction that generates an intermediate of the TCA cycle is a potential anaplerotic reaction. There is a reaction involving glutamate and glutamate dehydrogenase that generates more alpha-ketoglutarate, which adds to oxaloacetate in subsequent reactions.
427
Q

How does glutathione act to detoxify peroxides?

A
  • Forms GSSG with peroxide, then reconverted to glutathione via glutathione reductase, which uses NADPH
428
Q

Describe allosteric regulation of purine synthesis: what enzymes are regulated? Which allosteric modulators are responsible for this regulation?

A
  • PRPP synthetase (inhibited by ADP and GDP) - Amidophosphoribosyltransferase (inhibited by AMP, GMP; activated by PRPP) - Adenylosuccinate synthetase (inhibited by AMP) - IMP dehydrogenase (inhibited by GMP)
430
Q

Describe how the IP3-DAG signal transduction system works. Include all the necessary players from activation to deactivation.

A
  • Phospholipase C is activated by G-proteins (upon hormone/receptor binding) and hydrolyzes PIP2 (phosphatidylinositol 4,5 bisphosphate), a membrane lipid, generating two second messengers: IP3 (inositol 1,4,5 triphosphate) and DAG (diacylglycerol). - IP3 is water soluble and diffuses into cytosol where it binds to receptors on ER, triggering release of Ca2+ down its concentration gradient and into the cytosol. Ca2+ activates cytosolic protein kinase C (PKC) who have downstream effects. - DAG is liphophilic and remains membrane-associated, but easilty diffuses in the plane of the membrane. With Ca2+, it activates plasma membrane associated PKC. - Ca2+ released by the mechanism above can also bind calmodulin, causing it to interact with other protein kinases, phosphatases and ion channels. - Inactivation: 1.) receptor down-regulation/internalization/degration; 2.) protein phosphatases reverse action of PKC; 3.) Ca2+ pumped out of cytosol (into ER or to ECF); 4.) IP3 is dephosphorylated by phosphatase converting it into derivative that isn’t active; 5.) DAG is phosphorylated yielding phosphatidic acid or hydrolyzed to glycerol and FAs.
432
Q

Describe role of p53 in triggering cell cycle arrest or apoptosis in response to DNA damage. What gene encodes it?

A
  • p53 is normally an unstable protein, is ubiquinated and destroyed by proteasome when not stabilized - Phosphorylation by ATM/ATR (double-stranded breaks or UV damage respectively) increases stability of it, which leads to transcriptional control of many genes that regulate cell cycle arrest, repair of DNA and apoptosis - Encoded by gene TP53 - Active p53 is homotetramer. Missense mutations in one allele greatly reduce function. Any subunit mutation of the tetramer will cause aberrant function. Mutations act as dominant negative
433
Q

What are the consequences of PDH deficiency? Population affected? Symptoms?

A
  • Leads to chronic lactic acidosis, primary in kids. It leads to severe neurological problems and is frequenctly fatal.
434
Q

Draw function of glucagon presence (insulin absence) in regulation of FA synthesis.

A
436
Q

What are plasmalogens?

A
  • Plasmalogens are phosphoglycerides with an ether (as opposed to ester) linkage to glycerol. These are synthesized in peroxisomes are are components of the myelin sheath surrounding nerves.
437
Q

How is the urea cycle regulated? Explain

A
  • Carbomyl phosphate synthetase I = rate-limiting step - Carbomyl phosphate synthetase I is activated by N-acetylglutamate synthetase, which is activated by arginine - High levels of arginine are indicative of elevated peripheral blood ammonia levels
438
Q

How do FFAs travel in blood? Why is this transport method important?

A
  • They bind to hydrophobic pockets in albumin. - Don’t need to synthesize FFA transporters when we are in a starved state and need FFAs as albumin is always present.
439
Q

Name three common DNA lesions

A
  • Depurination: bond bw base and ribose hydrolyzed - Deamination: most often cytosine deamination generates uracil - Pyrimidine dimers: UV light dimerizes adjacent thymine bases on same DNA strand
441
Q

What is Beckwith-Wiedemann syndrome?

A
  • Example of uniparental disomy disorder- Incidence = 1/13000- Some types arise when child inherits homologues of a portion of chromosome 11 from father.- S&S: overabundance of IGF2, leads to kidney, adrenal and liver problems, resulting in severe hypoglycemia. Characterized by microcephaly, macroglossia (large tongue) and umbilical hernia. Susceptible to childhood cancer.
442
Q

Name 5 processes by which cells participate in development

A

1.) Gene regulation by TFs and chromatin modification2.) Cell-cell signaling (direct contact or morphogens)3.) Development of specific cell shape and polarity4.) Movement and migration of cells5.) Programmed cell death

444
Q

What enzyme is defective in alcaptonuria? Symptoms?

A
  • homogentisate oxidase - Symptoms: urine black upon standing, black pigmentation of cartilage and collage, joint destruction and arthritis
445
Q

Define dominant negative effect

A
  • Dominant negative effect: If the mutation produces an abnormal protein, the mutant protein may compete with the wildtype form. If the protein in question is part of a large complex, the presence of a few deformed mutant proteins may destabilize the structure. A dominant negative effect affects mostly structural proteins
446
Q

Precursor to histamine, how is it derived? What are cofactor requirements? Use?

A
  • Histidine - Decarboxylation of histidine - Pyridoxal phosphate - Produced by mast cells, involved in allergic reactions and in control of acid secretion by stomach
447
Q

Explain how 2,3-BPG; CO2 and H+ influence Hb’s affinity for oxygen. Provide physiological relevance.

A
  • Increase in 2,3-BPG decreases Hb’s affinity for o2 (binds in central cavity, stabilizing T-form through strong ionic interactions) promoting release - Increase in H+ conc (low pH) decreases Hb’s affinity as they bind His, forming positive residue which binds aspartate, stabilizing T-form and promoting o2 release - CO2 binds to N-terminal of Hb subunits forming carbamates. Carbamates carry a negative charge and participate in salt bridges that stabilize T-form, promoting o2 release Physiological relevance: In rapidly metabolizing tissue, CO2 and proton concentration is high, o2 requirement is high. Protonated His and carbamate formation stabilize the T-form of Hb resulting in offloading of o2 here. Conversely, in lungs, removal of CO2 (requiring proton with bicarb ion), causes deprotonation of Hb and conversion to R-form where conveniently o2 tension is also high. Amount of o2 carried to peripheral tissues increases as a result.
449
Q

Action of ricin

A
  • potent ribosome inactivating protein (RIP) found in castor beans - it removes adenine bases from rRNA
450
Q

Explain genome-wide association studies

A
  • GWAS refers to analysis of millions of SNPs to determine their connection with disease. Genetic polymorphisms underlie why certain individuals have a genetic susceptibility to certain disease or have different responsiveness to drugs.- From these studies, you get odds ratio that SNP is associated with disease state.- Note, SNPs are not necessarily disease-causing. SNPs often lie outside of coding regions, but show strong linkage with the mutation that causes the disease.
452
Q

Explain why a defect in cholesterol biosynthesis affects development

A
  • For correct functioning of Shh, the protein must interact with cholesterol. - Disturbances in cholesterol biosynthesis will therefore also have broad impact on development.- Defect = Smith-Lemli-Opitz syndrome, IEM in cholesterol synthesis
453
Q

What type of protein is YopH? What is it made by? What does it do?

A
  • YopH is a protein tyrosine phosphatase. - It is encoded by a plasmid in Yersinia pestis, the bacterium responsible for the bubonic plague. - It dephosphorylates certain key proteins and prevents host immune system from mounting an effective defense against the bacterium.
455
Q

How can prostaglandin synthesis be inhibited? Explain

A
  • NSAIDs and glucocorticosteroids(aka glucocorticoids) inhibit COX1/2 and prostaglandin synthase (PGH synthase)
456
Q

Name the ketone bodies. How are they used?

A
  • Ketone bodies: acetoacetate, acetone (result of spontaneous decarboxylation of acetoacetate) and beta-hydroxybutyrate. - Ketone bodies have the ability to cross membranes and can leave liver and travel to peripheral tissue. - Beta-hydroxybutyrate gets oxidized to acetoacetate via beta-hydroxybutyrate dehydrogenase (yields NADH) - CoA from succinyl-CoA is transferred to acetoacetate yielding acetoacetyl-CoA via enzyme acetoacetate:succinyl-CoA transferase - Thiolase produces 2 x acetyl-CoA from acetoacetyl-CoA, which function in the TCA cycle to produce energy. - Acetone cannot be converted to acetyl-CoA
457
Q

Describe synthesis of ketone bodies

A
459
Q

Define loss-of-function.

A
  • Loss-of-function: mutation reduces the protein’s activity
460
Q

Acetyl-CoA. a.) What is its structure? b.) What is its metabolic importance? c.) What is its source? Where does it come from?

A
  • a.) see picture - b.) It is an important intermediate and precursor molecule for TCA, FA biosynthesis, ketone body formation and cholesterol biosynthesis (leading to steroid hormones, bile acids and vit D production) - c.) It is produced as a result of glycolysis, beta-oxidation and AA deamination/oxidation. It is requires presence of vitamin B5.
461
Q

Through what two cell-signaling mechanisms does epinephrine function? What are the receptor types it binds to? Where are these receptors found? What G proteins are implicated in each?

A
  • Epinephrine can signal through cAMP and DAG-IP3 pathways. - It binds beta-adrenergic receptors in skeletal muscle through the cAMP (and adenylate cyclase via Gs proteins) signaling pathway to trigger glycogenolysis. - It binds alpha-1-adrenergic receptors in the liver through the DAG-IP3 (and PLC via Gq proteins) signaling pathway to trigger glycogenolysis.
462
Q

How is cholesterol synthesis regulated? Explain.

A
  • It is regulated at the HMG-CoA reductase reaction. 1.) AMP high (starved state): phosphorylation by AMP-dependent kinase, which inactivates enzyme and cholesterol synthesis is low during times of starvation 2.) Insulin high (well-fed state): desphosphorylation by protein phosphorylase activates enzyme, which means that cholesterol synthesis is high during times of well-fed 3.) Cholesterol inhibits the enzyme by feedback inhibition, which means that when cholesterol levels are high, cholesterol synthesis is low
463
Q

Describe/Draw how fructose is degraded.

A
464
Q

Which intermediate during ketone body synthesis can function in another pathway (besides acetyl Co-A)? What pathway? During what environmental conditions will this occur?

A
  • HMG-CoA = beta-hydroxy-beta-methylglutaryl-CoA - This molecule is used in the synthesis of cholesterol. - Will occur in well-fed times
465
Q

What is cholera? What is it caused by? How does cholera toxin work? What are the important symptoms of cholera?

A
  • Cholera is bacterial disease that causes massive diarrhea, can lead to death as the result of dehydration. - It is caused by Vibrio cholerae that produces the cholera toxin. - Cholera toxin catalyzes ADP-ribosylation of alpha-subunit Gs proteins at an arginine residue that causes the subunit to be in the GTP-bound active state. As a result, signaling by this molecule cannot be turned off, cAMP levels remain high, PKA remains active. CFTR is phosphorylated. Secretion of Cl, Na and water into the intestinal lumen, ie. massive diarrhea.
466
Q

From what compound is PRPP synthesized? What enzyme catalyzes this reaction

A
  • Ribose 5 phosphate - Ez: PRPP synthetase
467
Q

Contrast malformation, deformation and disruption. Examples

A
  • Malformation: intrinsic abnormality in developmental process. Eg. Polydactyly- Deformation: extrinsic influence on development of the affected tissue. Eg. Oligohydramnios- Disruption: destruction of developing tissue. Eg. Amniotic bands
468
Q

In a recessive disease with an incidence of 1/10000, how high is the allele frequency in the gene pool? How high is the carrier frequency?

A
  • f(a)^2 = 1/10000, then f(a) = 1/100 = 0.01- f(A) = 1-f(a) = 99/100 = 0.99- Since the disease is so rare, assume f(A) = 1, then frequency of heterozygote = 2f(a)f(A) = 0.02
469
Q

Outline synthesis of T3/4. What precursor molecule is used?

A
  • Synthesized from tyrosine residue on thyroglobulin, after synthesis of the molecules, proteolysis releases them as free T3/4
470
Q

What are/is disorder(s) that prevent proper use of galactose? Explain.

A
  • Galactosemia - Three types: a.) Classic galactosemia: most common / severe form where there is a galactose-1 phosphate uridyl transferase deficiency b.) Deficiency of galactokinase c.) Deficiency of UDP-galactose epimerase - Results in accumulation of galactose1-phosphate in liver and other tissues (CNS, kidney). Newborns present with milk intolerance and signs of liver failure, cataracts and intellectual disability. Also present with jaundice, lethargy and hepatomegaly. Diagnosis is by detection of galactose or galactose-phosphate in urine. Pts must receive a galactose-free diet (no lactose either, which is a glucose, galactose disaccharide).
471
Q

In general terms, describe how activation of p53 leads to efflux of cytochrome c from the mitochondria, ultimately resulting in caspase activation and cell death.

A
  • ATM/ATR activity stabilizes p53 (remember, ATM and ATR are activated by double-stranded DNA breaks and UV light/damaging drugs respectively) - Targets of p53 include proapoptotic proteins belonging to Bcl-2 family - Bcl-2 family proteins (not Bcl-2, which is antiapoptotic) stimulates BAX activity, which promotes release of cytochrome c from mitochondria - Cytochrome c binds to Apaf-1 in cytoplasm - Oligomerization of cyt c with apaf-1 occur - This recruits caspase 9, initiator caspase - Caspase 9 activates executioner caspases, such as caspase 3 - Cell is destined to death
472
Q

Outline synthesis of catecholamines. Where does each reaction occur?

A
474
Q

What is biotin? Which vitamin is it?

A
  • Biotin is a B vitamin, also known as vitamine B7. It is required by pyruvate carboxylase.
475
Q

What is the action of digitalis?

A
  • Digitalis is an inhibitor of the Na/K ATPase. Causes increase in FOC, decrease in HR.
476
Q

Explain transportation from nucleus to cytoplasm

A
  • Ran-GDP binds GTP and dissociates from GDP forming Ran-GTP - Ran-GTP binds exportin, binds cargo and moves through nuclear pore complex into cytoplasm - Hydrolysis of complex occurs, Cargo, exportin and Ran-GDP dissociate, Ran-GDP returns into nucleus
477
Q

How does NADH:NAD+ ratio determine predominant forms of ketone bodies in liver?

A
  • If NAD+ concentration is low in liver/kidney, acetoacetate will be converted to beta-hydroxybutyrate, causing an increased concentration of NAD+ and decrease in NADH.
478
Q

Glucogenic AAs?

A
  • All AAs except for TIPhe and L AAs
479
Q

Explain enzymatic defect leading to the development of acute fatty liver of pregnancy.

A
  • Fetus releases hydroxyacyl metabolites into the mother’s circulation as a result of long chain hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency in the fetus. FAs accumulate in mother’s liver and they present with acute liver failure in pregnancy (jaundiced).
480
Q

What apoproteins do chylomicrons associate with? Where?

A
  • Chylomicrons in enterocytes associate with ApoB-48 - After entering into serum, CM now associates with ApoC and ApoE, which is receives from HDL
481
Q

Name two functions of the Shh morphogen

A
  • Shh gradient secreted from notochord and floorplate of neural tube helps organizing the different cells in brain and spinal chord. Defects leads to midline defects- Shh also secreted from zone of developing limb to induce development of posterior limb elements.
483
Q

What is the role of glycogenin vs glycogen synthase?

A
  • Glycogenin: acts as a primer protein and generates short glucose chains via autoglycosylation of glycogenin - Glycogen synthase: takes over from glycogenin and transfers glucose residues from UDP-glucose - Both form alpha-1,4-bonds
484
Q

Define proto-oncogene. How does mutation in these genes contribute to development of cancer?

A
  • Proto-oncogene: promote cell growth - Loss of function causes excessively active growth promotion = stuck accelerator = oncogene = typically dominant mutations
485
Q

Describe how heme is synthesized

A
  • In mito: Glycine and succinyl-CoA acted on by ALA synthase = aminolevulenic acid (ALA) - 2 x ALA molecules acted on by PBG synthase (aka ALA dehydratase) and produce porphobilinogen (PBG) out in cytoplasm - 4 x PBG acted on by PBG deaminase with product fed into UPG III synthase to produce uroporphyrinogen III (UPG III) - UPG III acted on by UPG III decarboxylase to produce coporphyrinogen III (CPG III) - CPG III enters mito again and becomes protoporphyrinogen IX, which is acted on by ferrochelatase which adds Fe2+ to structure causing formation of heme B
486
Q

Explain what reaction(s) the cell can perform when there is a need for both NADPH and ribose. ATP is not required.

A
487
Q

Explain how bilirubin is excreted. What occurs when there is a defect in this?

A
  • Glucuronic acid is a prerequisite for bilirubin excretion. - UGT, which is an enzyme that catalyzes the conjugation of bilirubin with glucuronic acid leads to buildup of bilirubin and syndromes known as Crigler-Najjar/Gilbert.
488
Q

Describe synthesis of alpha-ketoglutarate from AAs

A
489
Q

Deficiency from niacin causes what diseases?

A
  • Pellagra - Hartnup disease
490
Q

How does the steroid hormone cortisol regulate transcription?

A
  • Cortisol is secreted by adrenal cortex - Binds to and activates receptors in cytoplasm, causing conformational change to that receptor - Accessory proteins dissociate, activated receptors dimerize - Dimerized receptors are imported into nucleus - Bind GRE elements on genome, regulate transcription positively or negatively - Typical result of cortisol is to stimulate gluconeogenesis, glycogen deposition, fat and protein degradation, inhibit inflammatory response
491
Q

What is Lesch-Nyan syndrome?

A
  • Hereditary syndrome with severe or complete deficiency of HGPRtase activity - Hyperuricemia, uric acid stones, intellectual disability and self-injurious behavior
492
Q

Describe creatine synthesis.

A
493
Q

What apoproteins do VLDL associate with? IDL? LDL? Where?

A
  • VLDL associates with ApoB100 in hepatocytes - VLDL in serum associates with apoE and apoC obtained from HDL - IDL remains associated with all 3 per above - LDL remains associated with all 3 per above
494
Q

What is meant by glucogenic AAs? Which are these? What is meant by ketogenic AAs? Which are these? Which are both?

A
  • Glucogenic = These AAs can be converted into glucose via TCA intermediates (alpha-kg, succ coa, fumarate, OAA) and pyruvate – anything that generates OAA, an intermediate in gluconeogenesis - Ketogenic = These AAs can be converted into acetyl-CoA and acetoacetyl-CoA, can never become glucose - Gluco/ketogenic AAs = TIPhe mnemomic = all T AAs (thr, tyr, trp), iso, phe - Ketogenic AAs = all L AAs - Glucogenic AAs = all others
495
Q

Which urea cycle product is a TCA intermediate? How is the TCA cycle connected to the urea cycle? Why is this important?

A
  • Fumarate production in urea cycle links urea cycle to TCA cycle in what is known as urea-TCA bicycle - Urea cycle requires ATP energy. Fumarate generates energy in TCA cycle to offset ATP requirements by TCA cycle - OAA can be converted into aspartate and feed into urea cycle
496
Q

How do growth factors stimulate cell division? Discuss growth factor binding and all intracellular pathway

A

1.) Binding growth factor stimulates dimerization and autophosphorylation on the tyrosine residues of the receptors (growth factor receptors contain intrinsic protein tyrosine kinase activity). 2.) Phosphorylation residues form docking sites*. 3.) Adaptor protein GRB2 binds phosphotyrosine, which recruits guanine nucleotide exchange factor Sos, which activate 4.) Sos activates g-protein Ras by promoting exchange of GDP (Ras + GDP = inactive) for GTP (Ras + GTP = active) 5.) Ras then activates Raf (aka MAP kinase kinase kinase) 6.) Raf activates MAP kinase kinase (aka MEK) by phosphorylating it 7.) MAP kinase kinase activates MAP kinase through phosphosphorylations, which translocates into the nucleus to regulate transcription of many genes, including stimulation transcription of gene encoding c-Fos 8.) c-Fos stimulates transcription of cyclin D, CDK4/6 9.) Cyclin D, CDK4/6 phosphorylates Rb (retinoblastoma) in Rb:E2F complex, causing dissociation of Rb from E2F. 10.) E2F is a transcriptional activator of cyclin E/CDK2 (late G1 cyclin), which also acts to phosphorylate Rb:E2F as in 9. E2F also is a transcriptional activator of cyclin A (s-phase cyclin) * Most signal Ras/MAP kinase pathway

497
Q

What is the clinical correlation of deficiency in uridine nucleotide synthesis?

A
  • UMP synthase mutation = orotic aciduria - Crystalluria, hypochromic megaloblastic anemia, growth retardation, neurologic abnormalities - Anemia is unresponsive to B12 and folic acid - Treatment with uridine
498
Q

What are some common causes of respiratory alkalosis?

A

Anxiety, pain, high altitude, mechanical hyperventilation

499
Q

Describe reactions of urea cycle. Where is each reaction taking place?

A
501
Q

Describe/Draw the oxidative/non-oxidative phase reactions of the pentose phosphate pathway.

A
502
Q

Regulation of pyrimidine synthesis

A

1.) UTP and CTP - Carbomyl phosphate synthetase II (inhibited by UTP, activated by PRPP) - CTP synthase (inhibited by CTP, activated by UTP) 2.) dTTP - thymidylate synthase (unknown)

503
Q

What is the name of the molecule that synthesizes mRNA?

A
  • RNA pol II
505
Q

What AAs are made from: alpha-ketoglutarate and oxaloacetate?

A
  • Asp, Asn, Arg, Glu, Gln, Pro
506
Q

Incontinentia pigmenti.

A
  • X-linked dominant. Result from a defect in NEMO (NFkappaB essential modulator) that results in rash in early infancy, mental retardation, microcephaly and defects in tooth development. - It is lethal in male embryos
507
Q

List three axes and discuss the function of each, what gene/factor is responsible for formation of this axis?

A

1.) Anterior-posterior axis- First visible structure defined by primitive streak with groove. Node is at the anterior end. Nodal gene is responsible for formation of this axis2.) Dorsal-ventral axis- Secreted proteins noggin and chordin are secreted from cells in the node and induce dorsal development in a concentration-dependent manner3.) Left-right axis- Asymmetric expression of the gene for the signaling protein Shh: sonic hedgehog from the notochord causes left/right asymmetry. This leads to asymmetric expression of nodal on the left-side that initiates development of the heart tube.

508
Q

Huntingtin Disease. Inheritance, defect, frequency

A
  • AD- Neurodegerative disorder causes by gain of function mutation. Triplet expansion in gene causes protein instability. Neurodegenerative disorder with late age onset, age dependent on number of CAG repeats. Premutation = reduced penetrance = 35-40 repeats. 40+ = fully penetrant. New mutations occur frequently in this disease.- 5/100000
509
Q

Describe how the family history or the pts genotype influence the prediction of risk (relative risk ratio and relative risk)

A
  • Incidence and prevalence data are different for ppl with different numbers of affected relatives.- In diseases with strong genetic component, prevalence for ppl with affected relatives is much higher than for general population- Therefore a measure known as relative risk ratio (lamda r where r indicates relationship) compares frequency of a dz in relatives of affected and unaffected probands.- Lambda r = prevalence of dz in relative r of affected person divided by prevalence in general population- Relative risk is a way to describe disease association of an allele: how much more likely a carrier of an allele is to develop the dz than a non-carrier. If RR for carrying allele Y is 3.5 then we tell pts: your risk is 3.5 times the risk of a non-carrier of allele Y
510
Q

The Km of an enzyme is independent of enzyme concentration while the velocity of the reaction (including Vmax) is directly proportional to enzyme concentration. Why?

A
  • Km reflects the substrate concentration at which v = Vmax / 2. Therefore, when changing the enzyme concentration, the rate will change in proportion to that. What is also true, is that increase substrate concentration will increase reaction rate to Vmax. Km is calculated by looking at rate constants, which are independent on enzyme concentration and mere just a reflection of the properties of the enzyme. No matter how much enzyme you have, substrate interaction with the enzyme are limited by its intrinsic properties.
511
Q

What is pertussis? What is it caused by? How does pertussis toxin work?

A
  • Pertussis is a respiratory bacterial infection. - It is caused by Bordetella pertussis, which produces the pertussis toxin. - Pertussis toxin catalyzes ADP-ribosylation of alpha-subunit Gi protein as a specific cysteine residue, which lowers the affinity of the G-protein for GTP. As a result, the G-protein is trapped in the inactive conformation and cannot inhibit adenylate cyclase resulting in increased cAMP levels. Unclear the exact mechanism that leads to respiratory issues; however.
512
Q

Synthesis of melatonin

A
513
Q

Describe how the cAMP signal transduction system works. Include all the necessary players from activation to deactivation.

A
  • cAMP, or cyclic AMP, is a secondary messenger. It is generated through the enzyme adenylate cyclase by converting ATP. - Gs proteins activate adenylate cyclase and cause rapid rises in cAMP levels. - cAMP acts as a secondary messenger by targeting protein kinase A (PKA, also known as cAMP-dependent protein kinase). It transfers gamma-phosphate groups from ATP to the hydroxyl groups of serine / threonine residues on the regulatory subunits, which cause conformational change and dissociation from the catalytic subunits, which then perform their specific downstream functions. - Deactivation of cAMP signaling: 1.) cAMP is hydrolyzed by cAMP phosphodiesterase to AMP and is no longer capable of activating PKA. 2.) intrinsic GTPase activity of G-protein returns protein to inactive states and adenylate cyclase is no longer activated. 3.) Hormone-receptor are no longer in active state (either internalized/degraded or inactivated by phosphorylation. 4.) Downstream enzymes phosphorylated by PKA are dephosphorylated by protein phosphatases and therefore are deactivated. - Gi proteins typically inhibit adenylate cyclase and cause rapid decreases in cAMP levels.
514
Q

Why does accumulation of uric acid lead to development of gout and kidney stones?

A
  • Uric acid is normally excreted in urine - It and salts have limited solubility, they are typically close to saturation in normal healthy individuals. Common salt = sodium urate - Deposition of sodium urate in kidney = kidney stone - Deposition of sodium urate in joints = gout (triggers inflammatory response)
515
Q

Why does an inability to generate THBtn decrease production of tyrosine, catecholamines and serotonin?

A
  • Tyrosine: synthesized by phenylalanine hydroxylase - Catecholamines: synthesized in part by tyrosine hydroxylase - Serotonin: synthesized by tryptophan hydroxylase - These hydroxylase enzymes require o2, NADH and THBtn
516
Q

By what mechanism does the toxin alpha-amantin function? How are ppl exposed to this? Symptoms? Treatment?

A
  • Death cap mushroom common in cool, damp regions of pacific coast - Genus/species: amanita phalloides - Contains alpha-amantin, which is a Pol II inhibitor - Nausea, vomiting, coma, hepatic encephalopathy from liver failure - Liver failure ~ 48 hour post-ingestion - Tx: gastric lavage, activated charcoal and liver transplant
517
Q

PDH complex. a.) Describe its structure? b.) Where is it located? What is its function? c.) What are its coenzymes, vitamins? d.) What are its substrates and products? Show the balanced reaction it catalyzes. e.) Explain mechanisms for regulation of PDH complex. Explain when it is active and inactive.

A
  • a.) PDH is a 3 enzyme complex. It contains a pyruvate decarboxylase (aka pyruvate dehydrogenase), a dihydrolipoyl transacetylase and a dihydrolipoyl dehydrogenase - b.) It is located in the mitochondrial matrix and serves to link glycolysis to the TCA cycle - c.) It requires 5 coenzymes: TPP, FAD/FADH2, NAD/NADH2, CoASH and lipoamide and 4 B-vitamins: thiamin (e) – B1, riboflavin – B2, niacin – B3 and pantothenic acid – B5. - d.) CH3COCOO- + NAD+ + CoASH = CH3COSCoA + CO2 + NADH + H+. The reaction is not reversible as the delta Go’ is too high. Too much free energy is lost. - e.) PDH is inactive when phosphorylated and is therefore activated by PDH phosphatase. PDH is deactivated when phosphorylated by PDH kinase. It is activated during times when ATP energy is needed: low concentrations of CoASH, pyruvate, ADP and NAD+ (substrates of reaction). These states deactivate the kinase, leading to active form of PDH. It is deactivated during times when ATP energy is not required: high concentrations of acetyl-CoA and NADH (products of reaction). These states activate the kinase, leading to inactive form of PDH.
518
Q

Describe changes in mRNA levels that occur as a result of insulin binding

A
  • Increase in acetyl-CoA carboxylase and FA synthase - Decrease in PEP carboxykinase
519
Q

Explain the process of FA elongation from a 16 carbon molecule. Why is this necessary? Where does it occur?

A
  • Cytoplasm has FA synthase that synthesizes FAs to 16 carbons = palmitate (16:0), which is a saturated molecule. - Many lipid structures in the body are longer than 16 carbons - Elongation occurs in mitochondrial as shown here:
520
Q

Which polymerase has proofreading activity?

A
  • DNA Pol delta
521
Q

How to distinguish between hepatic and post-hepatic jaundices?

A
  • Hepatic: watch for other marker of liver disease, such as ALT and AST - Post-Hepatic: watch for other markets of blocked bile ducts such as presence of alk phos in serum
522
Q

Explain the effect of glucagon-induced phosphorylation on glycogen phosphorylase and glycogen synthase.

A
  • Glycogen phosphorylase is activated - Glycogen synthase is inactivated.
523
Q

How do newborns become riboflavin deficient?

A
  • If a newborn is jaundiced, treatment requires use of “bili-lights”. As riboflavin is light-sensitive, newborns become riboflavin deficient.
524
Q

How does 5-fluorouracil function?

A
  • Inhibits thymidylate synthase, which blocks de novo dTMP synthesis, ultimately blocking cell proliferation - It is converted into FUMP by phosphoribosyltransferase in the cell - FUMP is phosphorylated by a kinase and converted into FUDP/FUTP - Ribonucleotide reductase converts it into FdUDP, with kinase converting it into FdUTP - FdUTP can be incorporated into DNA - FUTP can be incorporated into RNA - Biggest effect is FdUDP is acted on by phosphatase and product, FdUMP, inhibits thymidylate synthase
525
Q

Synthesis of glycine

A
  • PLP = active form of B6
526
Q

What enzyme inserts iron into heme molecule? Where in cell?

A
  • Ferrochelatase inserts Fe2+ into protoporphyrinogen IX molecule causing formation of heme B
527
Q

Describe the gene therapy trial for ADA-SCID

A
  • This was the first successful gene therapy trial in 1990. Cured SCID pt caused by absence of ADA: adenosine deaminase.- ADA gene was inserted into modified retro-virus- T-cells were isolated from pt. - Retrovirus was cultured with T-cells and inserted ADA into cells.- Cells that incorporated ADA gene into their genome were selected and grown in culture. - Cells were re-implanted into pt.
528
Q

Describe/draw the reactions of glycogen degradation (glycogenolysis).

A
529
Q

Describe beta-thalassemia

A
  • Reduced beta-chain of hemoglobin due to mutation in beta-globin gene at first exon/intron boundary. Mutation at 5’ splice site causes two aberrant mRNA products to be formed
530
Q

TCA cycle. a.) What are the 2 major functions of the TCA cycle? b.) What is the overall balanced reaction of the TCA cycle? c.) Draw the reactions of the TCA cycle. Include enzymes, cofactors. d.) What is the fuel for the TCA cycle? e.) Which reaction(s) liberate CO2? f.) Which reaction(s) liberate NADH? g.) Which reaction(s) liberate FADH2? h.) Which reaction(s) involve substrate-level phosphorylation? i.) How many ATPs/ATP-equivalents are produced from a molecule of acetyl Co-A? From pyruvate? From glucose? j.) Explain mechanisms for TCA cycle regulation. What enzymes are regulated?

A
  • a.) Serves to generate ATP and to generate biosynthetic precursors. - b.) CH3COSCoA + 2H2O + 3NAD+ + FAD + GDP + Pi = 2CO2 + 3NADH + 3H+ + FADH2 + CoASH + GTP - c.) see picture - d.) Acetyl-CoA - e.) isocitrate to alpha-ketoglutarate; alpha-ketoglutarate to succinyl-CoA - f.) isocitrate to alpha-ketoglutarate; alpha-ketoglutarate to succinyl-CoA; malate to oxaloacetate - g.) succinate to fumarate - h.) succinyl-CoA to succinate - i.) since 3 ATPS per NADH and 2 ATPS per FADH - 12 per acetyl CoA, 15 from pyruvate (considering PDH reaction) and 30 from glucose - j.) Regulation of the TCA cycle occurs through regulation of isocitrate dehydrogenase and alpha-ketoglutarase dehydrogenase complex. Isocitrate dehydrogenase activity increases when conc of ADP are high and decreases when NADH conc are high. Alpha-ketoglutarate dehydrogenase complex activity decreases when concentrations of succinyl-CoA and NADH are high.
531
Q

What are the three factors that lead to development of fatty liver in chronic alcoholism?

A

1.) alcohol metabolism in liver generates NADH and acetyl-CoA. High concentration of NADH blocks TCA cycle enzymes and forces acetyl-CoA into FA synthesis. 2.) Beta-hydroxyacyl dehydrogenase (beta-oxidation enzyme) requires NAD, which is at low concentration during alcohol metabolism. This means that FA breakdown by liver is slowed down and leads to accumulation of fat. 3.) Damaged liver tissue has reduced capacity to synthesize VLDLs to export FAs to adipose tissue

532
Q

Draw the Lineweaver-Burk plot and indicate what each intercept, slope and axes mean. Indicate what navigating around the graph means.

A
533
Q

Name the sphingolipidoses. Name the enzyme defect, accumulated lipid and presentation.

A

1.) Tay-Sachs Disease Enzyme defect: beta-hexosaminidase A Accumulated lipid: ganglioside GM2 Presentation: mental retardation, blindness, cherry red spot on macula, death before age 3 2.) Gaucher Disease Enzyme defect: beta-glucosidase (beta-cerebrosidase) Accumulated lipid: glucocerebroside Presentation: liver and spleen enlargement, erosion of long bones 3.) Fabry Disease Enzyme defect: alpha-galactosidase Accumulated lipid: ceramide trihexoside Presentation: skin rash, kidney failure 4.) Niemann-Pick Disease Enzyme defect: sphingomyelinase Accumulated lipid: sphingomyelin Presentation: liver and spleen enlargement, mental retardation 5.) Sandhoff Disease Enzyme defect: beta-hexosaminodase A and B Accumulated lipid: GM2 ganglioside and globosides Presentation: similar to Tay-Sachs, progresses more rapidly 6.) Metachromatic Leukodystrophy Enzyme defect: arylsulfatase Accumulated lipid: sulfatide Presentation: mental retardation

534
Q

Provide examples of defects in oxidative phosphorylation.

A
  • LHON: Leber’s Hereditary Optic Neuropathy is due to mutations in subunits of complex I and is characterized by sudden-onset blindness in young adults. - MERRF: Myoclonic Epilepsy and Ragged Red Fibers is due to abnormally shaped mitochondria with dimished cytochrome oxidase (complex IV) activity as the result of a point mutation in the mitochondrial gene for lysine tRNA. It is characterized by myoclonus and ataxia with generalized seizures. - MELAS: Mitochondrial Encephalopathy, Lactic acidosis and Stroke-like activity is due to abnormally shaped mitochondria with normal cytochrome oxidase activity that is caused by a point mutation in the gene for leucine tRNA. Early symptoms include muscle weakness, pain, recurrent headaches, loss of appetite, vomiting and seizures. Stroke-like episodes begin before age 40. Episodes involve transient hemiparesis (temporary muscle weakness on one side of body), altered consciousness, vision abnormalities, seizures and severe headaches resembling migraines. Episodes lead to brain damage and in some cases dementia.
535
Q

Types of apoproteins and functions?

A
  • Apoprotein A: activates LCAT (lecithin:cholesterol acyltransferase), which generates cholesterol esters from cholesterols. Extracts lipids from membranes for reverse transport. - Apoprotein B: structural protein, interacts with lipoprotein receptors and mediates uptake of particle into target cells - Apoprotein C: modulate lipoprotein lipase (LPL) activity, which liberates FFAs and glycerol from lipoproteins - Apoprotein E: bind to receptors to allow removal of remnant particles from circulation
536
Q

Synthesis of methionine

A
537
Q

What are inborn errors of metabolism relating to synthesis of succinyl-CoA from AAs?

A
  • AAs = VOMIT (valine, odd chain FAs, methionine, isoleucine, threonine) - 1.) deficiency in propionyl-CoA carboxylase resulting in propionic / propionyl acidemia - 2.) deficiency in racemase resulting in D-methylmalonyl aciduria - 3.) deficiency in mutase (or Vit B12, required as cofactor) resulting in methylmalonyl aciduria
538
Q

What is the clinical significance of ALT and AST?

A
  • AST found at highest levels in liver, but also in other tissues including cardiac muscle - ALT abundant in liver - Both are normally low in serum - When tissues are damaged, these enzymes are released into the serum - Elevation of AST indicates liver disease, but also acute MI, kidney damage, pancreatitis etc. - Elevation of ALT is more specific indicator of liver damage
539
Q

Explain the significance of CpG islands for gene expression

A
  • Methylation of DNA occurs on cytosine residues in areas rich in CG known as CpG islands- CpG islands are found upstream of genes close to the 5’ region- Methylation acts to silence part of the genome
540
Q

What two molecules are carriers of methyl groups?

A
  • SAM and THF
541
Q

What are the bonds/forces of each level of protein structure?

A
  • Primary structure: peptide bonds
  • Secondary structure: H-bonding
  • Tertiary structure: Cystine disulfide bridges (from two cysteines), hydrogen bonding, ionic interactions (salt bridges), hydrophobic forces
542
Q

What is MEOS? Explain.

A
  • Microsomal ethanol oxidizing system. This is a P450 cytochrome system that is induced in chronic alcohol abuse and has a higher capacity to process alcohol. Activation of this system causes oxidation of NADPH and weakens the cellular antioxidant defense mechanisms.
543
Q

How does carnitine deficiency present? What are the consequences?

A
  • Without carnitine, FAs cannot be imported into mitochondria to be used as energy source. - Pts present as fasting non-ketotic hypoglycemia. Ketones are unusually low in blood or / and urine.