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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/345

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

345 Cards in this Set

  • Front
  • Back

What is a nucelosome?

Histone core + linker DNA

Which bacterial DNA polymerase has 5´to 3´exonuclease activity? And why is it important?

Polymerase I


Primer removal and DNA repair

Which DNA polymerase has 3´to 5´exonuclease activity? And why is it important?

Polymerase I, II and 3


Proofreading of the newly synthesized DNA strand



What does it mean that replication is semiconservative?

Each DNA strand serve as a template for the synthesis of a new DNA strand producing 2 new DNA molecules, each with 1 old and 1 new DNA strand.

In which direction does DNA synthesis happen?

DNA synthesis is allways in a 5 ́to 3 ́direction.

What is the role of Dam methylase?

Methylates the newly synthesized DNA strand.

Methyl directed mismatch repair (the enzymes and their function)

MutS, MutL, MutH: winds up the DNA molecule




MutH: cleaves the unmethylated strand at methylated site




DNA helicase: unwinds the DNA strand




Exonuclease I: removes the newly synthesized fragment from the MutH cleavage site to just beyond the mismatch




DNA polymerase III makes the new strand

Base-excision repair pathway (the enzymes and their function)

DNAglycosylase: recognize common DNA lesions and remove the affected base (AP site is formed)




AP endonuclease: cut the DNA strand containing the AP site. A segment of DNA including the AP site is removed.




DNA polymerase I: replaces the removed DNA




DNA ligase: seals the remaining nick

Nucleotide excision repair (the enzymes and their function)

DNA lesion is detected.




Exinuclease: cleaves the DNA on each side of the damage.




DNA helicase: removes the DNA segment.




DNA polymerase (I in Ecoli, ε in human) : fills the gap




DNA ligase: seals the nick

What are the difference types of direct repair of DNA molecules?

- Repair of pyrimidinedimers with photolyase




- Direct repair of O^6-Methylguanine by O^6-Methylguanine-DNA methyltransferase




- Direct repair of alkylated bases by AlkB

What are the different types of short RNAs and their function?

miRNA (micro): Post-transcriptional regulators, silencing of many genes




siRNA (small interfering): Binds to the mRNA and silence it




saRNA (small activating): induce gene activation (long lasting)




snRNA (small nuclear): Processing of pre-mRNA in the nucleus. Part of the spliceosome.




snoRNA (small nucleolar): Guide chemical modifications of other RNAs (methylation or pseudouridylylation)




piRNA (piwi-interacting): transcriptional gene silencing of retrotransposons in germline cells.




rasiRNA (repeat associated small interfering RNA): Establishes and maintaining heterochromatin structure. Silencing transposons and retrotransposons.




qiRNA: inhibiting protein translation




tmRNA: rescues ribosomes (mRNA without stop codon). Facilitates degradation of non-normal mRNA.



What are the different types of RNA molecules and their function?

mRNA: Encode the amino acid sequence of one or more polypeptides specified by a gene or a set of genes.




tRNA: Read the information encoded in the mRNA and transfer an amino acid to the growing polypeptide chain in protein synthesis.




rRNA: Are constituents of ribosome.

Examples of ribozymes

Ribozymes = ribonucleic acid enzymes. They increase the speed of reaction.




Group I introns


Rnase P


rRNA


Spliceosomes


Hammerhead ribozym

What is the role of Sigma factor?

Sigma factor is a bacterial transcription initiation factor that enables specific binding of RNA polymerase to gene promoters. It controls promoter recognition and specificity.

What is the function of the Rho factor?

Prokaryotic protein involved in the termination of transcription.

What inhibits transcription in bacteria? (name and function)

Tetracycline: blocks binding of aminoacyl-tRNA to A-site of ribosome.




Streptomycin: prevents the transition from initiation complex to chain-elongating ribosome and also causes miscoding




Chloramphe-nicol: blocks the peptidyl transferase reaction on ribosomes




Erythromycin: blocks the translocation reaction on ribosomes




Rifampicin: blocks initiation of RNA chains by binding to RNA polymerase(prevents RNA synthesis)

What inhibits transcription in both bacteria and eucaryotes? (name and function)

Puromycin: causes the premature release of nascent polypeptide chains by its addition to growing chain end.




Actinomycin D: binds to DNA and blocks the movement ofRNA polymerase(prevents RNA synthesis)

What inhibits transcription only in eukaryotes? (name and function)

Cycloheximide: blocks the translocation reaction on ribosomes




Anisomycin: blocks the peptidyl transferase reaction on ribosomes




α-Amanitin: blocks mRNA synthesis by binding to RNA polymerase.

What is the function of 5´cap?

Protect mRNA and prevent degradation by endonucleases


Promotion of ribosome binding and translation


Regulates nuclear export

What is the function of poly (A)?

Protection


Transport


Translational enhancement

What is the difference between virus and retrovirus?

Virus contain both DNA and RNA, retrovirus contain only RNA.


Viruses have transcription process, whereas retroviruses have reverse transcription process. It needs to convert their RNA into DNA before it can insert it into the host genome.

What is transposon/retrotransposon?

DNA sequence that can change its position within the genome, sometimes causing mutations and altering the genome size.

What is the function of molecular chaperones?

Molecular chaperones: proteins assisting folding of nascent polypeptides, by preventing wrong folding.Molecular chaperones catalyze the formation of correctly folded, functionally active, native proteins, but they are not part of the product.

What is heat shock proteins?

Expression of many chaperones is induced by stress, such as by heat,because during heat-stress the probability of wrong folding is higher, therefore cells need more protection.These chaperons are called heat shock proteins (HSP’s).

Examples of diseases due to misfolding of proteins

Cystic fibrosis mutant


Parkinson’s disease


Alzheimer’s disease


Huntington’s disease

Hoe does protein degradation happens in eukaryotes and bacteria respectively?

Eukaryotes: Ubiquitination (signal of death for proteins) and proteasome (enzyme where proteins are degraded)




Bacteria: Lon (an ATP dependent protease) and other proteases

What are the 7 processes that can affect the steady-state concentration of a protein?

1. Transcription


2. Posttranscriptional processing


3. mRNA degradation


4. Translation


5. Posttranslational processing


6. Protein degradation


7. Protein trageting and transoprt

Gene regulatory mechanisms

1. Transcriptinal mechanims (promoter and RNA polymerases)




2. RNA porcessing (5´capping, 3´poly-adenylation, splicing and alternative splicing)




3. Translational mechanisms (miRNA inhibit translation and degrade mRNA, silencer RNAs degrading mRNA)




3. Epigenetic mechanisms (DNA methylation, histone modification, chromatin remodeling)

What are enhancers elements?

DNA sequence that increase theactivities of many promoters in eucaryotes.Enhancers function by serving as binding sites for specificregulatory proteins. An enhancer is effective only in the specific cell types in whichappropriate regulatory proteins are expressed.

What are the 3 transmit messages between cells?

1. Neurotransmitters - nervous system


2. Hormones - Endocrine system


3. Cytokines - immune system

Peptide hormones

Insulin, glucagon, somatostatin, hypothalamic (releasing) hormones, pituitary hormones




Synthesis: proteolytic cleavage of the precursor proteins




Mode of action: bind to receptors in the plasma membrane

Catecholamine hormones

Epinephrine and norepinephrine




Synthesis: brain, adrenal glands




Mode of action: act through plasmamembrane receptors




Effects: increased heart rate and blood pressure (increased delivery of O2 to tissues). Increased glucose and ATP production, fatty acid mobilization. Increased glucagon secretion and decreased insulin secretion.

Eicosanoids (fatty acid derivatives)

Derived from polyunsaturated fats and produced only when needed. They are paracrine hormones.




Prostaglandins, thromboxanes and leukotrienes




Mode of action: act through plasma membrane receptors.

What is the role of phospholipase A2?

- Arachidonate --> leukotrienes by lipoxygenase.


- Arachidonate --> prostaglandins and thromboxjnes by cyclooxygenase.

- Phospholiase A convert phospholipids to arachidonate.




- Arachidonate --> leukotrienes by lipoxygenase.




- Arachidonate --> prostaglandins and thromboxjnes by cyclooxygenase.

Steroids

Steroid hormones are endocrine hormones.


Derived form cholesterol: Cortisol, progesterone, testosteron.





What is the mode of action of steroid hormones?

Vitamin D

Mode of action: acts through nuclear receptors (intestinal Ca2+-binding protein) --> uptake of dietary Ca2+




Effects: works in Ca2+ homeostasis,regulating balance between Ca2+ deposition and Ca2+ mobilization from bone.

Retionic acid hormones

Regulation of gene expression


Biosynthesis: In liver from vitamine-A


Nuclear receptors

Thyroid hormones

Thyroglobulin-Tyr ---> Thyroglobulin-Tyr-I (iodinated Tyr residues) ---> Thyroxine (T4) and triiodothyronine (T3)




Mode of action: through nuclear receptors




Effects: stimulate energymetabolism in theliver and muscle



Describe the desensitization of the beta-adrenergic receptor by epinephrine.

The genes which expression are supressed by insulin.

PEP carboxykinase (gluconeogenesis)


Glucose-6-phosphatase (release glucose to blood)



The genes which expression are increased by insulin.

Glycolysis: Hexokinase 2, Hexokinase 4, phosphofructokinase-1, pyruvate kinase




Regulation of glycolysis/gluconeogenesis: PFK-2/FBPase-2




Pentose phosphate pathway: glucose-6-phosphate dehydrogenase, 6-phosphogluconate dehydrogenase




Fatty acid synthesis: pyruvate dehydrogenase, acetyl-CoA carboxylase, malice enzyme, ATP-citrate lyase, fatty acid synthase complex



What enzymes are regulated by PKA?

- Glycogen synthase (glycogen synthesis)


- Pyruvate kinase (glycolysis)


- Pyruvate dehydrogenase complex (pyruvate to acetyl-CoA)


- Phosphofructokinase-2/fructose-2,6-bisphospatase (glycolysis/gluconeogenesis)


- Hormone-sensitive lipase (triacylglycerol mobilization and fatty acid oxidation)

What are the metabolic functions of PKB?

- Translation initiation


- Translation elongation


- Increase glucose uptake (by stimulating GLUT4 transporter)


- Stimulate glycolysis (by increase prod. of PFK2)


- Increase glycogen synthesis (by inhibiting GSK-3)

What is AMPK stimulated by and what does it stumulates?

It is stimulated by stress, exercise, fasting ( increased [AMP])




It stimulates glucose uptake and transport, glycolysis, fatty acid uptake, beta-oxidation

What does AMPK inhibit?

HMG-CoA reductase (cholesterol synthesis)


Fatty acid synthesis (ACC - acetyl-CoA carboxylase)


Insulin secretion


Transcription

What happens if there is hypoxia (low pO2)?

1. Increased transcription of HIF-1α


2. HIF-1α is phosphorylated


3. HIF-1α increases transcription of other enzymes and proteins (glucose transporter, glycolytic enzymes, lactate dehycrognease, protease)


4. --> ATP production by glycolysis increases and complex IV properties are adapted to low pO2.



What happens if there is normoxia?

What is the transcriptional regulation of cholesterol synthesis?

SREBP activates HMG-COA reductase as a transcription factor.


SCAP (cleavage-activating protein) prevents release of SREBP




LDL receptor protein ↑


Pyruvate kinase ↑


Hexokinase 4 ↑


Lipoprotein lipase ↑


Acetyl-CoA carboxylase ↑




Glucose 6-phosphatase ↓


PEP carboxykinase ↓F


BPase-1 ↓

Which enzymes uses TPP (thiamin diphosphate) as a cofactor?

Catalyze oxidative decarboxylation:


- Pyruvate dehydrognease


- a-ketoglutarate dehydrogenase complex


- Branched-chain keto-acid dehydrogenase


- Pyruvate decarboxylase (in bacteria)




Also a coenzyme for transketolase in pentose phosphate pathway.

Which enzyme are vitamin B12 dependent enzymes?

- Methymalonyl CoA mutase


- Methionine synthase

Which enzymes uses biotin as a coenzyme?

- Acetyl-CoA carboxylase


- Pyruvate carboxylase


- Propionyl-CoA carboxylase


- Methylcrotonyl-CoA carboxylase

What function has riboflavin (B2)?

Water-soluble vitamin


Coenzyme in oxidation and reduction reactions (FAD)


Prosthetic group of flavoproteins

What function has nicotinic acid/nicotinamide (niacin)?

Coenzyme in oxidation and reduction reactions, functional part of NAD and NADP.

Lipid-soluble vitamins

Hydrophobic compounds. Can be absorbed efficiently only whenthere is normal fatabsorption Are transported in theblood in lipoproteins orattached to specificbinding proteins.




D - Calciferol


E - Tocopherols, tocotrienols


K - Phylloquinone: menaquinones


A - Retinol, b-Carotene

Water-soluble vitamins

Hydrophilic compounds. Function mainly as enzymeco-factors.




B1 - Thiamin


B2 - Riboflavin


B3 - Niacin


B6 - Pyridoxine


B9 - Folic acid


B12 - Cobalamin


B5 - Panthotenic Acid


B7 ;H-Biotin


C - Ascorbic Acid

What are the different types of glucose transporters and their function?


What is the difference between hexokinase II and IV?

Hexokinase II is in muscle cells, have high affinity for glucose and is inhibited by its product glucose-6-phosphate.




Hexokinase IV (glucokinase) is in liver cells, have low affinity for glucose and is inhibited by a regulatory protein (not G6P). It is activated by glucose.

How is phosphofructokinase-1 (PFK1) regulated?

Regulated allostericly




Activated by ADP, AMP and fructose-2,6-bisphosphate (F2,6BP).




Inhibited by ATP and citrate.

What is the role of fructose-2,6-bisphosphate in regulation of glycolysis and gluconeogenesis?

 F2,6P is formed by phosphofructokinase 2 (PFK2). PFK2 is activated by insulin and inhibited by glucagon (↑ [cAMP])

F2,6P is formed by phosphofructokinase 2 (PFK2). It is an allosteric regulator of PFK-1 and FBPase-1. PFK2 is activated by insulin and inhibited by glucagon (↑ [cAMP]).

How is pyruvate kinase regulated?

Activated by fructose 1,6-bisphosphate


Inhibited by ATP, alanine, acetyl CoA, long chain fatty acids.




Glucagon stimulates phosphorylation of pyruvate kinase, which inactivates the enzyme. This happens only in the liver.

What are the 3 possible catabolic fates of the pyruvate formed in glycolysis?

Gluocose --> 2 pyruvate


1. 2 lactate


2. 2 acetyl- CoA (enter citric acid cycle)


2. 2 ethanol + 2 CO2

One reaction which acetyl-CoA inhibit and one reaction which acetyl-CoA stimulate.

Inhibit: pyruvate kinase, pyruvate dehydrogenase complex




Stimulate: pyruvate carboxylase



How is pyruvate dehydrogenase complex (PDC) regulated?

Pyruvate dehydrogenase kinase (PDK) phosphorylates PDC --> INACTIVE


Pyruvate dehydrogenase phosphatase (PDP), dephosphorylates PDC ---> ACTIVE




PDK is stimulated by NADH and acetyl-CoA, and inhibited by CoA-SH, NAD+, ADP, pyruvate.




PDP is stimulated by Ca2+, Mg2+, insulin

How is the citric acid cycle regulated?

How is the gluconeogenesis regulated hormonally?

Insulin: inhibit


Glucagon: stimulate


Epinephrine, norepinephrine: stimulate


Glucocorticoids: stimulate

How is the glycogen synthase regulated?

GSK3 = glycogen synthase kinase 3.


Casein kinase phosphorylates GSK3 for activation.




PP1 =phosphorylase a phosphatase.

What are the different enzymes that regulates different processes in lipid metabolism?




(biosynthesis of FA, β-oxidation, cholesterol biosynthesis, ketonebodies, signal transduction)

- Regulation of biosynthesis of FA: ACC


- Regulation of β-oxidation: CAT


- Regulation of cholesterol biosynthesis: HMG-CoA-reductase, transcriptional regulation


- Regulation of ketonebodies: succinyl-CoA


- Regulation of lipid metabolism by signal transduction (AMPK, PPAR)

What are the hormones that regulates lipid metabolism?

Insulin, glucagon, glucocorticoids, epinephrine

In which intracellular compartments does the different lipid metabolism processes happen?

Cytosol: Fatty acid synthesis




ER: phospholipid synthesis, fatty acid elongation, fatty acid desaturation




Mitochondria: fatty acid oxidation, acetyl-CoA production, ketone body synthesis, fatty acid elongation

How is Acetyl-CoA carboxylase (ACC) regulated?

Activated by: insulin, citrate




Inhibited by: glucagon, adrenaline, AMP, palmitoyl-CoA

How is the formation of ketone bodies regulated?

Regulation of ketone body synthesis happens via HMG-CoA synthase.




Stimulated by acetyl CoA.


Inhibited covalently by succinyl-CoA

Write with structures the reaction that is defective in phenylketonuria (PKU).

Defect phenylalanine hydroxylase.

Defect phenylalanine hydroxylase.

Write with structures the reaction that is defective in Maple syrup urine disease.

Defect branched-chain alfa-kets acid dehydrogenase complex.

Defect branched-chain alfa-kets acid dehydrogenase complex.

Write with structures the reaction that is defective in Tyrosinemia I.

Defect fumarylacetoacetase.

Defect fumarylacetoacetase.

Write with structures the reaction that is defective in Tyrosinemia II.

Defect tyrosine aminotransferase.

Defect tyrosine aminotransferase.

Write with structures the reaction that is defective in Tyrosinemia III.

Defect p-hydroxyphenylpyruvtae dioxygenase.

Defect p-hydroxyphenylpyruvtae dioxygenase.

Write with structures the reaction that is defective in Citrullinemia type I.

Argininosuccintae synthetase deficiency.

Argininosuccintae synthetase deficiency.

Write with structures the reaction that is defective in argininocsuccinate aciduria.

Argininosuccinase deficiency

Argininosuccinase deficiency

Write with structures the reaction that is defective in homocytinuria I.

Cystathionine ß synthase deficiency.

Cystathionine ß synthase deficiency.

Write with structures the reaction that is defective in argininemia.

Argininase deficiency.

Argininase deficiency.

Write with structures the reaction that is defective in alkaptonuria.

Homogentisate-1,2-dioxygenase deficiency.

Homogentisate-1,2-dioxygenase deficiency.

Write with structures the reaction that is defective in methylmalonic aciduria.

Methylmalonyl-CoA mutase deficiency.

Methylmalonyl-CoA mutase deficiency.

What is the function of intestinal lipase?

Degradation of triglycerols to diacylglyderides and monacylgylcerols.

What is the function of apoprotein?

Lipid-binding protein in blood, transport phospholipids, cholesterol and cholesterol esters between organs.

What is the function of lipoprotein lipase?

Converts triacylglycerols to factty acids and glycerol.

What is the function of hormone-sensitive lipase?

Mobilization of stored triacylglycerols un adipocytes.




Stimulated by glucagon/adrenaline


-- > PKA activates hormone-sensitive lipase through phosporylation --> triacylglycerols are hydrolyzed --> free fatty acids are released into the blood.

What is the function of serum albumin in lipid metabolism?

Binds fatty acids and transports them through the blood.

What is the CORI cycle

When lactate is formed by active skeletal muscles (or RBC), it can be recycled. It is carried to the liver by the blood, and in the liver it is first converted to pyruvate, and then to glucose by gluconeogenesis. The glucose then returns to muscl...

When lactate is formed by active skeletal muscles (or RBC), it can be recycled. It is carried to the liver by the blood, and in the liver it is first converted to pyruvate, and then to glucose by gluconeogenesis. The glucose then returns to muscle through the blood and is again used for energy.

Galactosemia

Defect in either glactokinase, glactose-1-phosphate uridylyltransferase or UDP-glucose-4-epimerase.

What are the inhibitors of complex I, III and IV in the respiratory chain and the ATP synthase?

Complex I: amytal, rotenone, piercidin A




Complex III: antimycin A, myoxothiazol




Complex IV: CN- (cyanide) and CO




ATP synthase: oligomycin, venturicidin



What are the most common uncoupling agents in the ATP synthesis?

DNP (2,4-dinitrophenol


FCCP




Causing the oxidation to continue even if ATP synthesis is blocked.

What is the function of the malate-aspartate shuttle?

System that transport the NADH generated by glycolysis in the cytosol into the mitochondrial matrix. The mitochondrial membrane is not permeable to NADH.

Functions in liver, kidney, and heart mitochondria.

System that transport the NADH generated by glycolysis in the cytosol into the mitochondrial matrix. The mitochondrial membrane is not permeable to NADH.




Functions in liver, kidney, and heart mitochondria.

Glycerol-3-phosphate shuttle

NADH shuttle used in skeletal muscle and brain. 

NADH in cytosol is converted to FADH2 in the mitochondrial matrix
Electrons are transported to complex II.

NADH shuttle used in skeletal muscle and brain.




NADH in cytosol is converted to FADH2 in the mitochondrial matrix


Electrons are transported to complex II.

What are the sources of reactive oxygen species (ROS)?

- CoQ of the respiratory chain


- Production of ROS in the peroxisome


- Cytochrome P450 monoxygenases

What is the function of antioxidants and name some examples?

Enzymes and other molecules that help protect the cell from harmfull effects of ROS.




Superoxide dismutase


Catalase


Glutathione: oxidized to donate reducing equivalents to regenerate oxidized cellular molecules

Where does the gluconeogenesis takes place and what is its function?

Takes place in the liver.


Function is to maintain the blood glucose levels during fasting.

How is the gluconeogenesis regulated?

Activators: Acetyl Coa, glucagon, epinephrine, norepinephrine, glucocorticoids




Activators of glycolysis (inhibitor of gluconeogensis): ADP, AMP, citrate, fructose-2,6-bisphosphate, insulin.

What is the function of the penstose phosphate pathway and in which cells is it present?

Function: production of NADPH, RNA, DNA, ATP, FADH2.




Location: rapidly dividing cells, such as those of the bone marrow, skin and intestinal mucosa.

Which pathways requires NADPH?

Synthesis:


- fatty acid biosynthesis


- cholesterol biosynthesis


- neurotransmitter biosynthesis


- nucleotide biosynthesis




Detoxification:


- reduction of oxidized glutathione


- cytochrome P450 monooxygenases

What does glucose-6-dehydrogenase deficiency lead to?

Glucose-6-deficency lead to NADPH deficiency.




NADPH maintains the supply of reduced glutathione in the cells that is used to remove free radicals that cause oxidative damage.


Gluthahtione peroxidase: 2GSH + H2O2 ---> GSSG + 2H2O.




The G6PD / NADPH pathway is the only source of reduced glutathione in red blood cells (erythrocytes). If there is not enough reduced glutathione in the cells the free radicals produce Heinz bodies in the RBC, which is denatured hemoglobin caused by oxidative stress.



What is the Wernicke-korsakoff syndrome?

Mutation in transketolase gene, affinity to TPP is decreased --> thiamine deficiency --> severe memory loss, mental confusion, partial paralysis

Where is glycogen stored and what is the function?

Glycogen is stored in liver and skeletal muscle.




Liver glycogen is used to maintain blood glucose levels.




Muscle glycogen is used to generate ATP for muscle contraction when is needed.

Which enzymes are needed for glycogen synthesis?

- Hexokinas/glucokinase


- Phosphoglucomutase


- UDP-glucose pyrophosphorylase


- Glycogen synthase


- Glycogen branching enzyme

What is the function of glucogenin?

Primer for glycogen synthesis.

Which enzymes are needed for glycogen degradation (glycogenolysis)?

- Glycogen phosporylase


- Debranching enzyme


- Phosphoglucomutase


- Glucose-6-phosphatase

How is glycogen phosphorylase regulated?

PP1 is activated by insulin (glucose in the liver).

PP1 is activated by insulin (glucose in the liver).

How many citrate molecules are needed frommitochondria to cytosol to synthesize palmitate?

8 (Synthesis of palmitate need 8 acetyl-CoA = 8 citrate)




Acetyl CoA from pyruvate cannot be transported through the mitochondrial membrane to the cytosol, so it forms citrate together with oxaloactetate, which is transported out through a citrate transporter.




In the cytosol, citrate is reconverted to acetyl-CoA and oxaloacetate, and acetyl CoA process to fatty acid synthesis in the cytosol. Oxaloacetate is reduced to malte and is transported back into the mitochondria.

What is special with the desaturation of fatty acids in animals?

Animals can only put double bonds before the 10th carbon.




Therefore it is essential for animals to have fatty acids with double bonds >10 in their diet, like lineolate (--> arachidonate)

What inhibit the production of prostaglandins, thromboxjnes, leukotrienes?

Steroids inhibit the synthesis of arachidonate from phospholipids ---> no prostaglandins, thromboxjnes or leukotrienes.




Non-steroids (aspirin, ibuprofen) inhibit COX (cyclooxygenase) ---> prostaglandins or thromboxjnes.



What is the common precursor for both ketone bodies and cholesterol, and what is the enzymes for the different synthesis reactions?

HMG-CoA (beta-hydroxy-beta-methylglutaryl CoA)




HMG-CoA lyase --> acetoacetate (ketone body)


--> acetone (acetoacetate decarboxylase) and D-beta-hydroxybutyrate (Hydroxybutyrate dehydrogenase)




HMG-CoA reductase --> mevalonate (cholesterol synthesis)

What is the different lipoproteins?

Chylomicrons


VLDL


LDL


HDL

What regulates the cholesterol synthesis?

Activators: insulin




Inhibitors: glucagon, cholesterol, metabolites

What are the fates of cholesterol?

Propionemia

Deficiency in the enzyme propionyl-CoA carboxylase, inhibiting the conversion of propionyl-CoA to D-methylmalonyl CoA.

Methylmalonyl-uria

Deficiency of the enzyme methylmalonyl-CoA wpimerase, inhibiting D-methylmalonyl CoA to L-methylmalonyl CoA.

What are the fates for the amino group and the carbon skeleton of an amino acid?

Amino group: urea cycle




Carbon skeleton: citric acid cycle

What are the transporters of NH4+ from amin-acids to urea cycle?

Glutamate and glutamine




The amino group is transferred through transamination.

How is the transamination reactions?

alfa-ketogluatarate + NH4+ --> glutamate.

Glutamate + NH4+ --> glutamine.

An amino acid without NH4+ = alfa-keto acid.

alfa-ketogluatarate + NH4+ --> glutamate.




Glutamate + NH4+ --> glutamine.




An amino acid without NH4+ = alfa-keto acid.





What is oxidative deamination?

the release of NH4+ from glutamate, forming alfa-ketoglutarate. The enzyme for this reaction is glutamate dehydrogenase, which is the only enzyme that use either NAD+/NADP+.

What is transdeamination?

Transamination + oxidative deamination = transdeamination

Transamination + oxidative deamination = transdeamination

What are the sources of NH4+ in the liver?

- Amino acids from ingested proteins


- Alanine from muscle (glucose alanine cycle)


- Glutamine from muscle and other tissues (brain)

What are the sources of ammonia NH3 in the urea cycle?

- Amino acid degradation (liver and muscle)


- Ammonia secretion in kidney tubule form glutamine


- Nucleotide degradation (pyrimidine)


- Intestinal bacteria produce it from amino acids and urea





What is the problem with ammonia?

Ammonia is very toxic. It is quickly protonated to NH4+ in mitochondrial matrix and converted to urea, which is excreted in the kidney.




If the blood ammonium level increase it can cause cerebral edema and increased cranial pressure.

Which enzymes of the urea cycle is located in mitochondria and which is located in the cytosol?

Mitochondria: Carbamoyl phosphate synthetase I and ornithine transcarbamoylase




Cytosol: argininosuccinate synthetase, agininosuccinase and arginase

Which metabolic intermediates are common between citric acid cycle and urea cycle?

Aspartate, fumarate and malate

How is the urea cycle regulated? (short term and long term)

Long term: Regulation of the rates of the synthesis (transcriptional regulation) of the 4 urea cycle enzymes and carbamoyl phosphate synthetase I.




Short term: Allosterically activation. Arginine stimulates the synthesis of N-acetylglutamate by N-acetylglutamate synthase. N-acetylglutamate stimulates the formation of carbamoyl phosphate by carbamoyl phosphate synthetase I.



What are the difference between ketogenic and glycogenic amino acids?

Ketogenic: produce ketone bodies in the liver




Glucogenic: produce glucose and glycogen

Which amino acids are degraded to pyruvate? (6)

Alanine


Cysteine


Glycine


Serine


Threonine


Tryptophan

Which amino acids are degraded to alfa-ketoglutarate? (5)

Arginine


Glutamate


Glutamine


Histidine


Proline

Which amino acids are converted to succinyl-CoA (4)

Isoleucine


Methionine


Threonine


Valine

Which amino acids are converted to fumarate (2)

Phenylalanine


Tyrosine

Which amino acids are converted to oxaloacetate? (2)

Aspargine


Aspartate

How is the enzyme glutamine synthetase regulated? (allosteric and covalent)

Allosteric regulation: Each inhibitor produce partial inhibition, all inhibitors together shut down the enzyme.


1. Glycine


2. Alanine


3. Glucosamine-6-phosphate


4. Histidine


5. CTP


6. Carbamoyl phosphate


7. Tryptophan


8. AMP




Covalent regulation: adenylation --> inactive


- Adenylation = (AMP) molecule is covalently attached to a protein side chain


- Adenylation stimulated by: glutamine, Pi


- Deadenylation stimulated by: ATP, alfa-ketoglutarate

What is the most important antioxidant in RBC and what is the active amino acid making the anti oxidation effect?

Gluthathione


Antioxidation effect: cysteine




It protects proteins for oxidation in erythrocytes. Removes toxic peroxided formed during growth and aerobic metabolism: glutathione peroxidase reaction.




Produced form glutamate, glycine and cysteine.

What neurotransmitters is produced from the amino acids tyrosine, glutamate, histidine, tryptophan, methionine, arginine respectively?

Tyrosine: dopa, dopamine, norepinephrine, epinephrine




Glutamate: GABA (inhibitory neurotransmitter)




Histidine: histamine




Tryptophan: serotonin




Methionine: spermine, spermidine




Arginine: NO

The glucose-alanine cycle

Why is the overall energetic cost of urea synthesis is reduced?

Because 1 NADH is formed in the malate dehydrogenase reaction (aspartate-argininisuccinate shunt).

What is common between activation of fatty acids and activation of amino acids?

Both need energy. They uses ATP which are converted to AMP + PPi.

4 possible fates of fatty acids in the liver

1. Free fatty acids


2. Beta-oxidation --> Acetyl CoA




Acetyl-CoA:


3. Cholesterol synthesis


4. Ketone body synthesis


5. Citric acid cycle




6. Storage as lipids


7. lipoproteins

How many CO2 molecules are produced form one glucose molecule, what are the reactions producing those CO2?

6 CO2 molecules




- Pyryvate dehydrogenase complex reaction


- Isocitrate dehydrogenase reaction


- Alfa-ketoglutarate dehydrogenase complex reaction



What is the biological role of creatine and carnitine, ans what enzymes catalyzes them?

Creatine: phosphocreatine is an energy reservoir in skeletal muscle during light activity or rest. Can rapidly regenerate ATP from ADP


Creatine kinase: creatine <--> phospocreatine




Carnitine: transport long-chain fatty acids into the mitochondrion and regulate the intramitochondrial ratio of Acyl-CoA to free CoA.


Carnitine acyltransferase: carnitine + acyl-CoA <--> Acyl-carnitine

What is the role of DNA-dependent RNA polymerase?

An enzyme that produces primary transcript RNA. RNA polymerase is necessary for constructing RNA chains using DNA genes as templates, a process called transcription.

What is the role of RNA-dependent RNA polymerase?

RNA-dependent RNA polymerase or RNA replicase, is an enzyme that catalyzes the replication of RNA from an RNA template.

Name the substrates and the products of the light reaction of photosynthesis

2H2O + 2NADP+ ---> O2 2NADPH + 2H+

Write with structures the reaction where an inhibitory neurotransmitter is produced from anamino acid directly.

Glutamate decarboxylase reaction.

Glutamate decarboxylase reaction.

The simultaneous presence of which amino acid side chain and which positively charged ionis necessary for the chain elongation step of nucleic acid biosynthesis?

Aspartate, Mg2+

What are the small RNAs other than microRNA that have a role in splicing and rRNA maturation respectively?

Splicing: snRNA (small nuclear) - part of the spliceosome.




Maturation of rRNA: snoRNA (small nucleolar)

What is called the secondary genetic code?

= proofreading activity


Amino acid tRNA synthetase and tRNA interaction will recognize the tRNA that belongs to which amino acid. Get right anticodon forthe right amino acid.

Namea cell type that does not express MHC I on its surface.

RBCand neurons only

What can happen to glucose-6-phosphate in the liver?

1. Glycolysis: glucophospho isomerase (energy production)




2. Gluconeogenesis: glucose 6-phosphatase (remove P toincrease blood sugar concentration)



3. Pentosephosphate pathway: Glucose 6-phosphatedehydrogenase (Ribose and NADPH production)




4. Glycogen synthesis: phosphoglucomutase (glycogen storage)

Which transcription factor is the most important regulator in the expression of erythropoietin?

HIF1 α (Hypoxia-inducible factor 1, α unit) --> Erythropoietin --> RBC production (since oxygen levelaccommodate to it)

How to calculate the maximum amount of ATP produced from fatty acid degradation to CO2 and H2O?

1. Number of Carbons/2 = Number of Acetyl CoA formed




2. Number of rounds in the Beta-oxidation necessary for converting the whole fatty acid to Acetyl Co A units = Number of Acetyl CoA - 1 = NADH and FADH2




3. NADH yields 2.5 ATP and each FADH2 yields 1.5 ATP. Multiply the number of rounds with 4.




4. Multiply the number of Acetyl CoA times 10 (10 ATP in each round of citric acid cycle)




5. Minus two ATP that were used for the activation of the fatty Acid.

Why is it advantageous for the long fasting man that his brain is able to utilize ketone bodies(produced by the liver) as a fuel?

Ketone bodies are produced from fat, converted to acetyl-CoA and then transported to the brain where it is used for energy. This prevent amino acid degradation, ketone bodies is used instead.

4 factors/accommodations for transport ofGlc-6-P out of cell as free glucose

1. High glucagon levels = low glucose levels in the blood


2. Glucose-6-phosphatase enzyme (only in the liver) - covert glc-6-P --> glucose


3. GLUT7 transporter (transport glucose from ER


4. GLUT 2 transporter (release glucose to the bloodstream)

4 enzymes that release ammonia (NH3) directly

Glutamate dehydrogenase


Glutaminase


Serine dehydratase


Adenosine deaminase

Which immunoglobulin participate in the fetuspassive immune system. Why?

Passive immunity = when maternal antibodies are transferred to the fetus through the placenta.




IgG is the only immunoglobulin that can pass through the placenta. It protect against bacterial and viral infections in fetuses.

Name 3 reactions in the animal tissue as possible sources of NADPH.

Malic enzyme: malate <--> pyruvate




Glucose-6-phosphate dehydrogenase: glc-6-P <--> 6-phosphoglucono lactone




6-phosphogluconate dehydrogenase: 6-phosphogluconate --> ribulose-5-P

Which mitochondrial enzymes are required during conversion of glucose to fatty acid?

Pyruvat-Dehydrogenase-complex


Citrat-Synthase

What does processivity mean, in relation to polymerases?

Nucleotides added before the polymerase dissociates.




Polymerase I: 3-200


Polymerase II: 1500


Polymerase III: > 500 000



What is the Shine-Dalgarnosequence and what is its biological role?

Ribosomal binding site inthe mRNA, generally located 8 basepairs upstreams for the AUG start codon.

List hormone-familiesthat have nuclear receptors.

Retinoic acid


Vitamin D


Thyroid


Steriod

Which catalytic function ismissing in retroviral reverse-transcriptase compared to human and E coli DNApolymerase?

Proofreading action. It is not able to dorepair on the template, and mutations are very common (may cause immunityagainst drugs).

Name two metabolic pathwayswhich take place partially in the mitochondria and partially in the cytosol.

Gluconeogenesis


Urea cycle



Possible Fates of amino acids in the liver

1. Liver proteins


2. Nucleotides


3. Hormones


4. Porphorins


5. Urea Cycle


6. Gluconeogenesis


7. Acetyl-CoA:


TCA cycle


Fatty Acids

By whatmechanism can a G protein coupled hormone receptor regulate gene expression?

G protein --> Adenylate Cyclase --> cAMP --> PKA

What are the modifications on individual amino acids in posttranslational modification?

Phosphorylation, carboxylation, methylation, sulfation, hydroxylation on the side chains.

What are the different posttranslational modifications in protein synthesis?

1. Modifications on both terminal ends


2. Loss of sinal sequences


3. Modifications of individual amino acids


4. Glycosylation: attachment of carbohydrate side chains


5. Acylation:prenylation, farnesylation


acetylation/deacetylation


6. Addition of prosthetic groups


7. Proteolytic processing


8. Disulfide crosslink formation

What is the function of dolichol in post translational modification of proteins?

It has an isoprene structure that is important in the glycosylation of proteins.

Put in order of highest protein content of lipoproteins.

HDL > LDL> IDL > VLDL > chylomicron

Put in order of highest cholesterol content of lipoproteins.

LDL > HDL > IDL > VLDL >chylomicrons

What is the biological roleof the reaction catalyzed by cytochrome P450?

It detoxifiesthe body of drugs and toxins by hydroxylation.

Name 5 types of antibodies found in humans.

IgM, A, D, G,E.

Which antibody is mainly involved in allergic reactions?

IgE

List 3 differences between DNA and RNA polymerases

RNA polymerasedoesn’t need a primer


RNA polymerasehas no 3’->5’ exonuclease activity


No proofreading

What distinguishes the newly synthesized DNA strand from thetemplate?

The template ismethylated.

What is the role MutH/MutS/MutL?

They are the main components of methyl directed mismatch repair. They winds up the DNA molecule.

Name the factors responsible for promotor recognition inprokaryotes and eukaryotes.

Prokaryotes:Sigma Factor


Eukaryotes:TATA binding protein

What are the heme containing complexes of the respiratory chain?

Complex IV


Cytochrome C


Complex III

What is an allosteric inhibitor of carnitine acyl transferase?

Malonyl-CoA

In which glycolytic reaction is a C-C bond broken?

Aldolase reaction




Fructose-1,6-bisphosphate <--> dihydroxyacetonephosphate + glyceraldehydphosphate

Which enzyme enables yeast to synthesize glucose from acetyl-CoA

Isocitrate lyase


Malate synthase




Glyoxylate cycle



Name the 4 vitamins that give cofactors participating in alpha-ketoglutarate dehydrogenase reaction.

TPP (B1) -> Thiamine


FAD (B2) -> Riboflavin


CoA-SH (B5) -> Pantothenic acid


NAD (B3) -> Niacin

What are the irreversible steps in glycolysis?

Hexokinase (1), phosphofructokinase 1 (3), pyruvate kinase (10)

What are the irreversible steps in gluconeogenesis?

Pyruvate carboxylase(1), PEP carboxykinase (2), fructose-1,6-bisphosphatase (9), glucose 6-phosphatas (11)

4 fates of pyruvate in the liver + enzyme

1. Gluconeogenesis


- Pyruvate carboxylase, PEP carboxykinase, Enolase, Phosphoglycerat mutase, Phosphoglycerate kinase, glyceraldehyd phosphate dehydrogenase, triose phosphate isomerase, aldolase, fructose-1,6-bisphosphatase, phosphohexose isomerase glucose-6-phosphatase




2. Acetyl-Coa


-Aerobic


- Pyruvate dehydrogenase complex


3. Lactate


-Anaerobic


- Lactate dehydrogenase




4. Oxaloacetate


-Pyruvate carboxylase



Albinism(structure of reaction

An amino acid metabolism disorder
Either tyrosine absent/nonfunctional or cell membrane tyrosine deficiency

An amino acid metabolism disorder


Either tyrosine absent/nonfunctional or cell membrane tyrosine deficiency

Maximum ATP from fructose-1-p to H2O and CO2

F-1-p -> dihydroxyacetone-phosphate and glyceraldehyde. Glyceraldehyde needs to become phosphorylated, this means that 1 ATP is invested during glycolysis




31 or 33 depending on which transport is used



How cholesterol regulates it´s own mechanism

- 4 mechanism


Mostly regulated by changes in HMG-CoA reductase


1. Rate of synthesis of HMG-CoA reductase mRNA is regulated by sterol regulatory element binding protein - transcriptional regulation of cholesterole synthesis


2. Rate of translation of HMG-Coa reductase mRNA is inhibited by nonsterol metabolites derived from mevalonate and dietary cholesterol


3. Degradation of HMG-CoA reductase


- Two domains, cytoplasmic and membrane


- If the membrane undergoes structural changes this can make the enzyme more susceptible to proteolysis


4. Phosphorylation decreases the activation of HMG-CoA reductase

Amino acids and vitamins in collagen

Amino acids -> Glycine, proline, methionine, serine, arginine,hydroxproline etc


Glycine, proline and hydroxproline are in abundant


- Glycine is every third residue




Vitamins -> Vitamin C is important for enzymes which synthesize hydroxyproline

Describe briefly mRNA processing

CAP


- Attached to 5'end of RNA with triphosphate group


- Protect mRNA


- Ribosome binding


Poly A tail


- Protection, transport, translational enhancement


Splicing


Editing


Ribozymes

Function of miRNa and piRNA

miRNA (micro): Post-transcriptional regulators, silencing of many genes




piRNA (piwi-interacting): transcriptional gene silencing of retrotransposons in germline cells.

What is the secondary genetic code?

= proofreading activity

Amino acid tRNA synthetase and tRNA interaction will recognize the tRNA that belongs to which amino acid. Get right anticodon for the right amino acid.

Regulation of glycogen phosphorylase

- Allosteric/covalent


- Inhibitors/activators



Glucose is an allosteric inhibitor of glycogen phosphorylase


Insulin will also inhibit by affecting Gm or glycogen targeting protein, and phosphorylating it once, causing it to be associated with the GM complex.


In the complex the glycogen phosphorylase is inactive.


- Glycogen is synthesized



Regulated allosterically and by phosphorylation



Phosphorylase b kinase will be upregulated by


- glucagon (liver)


- epinephrine, calcium and AMP in muscle

Molecules needed for nucleotide synthesis
There are two pathways to nucleotide synthesis

1. Activated ribose + base -> nucleotide


2. Activated ribose+amino acid + ATP + CO2 + etc -> nucleotide

Reaction in fatty acid synthesis which releases CO2

Malic enzyme

Ketoacyl synthase, aka 3-ketoacyl-ACP synthase



Hormones with nuclear receptors
Steroids

Cholesterol -> progesterone


- Mode of action through nuclear receptors


- Cortisol, aldosterone, testosterone




Thyroid hormones


Retinoid hormones

Which enzymes degrade RNA primer

Ribonuclease


- A family of non-sequence-specific endonucleases which catalyze the cleaving of RNA

What simulates PKA, PKC

PKA is dependent on cellular level of cAMP

- Activated only when cAMP is present


- Glucagon and epinephrine trigger actiivation cascade




PKC


- Activated by increased DAG concentration or Ca2+ ions


- Signal transduction cascades



Head to tail reaction in cholesterol synthesis
1. HMG-CoA synthase

2. HMG-CoA reductase


3. 1st Prenyl transferase


4. 2nd Prenyl transferase


?



Name of vitamins

- TPP, PLP, FAD, NAD

TPP - thiamine pyrophosphate


Pyridoxal phosphate


Flavin adenin dinucleotide


Nicotinamide adenine dinucleotide

Reactions that yield NH4 directly
Glutamate dehydrogenase

Glutaminase


Serine dehydratase



Inhibitors of translation in eukaryotes and prokaryotes respectively

Inhibits prokaryote- Streptomycin, tetracycline, chloramphenicol, erythromycin, pyromycin (both)Inhibits eukaryote- Cycloheximide, puromycin
How does arsenate and arsenite influence energy metabolism?

Arsenite inhibts pyruvate dehydrogenase complex


- No TCA and respiratory chain -> no ATP -> fatal




Arsenate is structurally analog to phosphate, and will inhibits reactions with organic phosphate incorporation -> respiratory chain still runs, but less ATP is produced

Glycogen binding protein


How is it phosphorylated by insulin and epinephrine

phosphorylated on different sites


Insulin phosphorylates GM directly


while epinephrine uses a second site on GM via cAMP -> PKA


- Inhibits glycogen synthesis and enhances glycogenlysis in muscle

Which histone acyltransferase activates HIF1-alpha in the nucleus?

p300

What enzyme, and how is the molecular pathway to increase blood sugar?

Epinephrine/glucagon -> caMP -> PKA -> activates enzymes of gluconeogenesis

Allosteric and covalent regulator of glycogen synthesis?

1. Insulin -> PKB -> GSK3 -> glycogen synthase covalent




2. Insulin -> PP1




3. Glucagon will causes PKA to phosphorylate glycogen synthase


- Cleave phosphate when bound


- Will causes glycogen degradation

What is the reaction when ADP is the only substate?

2ADP -> ATP + AMP

What are some lysosomal storage diseases?

Tay-sachs diseases


Niemann-Pick


Gaucher


Krabbes diseases



What is the molecular basis of Alzheimers diseases?

Accumulation of beta-amyeloid plaques


Tau protein fibrils

Through which molecule is PKA, PKC, and AMPK activated?

PKA -> cAMP


PKC -> Ca2+ and DAG


AMPK -> High cc of AMP, leptin

How is iron absorbed, transported and stored?

Absorbed as Fe2+ by DMT1


Transported as transferrin


Stored in apoferritin, in ferric state

Difference between RNA virus and retrovirus?

RNA virus -> no transverse RNAse, fitted random


Retrovirus -> transverse RNAse, own viral RNA is transcribed into cDNA and inserted into genome of cell

Which proteins participate in the replication fork of prokaryotes?

SSB, DNA gyrase, helicase

Name 3 toxins which inhibit RNA-ribosylation

Cholera, dypheteria, pertussis toxin

Name 3 proteins which participate in replication fork of E. Coli?

Ligase


Helicase


Sigmoid factor

What is the role of MutH, Mut L and MutS proteins?

MutH: Cleaves unmethylated strand


MutL and S: seen in DNA methyldgroups, controlling which strands are cleaved

Which hormone, in which direction and in which tissue regulates GLUT-4

Insulin, glucose uptake and in muscle and fat tissue

If a mitochondria is decoupled, what is it decoupled from? Give protein that can perform decoupling

Respiratory chain si decoupled form ATP synthase


- Oxidation of the resp. chain and phosphorylation is decoupled




Decoupler proteins -> thermogenin UCP-1, 2,5-DNP, UCP-2, UCP-3

Name basic mechanism and molecular process which guarantee accuracy of translation

Secondary genetic code


Wobble formation


Proofreading


Ribosomal with aminoacyl t-RNa synthase

What are the similarities between glucose-alanine cycle and cori cycle regarding function?

Glycolysis in muscle


Gluconeogenesis in liver


Glucose transport from liver to muscle

Name differences and similarities in pentose phosphate pathway and glycolysis

PPP: CO2 is released


Glycolysis: NAD+ is involved




Both have same intermediates


- Phosphate esters and fructose-6-phosphate

What are mechanism where interferons inhibit synthesis of viral protein/integration in human genome?

Viral infection of the virus mRNA is degraded and thus removed from cell




By RNA-dependent protein kinases of EIF2 which phosphorylates and inactivates transcription

What initiates activation of zymogens?

Enteropeptidase

How can ROS inhibit insulin signaling?

TNF-alpha can activate ASK/JNK pathway through ROS -> phosphorylation of IRS-1

Which enzymes take over the ammonium ion removal in portal and venous hepatocytes?

Glutaminase

Glutamine synthase



4 diseases related to glycogen/glucose and enzyme/reaction?

Von Gierke: glucose-6-phosphatase


Morbus Pompe: Alpha-1,4-glucidase


Cori syndrome: debranching enzyme


McArdle: Glycogen phosphorylase

Desensitization of beta-adenergic receptors with constant stimulation, which molecule are involved?

Desensitization by internalization of the receptor molecules: Arrestin, adrenorecepotrs, g-protein, Beta-ark

Name 6 types of small RNA molecules, beyond microRNA

siRNA, snRNA, piRNA, snoRNA, rasiRNA, qiRNA

What are antibiotics which inhibit the prokaryotic and eukaryotic translation?

Puromycin


- Embed into A site of both bacteria and eukaryotic ribosomes, and block translation


Actinomycin D


- Bind DNA, prevent RNA


Cycloheximide


- Selectively inhibits eukaryotic ribosomes in cytosol




Strepto mycin and tetracycline


- Example of inhibits which block highly specific bacterial ribosomes, without affecting the eukaryotic ribosomes

What are properties that only apply to RNA polymerases?

No proofreading


RNA pol doesn't need primer


RNA pol has no 3-5 exonuclease activity

How is the poly-a tail formed + enzymes


- in abbreveations

RNA+ nATP -> RNA + (AMP)n + PPi




Under polyadenylation is mean to attach a chain to end of 3 end of mRNA


Polydenylation sequence is AAUAAA followed by a GU or U rich sequence

In MELAS and MERRF, which genes are mutated in mtDNA?


Genes for tRNA and rRNA

Glut-4 vesicles, carry which protein with GTPase function when released?

Rab-GTP

What molecule is responsible for pH in lysosymes?

Acid hydrolyse


V-class proton pumps

Name a mixed function oxidase enzyme requiring vitamin C as a cofactor, and one that doesn't


What is the cofactor requirement for the latter enzyme?

Vitamin C:


Proline hydroxylase




Not required squalene monoxygenase


- Cofactor required: NADPH+H+

Conversion of pyruvate in alanincycle?

Alanine cycle transports toxic ammonia from muscle to liver




Muscle


- Alanine aminotransferase will transfer amino group to pyruvate -> alanine




Alanine is transported via blood to liver


At liver alanine is converted to pyruvate and ammonia


- Aminotransferase




Gluconeogenesis uses pyruvate, creating glucose for muscle

In which senses are 7TM receptors found?

Vision, smell and taste


- Sweet, bitter, umami

Which senses does not have the 7TM receptors?

Salty taste


Proprioception


Pain


Acoustic, hearing

How are introns type II spliced?

Selfsplicing, lariat structure


Mitochondrial, chloroplast mRNA

Structure of replisome?

Replisome complex: primase, DNA pol, helicase and SSB

ATP dependent reaction of urea cycle

In mitochondria: Carbomoyl-phosphate -synthase-1


In cytosol: Arginosuccinate synthase

Through which cellular compartment, and through which enzyme is HMG-Coa produced from ketone bodies and cholesterol?

Cytosol: HMG-CoA reductase -> Cholesterol synthesis


Mitochondria: HMG-CoA lyase -> ketone bodies

Give the name of the NADPH dependent reactions of cholesterol synthesis?

HMG-CoA reductase


Squalene synthase


Cyclization of squalene to lanosterol

Name an allosteric inhibitor and a reaction based inhibitor of carnitine acyltransferase 1

Allosteric inhibitor: Malonyl- CoA


Reaction based: Acetyl-CoA carboxylase

Give the name of reactions where NADPH is produced in RBC

Glucose-6-phosphate dehydrogenase


6-phosphogluconate dehydrogenase




Malic enzyme

What is a reaction where 2 trioses make a hexose?


- with structures

Aldolase reaction

What is the first common intermediate of lactate and glycerol in glucose formation with structural formula

Glyceraldehyde-3-phosphate

Give the name of mitochondrial enzymes for the conversion of glucose into fatty acid?

Acetyl-Coa for fatty acid synthesis


Pyruvate dehydrogenase complex


Citrate synthase


- citrate into cytosol by ATP-citrate lyase - > Acetyl-CoA

Why is mtDNA more fragile to ROS than DNA?

No repair mechanism


In the last step of resp. chain ROS can make a vicious cycle producing more ROS -> broken mitochondria

What is the the most common/strongest antioxiadant in humans?


What is it's reaction with hydrogen peroxide?

Glutathione


Glutathione peroxidase

What are 3 hormones that phosphorylated by glucagon indirectly via cAMP?

Phosphofructokinase-2


Pyruvate dehydrogenase complex


Glycogen phosphorylase kinase

Reaction/function with ATP, GTP, UTP, CTP, except for RNA/DNA biosynthesis, energy metabolism/storage

ATP: Signal transduction, second messenger (cAMP)


GTP: Protein synthesis


CTP: complex lipid synthesis


UTP: Conversion of galactose to glucose-phosphate

Why is not HIF-1 alpha cobalt hydroxylated or ubiquinated?

HIF-1 alpha has a transcription factor


Proliferation


When bound to Fe2+/3+ it is broken as hydroxylated and the ubiquinated and then degraded


Substituted cobalt Fe+ -> HiF-1 aplha is not -OH and not ubiqu

High blood suger


Which enzyme is active, and what is it´s action of glycogen phosphorylase?

Insuline sensitive protein kinases


It will inhibit glycogen phosphorylase, which degrades and causes no glycogenlysis -> blood sugar drops

Describe briefly the JAK-STAT pathway?

JAK= janus kinase


STAT= signal transducer and activator of transcription


JAK will phosphorylate STAT -> STAT dimerization and moves into nucleus where it participates in gene regulation and expression


Signal transduction via JAK-STAT protein is used by cytokines, interleukins, interferons and erythropoietin

What transport systems in the mitochondria needs vitamin B2 - riboflavin and vitamin B3 - niacin

Protonand electron transfere


Glyceraldehyde-3-p dehydrogenase


NADH transfers its electrons complex II

What vitamins are in the pentose phosphate pathway?

Vitamin B3 - niacin - because of NADP+


Vitamin B1 - thiamine -> TPP in the transketolase reactions

Two enzymes which has a soutbility and metabolite, which can act through DNA enhancement?

Vitamin A -> retinoic acid


Vitamin D -> calciferol

Why can elF2 be important for protein synthesis?

Forms a complex with tRNA and GTP in translation

Affected enzymes in galactosemia?

Galactokinase


Galactose-1-phosphate uridyl transferase


UDP- galactose- 4 epimerase

cataract formation by galactos-metabolite. Write reaction with structures

Galactokinase reaction

What reaction is related to Propionic acidemia

Proprionyl-Coa carboxylase reaction

Tyrosinemia type I

Fumarylacetoacetate

Tyrosinemia type II

tyrosine aminotransferase

Tyrsinemia type III

p-hydroxyphenyl pyruvate dehydrogenase

What activates PKA, PKB, PKC, PKR, AMPK, PKG

cAMP -> PKA


cGMP -> PKG


PDK-1 -> PKB


Ca2+, DAG and PDK -> PKC


AMP, leptin -> AMPK


dsRNA -> RKR

What intracellular enzymes are activated during hyperglycemia?

Excessive nutration load -> ROS -> H2O2 -> phosphorylation of IRS-1


mTOR, PKC, JNK, TNF, IKK can phopshorylate IRS-1 -> supressed insulin reistance eventually

Why is folic acid a target I cancer treatment?

Follicle acid antagnoist will inhibitconversion of folic acid to tetrahydrofolic acid


Indirectly inhibit purine synthesis, which stops rapidgrowth of cancer cells

Most potent endogenous vasocontrictor

Endothelin type 1

Enzymes of direct DNA repair?

O6-methylguanin-DNA methyltransferase


AlkB

What is the mechanism of ion channels selectivity?

Specific ions can enter due to specific arrangement of carboxyl groups in the ion channel

A reaction with direct release of ammonia?

Glutaminase


serine dehydratase



3 enzymes that contain copper?

Complex IV of resp chain


superoxide dismutase


Bilirubin oxidase

What is the protein structure vulnerability of dopaminergic cells in Parkinson's disease?

Parkin: Lack of ubiquination -> accumulation


Alpha-syncein stabilization


Protofibrils

Chaperone

Formation of correctly folded, functionally active, natic proteins -> not a part of the product

name motorproteins with antergrade and retrograde transport

Antegrade -> kinesin


Retrograde -> dynein

How does epinephrine/adrenaline regulate glycogen synthesis?



cAMP -> PKA pathway


will inhibit glycogen synthase via phosphorylation (covalent)


Activate glycogen phosphorylase via phosphorylation (covalent) -> By GM protein and inhibition of PP1

What are the sensory transmembrane proteins of the unfolded protein response


Sensor for UPR

IRE-1


PERK


ATF-6

How and why can muscle tranining inhibit mtDNA erros

training causes temporary increase in ROS -> NRF2 activation -> induction of genes for ROS homeostasis

Explain the mechansim of chaperone

Folding and quality control

3 enzymes with zinc

superoxide dismutase


Alcohol dehydrogenase


carbonic anhydrase

write the reaction in which the kidney compensates for metabolic acidosis

Carbonic anhydrase

Right shift

What enzymes remove ammonium ions from periportal and perivenous hepatocytes?

Glutamine synthase - perivenous


Carbamoyl phosphate synthethase - periportal

What is the effect of chloramphenicol in humans?

Antibiotics


Inhibits peptidyl transferase activity of 50s ribosomes in subunit of prokarytoes

What is the secondary genetic code?

It has proofreading activity


Interaction between an aminoacyl-tRNA synthethase and tRNA guarantee the accuracy of the protein biosynthesis

Role of THF in cancer?

Folic acid in active state -> THF -> purine synthesis

How can ROS inhibit insulin transduction

ROS- PARP activation -> apoptosis

What are the proteins in the replication fork of prokarytos?

SSB, helicase, primase, polymerase, ligase, topoismoserase, gyrase

3 diseases caused by 1. point mutation, 2. deletion and 3. recombination

1. Krebs diseases


2. Alpha thalassemia


3. Chronic leukemia

UV radiation causes what?


Repair mechanism?

Causes pyrimidine dimmers


Eukaryotes -> nucleotide excision repair


Prokaryotes -> photolyase

reaction and equation where a hexose arises

Fructose-1-phosphate dehydrogenase


Aldolase

Definition of sequence homology

Measurement of similarity of proteins and nucleotides

Heme containing components of respiration chain

complex III, Complex IV and cytochrome C

What changes does collagen have on inside and outside?

Inside -> hydroxylation and glycosylation


Outside-> aggregation of proteins

How does high blood sugar induce insulin release?

Glucose -> beta cells in pancreas -> glycolysis -> ATP -> Potassium channel -> depolarization > Ca2+ -> insulin release

What is the anti-inflammatory response of corticosteroids?

PLA-2 inhibitor


IkB activator


PPAR

Name 4 vitamins in the a-ketoglutatarate dehydrogenase complex

B1 -> thiamine - TPP


B2 -> Riboflavin -> FAD


B3 -> niacin -> NAD


B6 -> pyridoxine -> CoA?

Which component of telomerase determine new DNA sequence?

RNA

Which molecule is bilirubin conjugated to in liver?

Glucuronic acid -> water soluble

Mechanism in which insulin inhibits fatty synthesis?

Inhibits traicylglycerid lipase


activates acyl-CoA carboxylase

Inhibitor and activator of mitochondrial permeability

Activator -> pyridoxal phosphate


Inhibits -> cyclosporine

what are the prostatic groups in cytochrome oxidase

CuA


CuB


Heme a


Heme B

Enzymes need to make glucose from glycogen

Glycogen phosphorylase


Debranching enzyme


Glucose-6- mutase


Glucose-6-phosphatase

What reaction uses vitamin k as a catalyst?

Vitamin k dependent carboxylation

3 enzymes with ribozyme activity

Class I introns


23s RNA


Spliceosome


Hammerhead ribozyme

How is viagra inhibited?

Slidenafil inhibits PDA 5a

4 protein regulated by NF-k-B

cytokines


Chemokines


iNOs


COX II

What proteins are released from mitochondria during apoptosis?

Omi


cyt c


endo G


AIF


Diablo

What enzymes are needed for insertion of a DNA segment

Restriction endonuclease


DNA ligase

Which receptors does PPAR communicate with?

PXR

3 main groups of proteins produced in liver?

Steroids


Coagulation factor


Albumin

How are steroid hormones act on lipid storage?

Steroid bind to HRE -> PEP carboxylase -> expression of glycerol-3-phosphate decreased

Function of 14-3-3 protein in FOXO

Can bind to FOXO and keep it in the cytosol

how is Ca+ prostaglandin synthesis activated

Activated phospholipase A2 -> arachidonic acid -> prostaglandins

What is the standard abbrevation for the reaction for p450

R+O2+NADPH+H -> H2O + NADP+ ROH

Products of phospholipase A2?

Lysophospholipid and fatty acid from phospholid

What distinguishes the newly created from the template strand in DNA

Newly created is methylated

3 proteisn that bind to both ends of mRNA?

PAB


elF4F


PBR

difference between hemoglobin A and S

Hb-s has a point mutation in the 6 position

3 functions of Apo/lipoproteins

Endocytosis of lipoproteins


activation of LCAT


Receptor ligand in the liver

What are the products of the RNA pol I, II, III?

RNA POL I -> rRNA 18, 5.8, 28s


RNA pol II -> mRNA, snRNA


RNA pol III -> tRNA, 5sRNA

Name 2 DNA enzymes which can´t proofread?

Taq polymerase


DNA pol I

Which enzyme recognizes DNA break damage?

PARP

what are the steps in tRNA maturation?

Splicing

Remove the 5 leader


3 tail attaches to CCA


Base editing


3 modifications in the ER?

Glycolysation


Farnesylation


Modification of amino acids

What does the transcription factor FOXO do?

Stimulate synthesis of apoptotic proteins and cell cycle arrest

4 compounds released by CCK?

Trypsin


Pepsin


Chymotrypsin


Alpha amylase

subunits of RNA poly

2 alpha, beta, beta', omega and sigma

What genes does HIF-1 alpha regulate?

GLUT 4


EPO


EGF


VEGF

What is the same in smell and taste?

7TM receptors and g protein

Biological role of PARP?

Increased production of lipase


Fibroblast into adipocytes

Most important function of HSP 70?

A chaperone in protein folding

starting compound for sphingolipid biosynthesis?

Serine


Palmitoyl-Coa

6 proteins that are components of skeletal muslce?

Action, myosin, tropomyosin C, actinin, dynein, kinesin

2 mechanism by which PKA influences metabolism?

Glycogenesis


Activation of CREB to CRE -> gene expression

4 enzymes in live with glucose-6-p as substate?

Phosphohexose isomerase


Glucose-6-phosphatase


Glucose-6-phosphate dehydrogenase


Phosphoglucomutase

Which enzyme does RNA dependent DNA polymerization?

Telomerase

3 enzymes which use biotin as cofactor?

Pyruvate carboxylase


Acetyl-CoA carboxylase


Propionyl-CoA carboyxlase

What are two mitochondrial proteins that not imported into the mitochondria?

proteins of complexes 1,3, 4 and 5

What nuclear receptors enhances glucose uptake?

PARP- gamma

What process in fatty acid metabolism are influenced by PPAr in adipocytes

Intracellular receptors


Many ligands such as glucocorticoids stimulate lipolysis -

what is the name of the DNA polymerase in mitochondria, and how does it differ from nuclear DNA polymerase?

mtDNA polymerase gamma


Replication not limited to s-phase


Both strands are transcribed at once

Role of vitamin C in oxygen sensation in humans?

Cofactor in hydroxylation of HIF-1 alpha