• 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

1/301

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;

301 Cards in this Set

  • Front
  • Back

What are the different types of glucose transporters, and in which tissue are they found?

GLUT1


- RBC, brain


- transport across blood-brain barrier




GLUT 2


- Liver, kidney, pancreas,


- Regulation on insulin release




GLUT3


- Brain, placenta, others


- Uptake into neurons,




GLUT4


- Muscle, adipose tissue


- Insulin-mediated transport




GLUT5


- Kidney and gut


- Absorption of fructose

What are the main differences between hexokinase II and IV



Hexokinase II


- Muscle cells


- High affinity for glucose


- Inhibited by glucose-6-phosphate




Hexokinase IV


- Liver cells


- Low affinity for glucose


- Inhibited by regulator protein, activated by either glucose or fructose-6-phosphate


- Regulator protein will cause sequestration in the nucleus


- Not allosteric inhibited by glucose-6-phosphate

what are some substrate-level phosphorylations?

Glycolysis


- Phosphoglycerate kinase


- Pyruvate kinase




Kreb´s cycle


- Succinate-CoA lyase


- Phosphoenolpyruvate carboxykinase

How is the regulation of phosphofructokinase-1 and fructose bisphosphatase 1 connected?

They have a mutual regulator, F26BP which is an allosteric regulator of PFK-1 and FBPas-1


Insulin and glucagon is mediated by F26BP


PFK-1 is inactive without the presence of F26BP regulation




F26BP levels are regulated by PFK-2 and FBPase-2


When PFK2 is active


- F26BP levels rise, this increases, which stimulate PFK-1, which stimulates glycolysis


- Active due to stimulation of insulin on pp, and the cleaving of the phosphate




When FBPase-1 is active


-Levels of F26BP will decrease. This will stimulate FBPase-1, and since PFK-1 need F26BP regulation to function,it will decrease glycolysis


- Stimulate gluconeogenesis


- Glucagon will stimulate cAMP secretion, which stimulates PKA to phosphorylate



What regulates PFK-1 except for F26BP?

High ATP and citrate levels


- Inhibit PFK-1




High levels of ADP and AMP


- activation of PFK-1




High levels of AMP will also strongly allosterically inhibit F-1,6-bisphosphatase

Arsenate poisining, and what is a reaction which can use arsenate?

Arsenate AsO4, is structurally and chemically similar to phosphate. This allows many enzymes, which require phosphate to use arsenate instead, making it very dangerous.


Arsenate is very toxic




Glyceraldehyde-3-phosphate dehydrogenase


- Will create 1-arseno-3-phosphoglycerate instead

What are the types of pyruvate kinase?

Pyruvate kinase L


- Found in liver only, an isoenzyme of


- Regulated by glucagon via PKA (cAMP)


- Covalently regulated




Pyruvate kinase M


- Found in all other glycotic tissues, also liver


- Not regulated by glucagon, or covalently


- Activated by F16BP


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



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

1. Pyruvate -> ethanol + CO2


- Fermentation to ethanol in yeast


- Hypoxic or anaerobic condition in bacteria




2. Lactate


- Fermentation to lactate in very active muscles, erythrocytes


- Also occurs in some cells under aerobic, as they lack mitochondria


- Anaerobic conditions




3. Pyruvate -> Acetyl CoA


- Aerobic condition


- Enters citric acid cycle

What is the short- time and longtime regulation of glycolysis?

Short time


- PFK1, hexokinase, pyruvate kinase




Longer time


- Glucagon, epinephrine, insulin


* Regulate glucose transporters GLUTx

How does acetyl-CoA regulate pyruvate dehydrogenase complex and pyruvate carboxylase?

Acetyl-CoA is a positive allosteric modulator of pyruvate carboxylase


- Creates oxaloacetate, and continues in the gluconeogenesis




Acetyl-CoA is negative allosteric modulator of pyruvate dehydrogenase complex

When does acetyl-CoA accumulate?

When the cell energy needs are met


- the oxidative phosphorylation slows


- NADH/NAD+ ratio rises


- Change in ratio, will inhibit the citric acid cycle


- Acetyl-CoA accumulates

How is pyruvate dehydrogenase complex regulated?

Inhibited by ATP, acetyl-CoA, NADH and fatty acids


Activated by AMP, CoA, NAD+, Ca2+




It's phosphorylated by PDK kinase and dephosphorylated by pdk phosphatase.


Kinase is stimulated -> inhibited PDK


- NADH


- ATP


Kinase is inhibited -> activation of PDC


- CoA-SH


- NAD+


- ADP


- Pyruvate




Stimulation of phosphatase -> Dephosphorylation of PDC -> activation


- Ca2+


- Mg+


- Insulin

How is citrate synthase regulated?

Inhibited by NADH, succinyl- CoA, citrate, ATP


Activated by ADP

How is isocitrate dehydrogenase regulated?

Activated by Ca2+ and ADP


Inhibited by ATP

How is a-ketoglutarate dehydrogenase complex regulated?

Activated by Ca2+


Inhibited by succinyl-CoA, NADP


- Succinyl-CoA is the product

What enzyme does substrate-level phosphorylation in the citric acid cycle?

Succinyl-CoA synthase




CoA-S is removed and replaced with a Pi


- Pi is donated to the enzyme and replaced by oxygen.


- Pi is donated from enzyme to GDP -> GTP



What is the role of citric acid cycle in anabolism?

anabolism, we use energy to create a product


Citrate -> Fatty acids and steroids


A-ketoglutarate -> Glutamate


- Glutmate is used from glutamine, proline, arginine or purines




Succinyl-CoA -> porphyrins, heme


Oxaloacetate -> Aspartate, asparginine


- Asparagine -> pyrmidines


Oxaloacetate -> phosphoenolpyruvate


- PEP -> Glucose


- PEP -> serine, glycine, cysteine, phenylalanine, tyrosine, tryptofan

What are anaplerotic reactions? what are some anaplerotic reactions?

Reactions which replenish the intermediates of citric acid cycle, as they are removed for synthesis of glucose, fatty acids, amino acids etc.




Pyruvate to oxaloacetate


- Pyruvate carboxylase


Aspartate to oxaloacetate


- Reversed reaction of aspartate transaminase




Glutamate to a-ketoglutatarate


- Glutamate dehydrogenase




B-oxidation of odd chain fatty acid


- Final oxidation forms succinyl-CoA

How is hexokinase I, II, III and IV regulated?

Hexokinase I, II, III


- Inhibited by their own product -> Glucose-6-phosphate




Hexokinase IV - Glucokinase


- Not inhibited by glucose-6-phosphate


- regulated by sequestration in the nucleus via regulator protein


- Regulator protein is activated by -> inhibition of hexokinase IV


* Glucose and fructose-6-phosphate



How does galactose enter glycolysis?

Galactose -> Galactose-1-p -> UDP-galactose -> UDP-glucose -> Glucose-1-phosphate -> Glucose-6-phosphate



Galactokinase -> Gal-1-p


UDP-glucose:Galactose-1-phosphate uridyltransferase -> UDP-Gal


UDP-glucose 4-epimerase -> UDP-Glc


UDP-glucose: Galactose-1-phosphate uridyltransferase -> Gal-1-p


Phosphoglucomutase -> Gal-1-p





How does fructose enter glucolysis?

Fructose -> hexokinase


Enters at fructose-6-phosphate -> step 3

How does Mannose enter?

Mannose -> mannose-6-p


- Hexokinase




Mannose-6-p -> fructose-6-p


- Phosphomannose isomerase



How does fructose -1-phosphate enter glycolysis?

F-1-p -> glyceraldehyde + dihydroxyacetonephosphate


- F-1-p aldolase




Glyceraldehyde -> glyceraldehyde-3-p


- Triose kinase




Dihydroxyacetone phosphate -> glyceraldehyde -3-p


- triose phosphate isomerase




Enters at 6 stage of glycolysis

How does glycogen enter glycolysis?

Glycogen -> Glucose-1-p


- Phosphorylase



Glycogen-1-p -> glc-6-p


- Phosphoglucomutase

What is an enzyme in gluconeogenesis, found in both mitochondrial and cytosol, and why?

Mitochondrial malate dehydrogenase and cytosolic malate dehydrogenase




Pyruvate is converted to oxaloacetate by pyruvate carboxykinase


Oxaloacetate can't pass the mitochondrial membrane, and is therefore converted to malate by mitochondrial malate dehydrogenase.


When it has passed the mitochondrial membrane, it's converted back to oxaloacetate, and it continues the glucogeonetic pathway

What are the activators of gluconeogenesis and glycolysis?


Gluconeogenesis


- Acetyl-CoA, Glucagon, Epinephrine, Norepinephrine, Glucocorticoids




Glycolysis


- Inhibit gluconeogenesis


- ADP, AMP, citrate, fructose-2,6-bp, insulin

Cori cycle

Lactate is formed by active skeletal muscle in anaerobic environment or by erythrocytes.


Lactate is carried in the blood to the liver, where it´s converted back to pyruvic acid, then glucose via gluconeogenesis.


- Convertion requires 6ATP



Alanine cycle

Alanine transports ammonia from skeletal muscle to the liver


In the muscle, muscle protein is degraded to amino acids which inturn gives NH4+. In the alanine cycle we can transport both NH4 and pyruvate to the liver. Where NH4 enters urea cycle, and pyruvate enters gluconeogensis.




We can covert glutamate to glutamine and transport it to liver.


Or by alanine aminotransferase, we can transfer the amino group to pyruvate and transport both

What is the hormonal regulation of gluconeogenesis?

Insulin -> inhibit


Glucagon -> stimulate


Epinephrine, norephinephrine - > stimulate


Glucocorticoids -> stimulate

SREBP-1c


How does it regulate gluconeogenesis and glycolysis?

SREBP-1c - sterol regulatory element binding proteins


Synthesis is increased by insulin and decreased by glucagon


SREBP-1c will increase synthesis of


- Hexokinase IV, pyruvate kinase, lipoprotein lipase, acetyl-CoA carboxylase, fatty acid synthase complex




SREBP-1C will decrease


- Synthesis of PEP carboxykinase, FBPase-1, glucose-6-phosphatase

How does CREB regulate gluconeogenesis and glycolysis?

cAMP response element binding protein


Synthesis increased by glucagon via cAMP


Increases synthesis of PEP carboxykinase and glucose-6-phosphate

How does FOXO1 regulate gluconeogenesis and glycolysis?

Insulin will have the effect of turning off expression of the genes


- Insulin will activate a signalling cascade, which leads to activation of PKB. - PKB will phosphorylate FOXO1 in cytosol


- FOXO1 will be attached t ubiquitin, which will mark it for destruction via proteasome.


- FOXO1 which remains dephorphorylated can enter the nucleus and attach to DNA.


- To trigger transcription of it's associated genes




Increases synthesis of gluconeogenetic enzyme


Supresses synthesis of glycolytic enzymes, pentose phosphate pathway enzymes and triacylglycerol synthetic enzymes

How does chREBP regulate gluconeogenesis and glycolysis?

ChREBP is found inactive in the cytosol


- 2xPhosphorylated, cant move into nucleus


- PP2A dephosphorylates it, allowing it to enter nucleus


- In nucleus second dephosphorylation by PP2A


- This allows it to be associated with mix


- ChREBP-Mix complex will bind to a carbohydrate response element, and stimulate transcription




PP2A is allosterically activated by xylulose-5-phosphate


- xylulose-5-phosphate is a signal that other pathways which utilize glucose, has enough substrate




ChREBP will therefore turn synthesis of several enzymes


- Pyruvate kinase, fatty acid synthase and acetyl-CoA carboxylase

What is the function of NADPH created by the pentose phosphate pathway?

needed for synthesis


- Fatty acid biosynthesis


- Cholesterol biosynthesis


- Neurotransmitter biosynthesis


- Nucleotide biosynthesis




Detoxification


- Reduction of oxidized glutathione


- Cytochrome P450 monoxygenases




Also needed to counter the damaging effects of oxygen radicals

Wha it the fat of pentose from pp pathway?

needed in rapidly dividing cells


- Bone marrow, skin, intestinal mucosa, tumors




Uses ribose-5-phosphate


- RNA, DNA, ATP, NADH, FADH2, CoA

What enzymes is the oxidative phase made up of, and how are they regulated?

Glucose-6-phosphate dehydrogenase


Lactonase


6-phosphogluconate dehydrogenase


Phosphopentose isomerase

When is the non-oxidative phase active?

In tissues which primarily require NADPH, pentose phosphates will be recycled back into glucose-6-p




Transketolase and transaldolase

What are 4 fates of glucose-6-phosphate?

1. Ribose-5-phosphate is needed


- Fructose-6-p into ribose-5-p


- Glyceraldehyde-3-p into ribose-5-p




2. Ribose-5-phosphate and NADPH is needed


- Glucose-6-p -> Ribulose-5-p+ NADPH -> Ribose-5 p




3. NADPH needed


- Ribulose-5-p produced, but recycled




4. NADPH and ATP needed


- Ribulose + NADPH is first made, then ribose is sent into glycolysis via fructose-6-p and glyceraldehyde-3-p

How are reactive oxygen created in mitochondria?

Created when the rate of electrons entering the respiratory chain, and electrons transferred through are mismatched.


1. Superoxide radical production will increase at complex I and III


2. Partially reduced ubiquinone radical will donate electron to O2


3. Superoxide will act as a aconitase to release F2+


4. Fenton reaction will form reactive hydroxyl free radical -> OH



How does NADPH and glutathione protect against ROS?

Reduced glutathione or GSH will donate it's electrons to H2O2, reducing it to H2O and O2.


GS-SG will be regenerate to oxidized form by NADPH




For this we need glucose-6-phosphate dehydrogenase as it produces NADPH+H


In mitochondria nicotinamid nucleotide transhydrogenase is used instead, which transfers electrons from NADH to NADP



What are some deficiencies which prevent glutathione and NADPH from protecting against ROS?

Glucose-6-phosphate dehydrogenase deficiency


- Causes Heinz-bodies in red blood cells




Wernicke-Korsakoff syndrome


- Mutation in transketolase gene, which decreases affinity to TPP

What is muscle glycogen used for, and what is liver glycogen used for?

Muscle glycogen is used to generate ATP for muscle contraction


Liver glycogen is used to maintain glucose blood levels

What is the structure of glycogen?

Chain structure, consisting of multiple chains with branches




a-1,6 linkage


- Connects branches to main chain




a-1,4 linkage


- Connects the glucose molecules together




On the end of the main chain, and on all branches we find reducing ends

Glycogenin and how is a primer formed?

The primer for glycogen synthase


Tyrosine residue of the protein glycogenin, will form the first glycosidic bond between glucose and UDP-glucose




1. Primer consist of a glycogen molecule of 8 glucoses, remaining from previous degradation


2. Glycogen molecule attaches to glycogenin through tyr


3. One ATP is needed to start glycogen synthesis

How is a glucose molecule added during glycogen synthesis?

1. Glucose enters liver or muscle cell -> Becomes phosphorylated by hexokinase II or IV


2. Phosphoglucomutase will convert G6P to G1P


3. G1P will react with UTP to form UDP-glucose, catalyzed by UDP-glucose phosphorylase


4. Glycogen synthase transfers UDP-glucose to non-reducing end of a glycogen primer


5. The UDP is released and reconverted to UTP via ATP

How are branches formed in glycogen?

Formation of branches starts when a chain becomes 11 or more glucose residues.


At this point an oligomere of 6-8 glucose molecules is removed from the non-reducing end of the chain


This is reattached to the chain via a-1.6 linkage by glycogen branching enzyme


This process adds another non-reducing end to the glycogen chain

What are the steps of glycogen degradation or glycogenolysis?

1. Glycogen phosphorylase removes glucose residues from non-reducing ends


- Release 1 glucose-1-phosphate at the time


- Only remove until there are 4 glucose units until branching point


2. Debranching enzyme


- Removes 3 out of 4 glucose left on the branch


- Attaches them to non-reducing end of another branch


- Debranching has a transfer like activity


3. Remaining glucose removed by a-1,6 glucosidase


- releases free glucose


4. Glucose-1-phosphate -> glucose-6-phosphate by phosphoglucomutase


- In liver G6P are converted to glucose by glucose-6-phosphatase and released into blood stream via GLUT2




Glucose-6-phosphatase is an integral membrane protein of ER


- Prevents it from cleaving all G6P in the cell

How is glycogen phosphorylase regulated?



Glycogen phosphorylas has 2 forms

1. Glycogen phosphorylase a


- More active


- Activated by Phosphorylase b kinase by phosphorylation


- Epinephrine, Ca2+, AMP will activate the kinase in muscle,


- Glucagon will activate the kinase in liver


- Will activate will activate via cAMP -> PKA -> phosphorylase kinase b




2. Glycogen phosphorylase b


- Less active


- Not phosphorylated


- When muscle returns to rest, phosphorylase a phosphatase or PP1 removes phosphate groups, converting it




Glycogen phosphorylase in liver has glucose sensor


- Glucose will bind to an allosteric site of phosphorylase a, and this causes a change in conformation which exposes it's phosphorylated groups


- The PP1 will convert it to phosphorylase b,


- Reducing the activity, and slowing glycogen breakdown


- Also stimulated by insulin indirectly

How is glycogen synthase regulated?

Glycogen synthase has two forms, b and a


1. Glycogen synthase a


- Active form


- Converted by Phosphoprotein phosphatase 1


2. Glycogen synthase b


- Inactive form


- Converted by glycogen synthase kinase 3




GSK-3 phosphorylates glycogen synthase b -> inhibiting glycogen synthesis


- Inhibited by insulin


- GSK-3 requires prior phosphorylation or priming from casein kinase II


- Insulin will trigger activation of glycogen synthase b by blocking the activity of GSK3, and activating PP1 in muscles.


- insulin will phosphorylate GSK3 via PKB/PKA




PP1 -> dephosphorylates glycogen synthase b -> glycogen synthase a


- Promotes synthesis


- Removes phosphoryl groups from all 3 enzymes

How is GSK-3 regulated?

GSK-3 phosphorylates glycogen synthase b -> inhibiting glycogen synthesis


- Inhibited by insulin


- GSK-3 requires prior phosphorylation or priming from casein kinase II- Insulin will trigger activation of glycogen synthase b by blocking the activity of GSK3, and activating PP1 in muscles.


- insulin will phosphorylate GSK3 via PKB/PKA

What enzyme can remove phosphoryl groups from all three of the enzymes phosphorylated in response to glucagon in liver and epinephrine in muscle/liver? How is it regulated?

PP1 can remove phosphate groups from phosphorylase kinase, glycogen phosphorylase and glycogen synthase


PP1 is not found in free form, but tightly bound to its traget protein by glycogen-targeting proteins.


- Bind glycogen, each of the three enzymes


Regulated


- Positive allosteric regulation by glucose-6-phosphate


- Negative covalent phosphorylation by PKA

How is glycogen-argeting protein Gm regulated?

Gm binds glycogen to enzymes, like PP1


Gm can be phosphorylated at 2 different site, which give a different effect


- in response to epinephrine and insulin




1. insulin stimulated phosphorylation


- Phosphorylation at site 1


- Activates PP1, which dephosphorylates phosphorylase kinase, glycogen phosphorylase and glycogen synthase


- Insulin inhibit glycogen breakdown, and stimulate glycogen synthesis




2. Epinephrine stimulated phosphorylation


- Phosphorylation at site 2


- Dissociation from PP1, inactivating it


- Preventing access of PP1 to glycogen phosphorylase and synthase


- PP1 is also connected to phosphorylated inhibitor 1, inactivating it


- Epinephrine or glucagon will stimulate glycogen breakdown, and inhibit glycogen synthesis


-

In which region is glycogen synthase kinase 3 phosphorylated, and by which kinase?

Pseudosubstrate region, PKB



What is the hormonal regulation of glycogen synthesis and degradation?

Insulin


- Phosphorylation of site 1 on Gm -> PP1 activated -> synthesis of glycogen




Epinephrine


- Stimulates PKA -> Phosphorylates site 2 on Gm -> PP1 inhibited -> degradation of glycogen

What are the possible pathways in enzyme regulation and degradation?


- 10

1. Extracellular signal


- Hormonal


- neural


- Growth factos


2. Transcription of specific gene(s)


3. mRNA degradation


4. mRNA translation on ribosome


5. Protein degradation


- Tagged by ubiquitin for degradation by proeasomes


6. Enzyme sequestered in subcellular organelle


- Segregation of enzyme


7. Enzyme binding substrate


8. Enzyme binds ligand (allosteric)


9. Enzyme undergoes (de)phosphorylation


10. Enzymes combines with regulatory protein



Where does fatty acid synthesis, elongation and desaturation happen?

Fatty acid synthesis happens in cytosol

- until C16


Fatty acid elongation happens in ER and mitochondria


Fatty acid desaturation in ER

What is the function of acetyl-CoA carboxylase in fatty acid synthesis?

Synthesis of malonyl- CoA from acetyl-CoA


Acetyl-CoA har 3 regions


1. Biotin carrier protein


2. Biotin carboxylase


3. Biotin transcarboxylase




This is the first step of fatty acid synthesis, as biotin carboxylase transfers HCO3 to biotin carrier protein with the help of ATP-> ADP + Pi.


Then transcarboxylase transfers activated CO2 biotin to acetyl-CoA

what are the enzymes of fatty acid synthase?

8 enzymes in a complex

4 basic enzymes


- Ketoacyl synthase


- Ketoacyl reductase


- Hydroxyacyl dehydratase


- Enoyl reductase




4 other


- Acetyl carrier protein - ACP


- Acetyl-CoA-ACP acyltransferase - AT


- Malonyl-CoA-ACP acyltransferase - MT


- Palmitate deacylase - PD





What are the steps of fatty acid synthesis, which enzymes, cofactors and products do we find?

Before fatty acid synthesis we must activate Acetyl-CoA and malonyl-CoA




Step 1 - Condensation


- Ketoacyl synthase


- Condensates the substrates, creating acetoacetyl-ACP


- CO2 is released




Step 2 - Reduction


- Ketoacyl reductase reduces NADPH + H -> NADP+


- D-B-hydroxybutyryl-ACP is formed




Step 3 - Dehydration


- Hydroxyoacyl dehydratase removes water from the substrate


- Water is removed




Step 4 - Reduction


- Enoyl reductase reduces NADPH+H -> NADP+


- Double bond is saturated


-Saturated acyl group is formed, lengthened by two carbons





What is the source of cytosolic acetyl-CoA?

Almost all cytosolic acetyl-CoA used in fatty acid synthesis is formed in the mitochondria from pyruvate oxidation and from the catabolism of the carbon skeletons of amino acids


Inner mitochondrial membrane is impermeable to acetyl-CoA


- Because of this it´s converted to citrate, before shuttled into the cytosol via a citrate transporter


- in cytosol citrate is reconverted to acetyl-CoA and oxaloacetate


- Acetyl-CoA enters fatty acid synthesis


- Oxaloacetate is reduced to malate by malate dehydrogenase, and shuttled via Malate-a-ketoglutarate transporter




Cytosolic NADPH is also created as malate is converted to pyruvate via malic enzyme





What is the sources of cytosolic NADPH?

The malate formed from oxaloacetate during the shuttle of acetyl-CoA to cytosol can also be used for production of NADPH

Malate to pyruvate via malic enzyme will produce NADPH




NADPH is also produced in the pentose phosphate pathway


- Glucose-6-phoshphate dehydrogenase


- 6-phosphogluconate dehydrogenase


How is acetyl-CoA carboxylase (ACC) enzyme I regulated?

Found in all cells, but mostly adipose and mammary glands


Inactivated by AMP-activated protein kinase


- Glucagon, adrenaline, AMP, palmitoyl-CoA -> Phosphorylation of ACC


- Glucagon inhibits protein phosphatase A2


Activated by insulin and citrate


- Insulin -> insulin-dependent protein phosphatase dephosphorylates ACC, activating it - Protein phosphatase 2A




Inactive carboxylase + citrate -> partialy active carboxylase


- Inhibited by palmitoyl-CoA

What is mechanism of fatty acid elongation in animal cells?

Takes place in ER and mitochondria


Palmitate is the precursor for most longer fatty acids


Similar mechanism as fatty acid synthesis, but with other enzymes, and using CoA instead of ACP


Fatty acids are elongated on the head




Elongation starts with donation of carbons by malonyl-CoA


Then reduction, dehydration and reduction

What is the mechanism of desaturation in vertebrates?

Happens in smooth ER


Fatty acid desaturation is needed for cell membrane fluidity in different temperatures


Double bond introduced via an oxidative reaction by fatty acyl-CoA desaturase




During this process 2 different substrates will at the same time undergo electron oxidations.


- Electrons will flow via cytochrome and cytochrome reductase


1. NADPH will give its electrons to cytochrome reductase - FAD -> FADH2


2. Cytochrome reductase (FADH2) will give it's electrons to cytochrome - Fe3+ -> Fe 2+


3. Cytochrome (Fe2+) will give them on to the fatty acid



At which carbons of a fatty acid can animals add double bands, and why is this important?

Animals can only put double bonds before 10th carbon atom


Because of this, it's essential for animals to have fatty acids with double bonds after 10C in their diet


These can be made into arachidonate, which can turn into prostaglandins, thromboxanes and leukotrienes

What can be produced from arachidonate, and how is the synthesis regulated?

Arachidonate is produced from phospholipids

And can produce prostaglands, thromboxanes and leukotrienes


- By enzymes in smooth ER


- COX-1 -> synthesis prostaglandins that regualte secretion of gastric mucin


- COX-2 -> synthesis prostaglands that mediate inflammation, pain, fever


- Thromboxanes are produced on the pathway involving COX-1




Steroids will inhibit synthesis of arachidonate


- No prostaglandins, thromboxanes or leukotrienes


Non-steroids like aspirin and ibuprofen will inhibit cyclooxygenase


- No prostaglandins or thromboxanes

What are essential fatty acids for mammals?

Linoleate and alpha-lineoleanate

How can glycerol-3-phosphate be synthesized in liver and adipose tissue?

In liver


- Glycerol kinase


- Glycerol-3-phosphate dehydrogenase




In adipocytes


- Glycerol-3-phosphate dehydrogenase

Phosphatidic acid

Diacylglycerol-3-phosphate

What are the different head groups for phosphatidic acid? what else can phosphatic acid produce?

Head groups can be ethanolamine, serine, inostiol




Triacylglycerol can be produced from phosphatidic acid


- Phosphatidic acid phosphatase


- Acyl transferase


How is lipid synthesis regulated?

Insulin


- Favors synthesis of both acetyl-CoA and fatty acids -> triacylglycerol synthesis

Diabetes mellitus


- How does this effect fatty acid synthesis

Diabetes mellitus is the lack of insulin


This causes a decrease in fatty acid synthesis and causes ketone body production

Synthesis of triacylglyceride

1. Production of glycerol-phosphate


- Either via glycerol-3-phosphate(liver and fat) or glycerol kinase (liver)


2. Production of phosphatidic acid


- Acyl transferase twice


3. Production triacylglycerols and glycerophospholipids


- Phosphatidic acid phosphatase + acyl transferase -> triacylglycerol


- Attachment of head group -> glycerophospholipids

What is glycerneogenesis, whats the steps?

A shortened version of gluconeogenesis


1. Pyruvate


- Pyruvate carboxylase


2. Oxaloacetate


- PEP carboxykinase


3. Phosphoenolpyruvate


- Multistep


4. Dihydroxyacetone phosphate


- Glycerol-3-phosphate dehydrogenase


5. Glycerol-3-phosphate


6. Triacylglycerol synthesis

What´s the role of glycerneogenesis?

-Adipose tissue it's coupled with reesterification of free fatty acids to control the rate of fatty acid release in blood


- In fasting humans, it supports the synthesis of glycerol-3-p enough to account for 65% of fatty acid reesterification to triacylglycerol

Regulation of glyceroneneogenesis?

Glucocorticoid hormone


- Stimulates glycerneogenesis in liver


- Inhibits glycerneogenesis in adipocytes




Thialozolidine


- Increases glyceroneogenesis -> increasing resynthesis of triacylglycerol in adipose tissue -> Reduces amount of free fatty acid in blood


- Treats type II diabetes

Triacylglycerol cycle

Triacylglycercol cycle shows how triacylglycerols are broken down and synthesized during starvation


In the liver triacylycerols are produced from glycerol-3-phosphate and transported to blood and adipose tissue


In Blood lipoprotein lipase releases glycerol, and fatty acids are used for fuel for tissues


In adipose tissue fatty acids are resynthesizes into triacylglycerols, and glycerol is released

What are the basic type of complex lipids?

1. Phospholipids


Glycerophospholipids


- 2 fatty acids + alcohol


Sphingolipids


- Sphingosine + fatty acid + choline


2. Glycolipids


Sphingolipids


- Sphingosine + fatty acid + mono/oligosaccharide


Galactolipids


- 2xfatty acids + mono +SO4


3. Aracheabacetial ether lipids



What are the two strategies for synthesizing complex lipids?

Strategy 1


Diacylglycerol activated with CDP and then add head group


- Remove CMP


Strategy 2


Diacylglycerol + head group activated with CDP.


- Remove CMP

What are the 4 steps of assembling a phospholipid?

1. synthesis of backbone of molecule


- Glycerol or sphingosine


2. Attachment of fatty acid


3. Addition of hydrophilic head group


4. Alteration or exchange og head group


- This is where the two strategies seperate

Which lipids are precursors of CDP-Diacylglycerol?

Phosphatidylglycerol


- CDP-diacylglycerol + glycerol




Cardiolipin


- CDP-Diacylglycerol + phosohatidylglycerol




Phosphatidylinositol


- CDP-diacylglycerol + inostiol

How do inositol phosphates function as intracellular signaling molecules

Splitting by phospholipase C


-Diacylglycerol -> DAG


- Inositol-1,4,5-triphosphate -> IP3




Splitting by phophatidyl-inositol 3 kinase


- Phosphatidyl-inositol 3,4,5 triphosphate or PIP3

What are the roles of IP3 and DAG?

IP3 and DAg both contribute to activation of protein kinase C, by raising cytosolic Ca2+ concentration and IP3 also activates Ca2+ dependent enzymes.


1. Hormone binding


- Binds to specific receptor


2. GDP-GTP exchange on G protein


3. Activation of PLC by GTP-G


4. Cleavage of PIP2 to IP3 and DAG


5. Binding of IP3 to ER IP3 -receptor evoking Ca release


6. DAG and Ca2+ activate PKC


7. PKC phosphorylates target proteins triggering an hormone response

What is the intracellular effects of insulin mediated by?

PIP3

What is HMG-CoA?

a common precursor for both ketone bodies and cholesterol


Formation of ketonebodies occur in the mitochondria, while the synthesis of cholesterol happens in the cytosol

HMG-CoA is a branchpoint for which two products? and in which compartments and by which enzymes is this done?

HMG-CoA functions as a branchpoint for synthesis of cholesterol in cytosol and ketone bodies in cholesterol




Synthesis of cholesterol


HMG-CoA -> Mevalonate via HMG-CoA reductase


- 2 NADPH -> NADP+, CoA removed




Synthesis of ketone bodies


HMG-CoA -> Acetyl-CoA + acetoacetate via HMG-CoA lyase



Conversion of ketone bodies?

3 ketone bodies -> Acetoacetate, acetone, D-B-hydroxybutyrate




Acetaoacetate


- Created from HMG-CoA lyase,


- Also created as when fumarylacetoacetate is split by fumarylacetoacetase




Acetone


- Acetoacetate decarboxylase, removes CO2 from acetoacetate




D-B-hydroxybutyrate


- D-B-hydroxybutyrate dehydrogenase from acetoacetate


- Acetoacetate is reduced to form



How are acetoacetate activated for metabolism?



Done by B-ketoacyl-Coa transferase

Succinate-CoA -> Succinate




Acetoacetate -> Acetoacetyl-CoA

What are the steps of cholesterol synthesis?


What is the rate determining step?

1. Synthesis of mevalonate from HMG-CoA


- Rate determining step


2. Mevalonate is converted into chain of activated isoprenes


- ATP is utilized to attach phosphate groups activating isoprenes


- Geranyl pyrophosphate -> 10 Carbons


3. After several steps squalene is formed -> 30 carbons


- Precursor is farnesyl -pp -> 15C


4. cholesterol is formed from squalene after a sequence of reactions


- Cholesterol is 27 C


- One of the enzymes is cyclase

Esterification of cholesterol

cholesterol esters are formed in the liver by ACAT


- Cholesterol + fatty acyl-CoA -> Cholesteryl ester + CoA


ACAT catalyzes the transfer of fatty acids from CoA to the hydroxyl group of cholesterol


- creates a hydrophobic form




Cholesterol esters can also be formed form LCAT


- LCAT catalyzes the formation of cholesteryl ester from lecthine


- Cholesterol + Lecithine -> Cholesteryl ester + Lysolecithine


- Lecithine -> phosphatidylcholine



What are the types of lipoproteins? what do they contain and what is their destination?

1. Chylomicrons


- Dietary lipids


- From GI to liver


2. VLDL


- Endogenous lipids, cholesterol


- From liver to blood


3. LDL


- cholesterol


- From blood to tissue


4. HDL


- Excess Cholesterol


- From tissue to liver

How is cholesterol synthesis regulated in response to cholesterol levels?

System which does transcriptional regulation of the gene encoding HMG-CoA reductase


The gene coding for HMG-CoA reductase, and 20 others gene affiliated with cholesterol are controlled by sterol regulatory element binding proteins or SREBPs.


Only the soluble amino terminal domain of SREBP can function as a transcriptional activator.


- This domain has no access to nucleus when it's a part of SREBP molecule


To activate the transcription of HMG-CoA reductase, the transcriptional active domain must be proteolytically cleaved by SCAP




When cholesterol levels are high, SREBP is inactive, secured to ER via SREBP cleavage-activating protein


- SCAP binds to cholesterol, acting as a sterol sesnor


- When cholesterol levels decrease, conformational changes causes release of SCAP-SREBP complex from ER




In golgi SREBP is cleaved twice, and the second cleavage will release the amino-terminal domain into the cytosol


- Travels to nucleus and activates target genes


- Short life, rapidly degraded by proteasomes



How is HMG-CoA reductase regulated by hormones and by

Insulin activates HMG-CoA reductase


- Dephosphorylates the enzyme


Glucagon inhibits HMG-CoA reductase


- Phosphorylates the enzyme




Phosphorylated by AMP-activated protein kinase




Unidentified metabolites of cholesterol stimulate proteolysis of HMG-CoA reductase

What is the effect of cholesterol entering the cell on cholesterol synthesis?

1. Down regulation of receptor mediated endocytosis


2. Inhibition of cholesterol synthesis


3. Promotion of cholesteryl esters


4. Inhibition of endocytotic receptor synthesis

What's the theory of glucose fatty acid cycle?

If plasma glucose is decreased, fatty acids are mobilized (fasting, glucagon) and increased levels of fatty acid will decrease glucose utilization. This restores the glucose level, and maintains homeostasis

AMP dependent protein kinase - AMPK

ADP produced in synthetic reactions is converted to AMP by adenylate kinase


AMP will activate AMPK, which regulates the anabolic and catabolic pathways by phosphorylating the enzymes




AMPK will inhibit anabolic processes


- Process using ATP -> ADP




AMPK will stimulate catabolic process


- ADP -> ATP




Stress, fasting and exercise activates AMPK



What reactions does AMPK stimulate?


AMPK stimulates catabolic processes


- Stimulates glucose transport (GLUT1,4)


- Stimulates PFK-2 -> Cardiac glycolysis


- Stimulates fatty acid oxidation and glucose uptake in skeletal muscle


- Fatty acid oxidation, glucose uptake and glycolysis in heart

What reactions are blocked by AMPK?

HMG-CoA reductase


Transcription


Acetyl-CoA carboxylase - FA synthesis


Synthesis of Cholesterol, triacylglycerol, glycogen, protein

Regulation of ketone body synthesis via HMG-CoA synthase

Liver contains two types of HMG-CoA synthase


- cytosolic for cholesterol synthesis


- Mitochondrial for ketone body synthesis




The mitochondrial is rate limiting for ketone body synthesis


The mitochondrial HMG-CoA synthase will undergo covalent modification via succinylation/desuccinylation of pre-exisiting enzyme

What is PPAR, what's their role?

PPAR - Peroxisome proliferator-activated receptors


- Nuclear receptors


- Transcription factor, regulating expression of genes


- Effect on lipid transport




PPAR has roles in


- alpa (liver, heart, muscle, kidney) -> Regulates FA metabolism, HDL-cholesterol


- beta (anywhere) -> Kartinocytes differentiation, wound healing, mediated VLDL signaling


- gamma (adipocytes, macrophages) -> Adipocyte differentiation, lipid storage, glucose homeostasis

Farber disease

Enzyme deficiency which results in accumulation in lysosomes of PAS-positive lipids consisting of ceramide

What is the fate of the amino group and carbon skeleton of an amino acid?

Amino group


- Biosynthesis of amino acids, nucleotides, and biological amines


- Carbamoyl phosphate -> Urea cycle -> Urea




Carbon skeletons


- A-keto acids -> Citric acid cycle



How are amino groups incorporated into biomolecules?

They are incorporated via glutamate and glutamine


- Glutamate is the source of amino groups for most other amino acids, through transmination reactions


- Glutamine is source of amino groups for biosynthetic process

What are the 3 enzymes of the ATP-dependent pathway of protein degradation?

E1: Ubiquitin-activating enzyme


E2: Ubiquitin-conjugating enzyme


E3: Ubiquitin-protein ligase

What are the two pathways to protein degradation?

1. ATP-dependet pathway used for degradation of defective proteins, and those with short half-lives


- Involves ubiquitin, which becomes covalently linked to proeins slated for destruction via an ATP-dependent pathway


-Involves E1, E2, E3


- Ubiquinated proteins are degraded by proteasome




2. A second system is found in lysosomes. This recycles the amino acids of membrane proteins, extracellular proteins and those with longer half-lives

Proteasomes

Large complex which degrades ubiquitinated proteins


Complex has two main types of subcomplexes


- Regulatory Caps on either side


- Barrel like core




Core has 4 rings


- Outer rings are formed by seven alpha subunits


- Inner rings are formed by seven beta subunits

What is the role of PLP, what is it's derivative

A derivative of vitamin B6


PLP is an intermediate amino group carrier


- accepts and donates aminogroups




This reactions goes from left to right, then from left to right


- Ping-pong reaction mechanism




PLP + amino acid -> Shiff base intermediate -> Quinonoid intermediate -> Pyroxidaxmine phosphate + keto acid

What are some PLP requiring enzymes?

Aminotransferase, tryptophan synthase, cysthionine synthase and lyase, glutamate decarboxylase, serine dehydratase, ornithine decarboxylase etc

Glutamate hydrogenase reaction


- Cofactor, where, regulation

Happens in hepatocyte mitochondrial matrix


Works with both NAD+ and NADP+


Allosterically regulated


- ADP(GDP) stimulates


- GTP (ATP) inhibits




Oxidative deamination



What is oxidative deamination?

Release of NH4+ from glutamate, forming a-ketoglutarate


- Happens in glutamate dehydrogenase

Transdeamination pathway

Transamination + oxidative deamination -> transdeamination

Transamination + oxidative deamination -> transdeamination



How does NH4+ arrive to the liver?

1. Amino acids from ingested proteins


2. Alanine from muscle


- Glucose-alanine cycle via pyruvate and glutamate


3. Glutamine


- From muscle and other tissues

What are the sources of ammonia for urea cycle?

Amino acid degradation


Ammonia secretion in kidney tubules from glutamine


Degradation of pyrimidine bases


Produced by intestinal bacteria

What are the ATP dependent reactions of the urea cycle, and where do they happens?

Carbamoyl phosphate synthase I


- Requires 2ATP


- NH3 reacts with bicarbonate to form carbamoyl phosphate which enters cycle


- Reaction takes place in the mitochondria




Argininosuccinate synthetase


- Requires ATP -> AMP


- Happens in cytosol


- Aspartate enter the urea cycle from the citric acid cycle

aspartate-argininosuccinate shunt

Argininosuccinate will form arginine and fumarate via argininosuccinase or lyase


This fumarate can be converted to malate and enter the citric acid cycle


This connection is the aspartate-argininosuccinate shunt of TCA

Malate-aspartate shuttle


What are the steps?

shuttle of NADH in the liver, kindey and heart mitochondria


1. NADH+H is transferred to cytosolic oxaloacetate -> Malate


2. Malate crosses the inner membrane via malate-a-ketoglutarate transporter


- Ketoglutarate is transported in the opposite direction


3. Malate dehydrogenase gives oxaloacetate and NADH+H


4. Oxaloacetate is first transaminated to aspartate


5. Aspartate is transported out via glutamate-aspartate transporter


- Glutamate is transported in the opposite direction


6. Oxaloacetate is regenerated in cytosol

How is the overall energetic cost of urea synthesis reduced?

It used 3 ATP -> 2 ADP+AMP + 4Pi




1 NADH is formed in malate dehydrogenase reaction


- 1 NADH -> 2.5 ATP

How is carbamoyl phosphate synthase I regulated?

Arginine will stimulate N-acetylglutamate synthase, and N-acetylglutamate will stimulate carbamoyl phosphate synthetase I

What are the major goups of amino acids according to their endproducts?

Ketogenic


- Can yield ketone bodies in liver




Glucogeneic


- Can be converted to glucose


Mixed


- Both ketogenic and glucogenic

What are the purely ketogenic amino acids?

Leucine and lysine




-> Acetyl-Coa -> ketone bodies or citrate

What are the glucogenic amino acids?

Aspargine


Aspartate


Methionine


Valine


Arginin


Glutamine


Glutmate


Hisitidine


Prolin




They can become either pyruvate or oxaloacetate -> glucose

What are the mixed amino acids?

Tryptophan


Phenylalanine


Tyrosine


Threonine


Isoleucine


Alanine


Cysteine


Glycine


Serine

What are the Coenzyme B12 dependent reactions?


- What does deficiency cause?

Methylalonyl-Coa mutase


Rearrangement of L-methylmalonyl-CoA to succinyl-CoA




Methionine synthase reaction


Metabolic folates trapped in N5-methyl form

What are cofactors which transfer one-carbon groups? And in which oxidation states?

Biotin


- Most oxidized


- COO-




Tetrahydrofolate -> THF


- Intermediate oxidized state


- Methylene, methenyl, formyl, formimino groups




S-adenosylmethionine


- Most reduced


- Methyl

How is glutamine synthase regulated?

Allosterically regulated


- Each inhibitor will produce a partial inhibition, but all will together shut down the enzyme




Glycine, alanine, Glucosamine-6-p, Histidine, CTP, carbamoyl phosphate, tryptophan and AMP

What is the function of S-adenosylmethionine?

AdoMet is used for methyl group transfers


Synthesized from ATP and methionine by methionine adenosyl transferase




Often uses methylene-THF as a methyl donor

Reduction of N2 gas to ammonia



N2 + 3 H2 -> 2 NH3

Triple bond between N


Activation energy of N-fixation is very high



Nitrogen fixation

Done by nitrogenase enzyme complex


Hydrolysis of 16 ATP


N2 + 10H+ +8e- + 16ATP -> 2 NH4+ + 16ADP + 16Pi + H2

What regulates the amino acid biosynthesis?

Allosteric regulation


The enzyme regulated is often the first in the sequence, and it's inhibited by the end product of the pathway


- negative feedback


Regulation of the various synthetic pathways is coordinated

What causes hyperammonemia?

Urea cycle defects normally lead to hyperammonemia or build up of intermediates




Permanent activation of glutamate dehydrogenase also causes hyperammonemia

Congenital hyperammonemia type I

Carbamoyl phosphate synthetase

Congenital hyperammonemia type II

Ornithine transcarbamoylase

Citrullinemia

Argininosuccinate synthetase

Argininosuccinate aciduria?

Argininosuccinase

Argininemia

Arginase

N-acetylglutmate synthase deficiency

N-acetylglutamate synthase

Maple syrup urine disease

Defect on branched-chain a-keto acid dehydrogenase complex

Hyperglycinemia

Defect of the glycine cleavage enzyme

Methylmalonic acidura

Defect on methylmalonyl-Coa mutase

Homocystinuria I

Defect of cystathionine B-synthase



Phenylketonuria

Defect on phenylalanine hydroxylase

Tyrosinemia Type II

Defect on tyrosine aminotransferase

Tyrosinemia type III

Defect of p-hydroxyphenylpyruvatedioxygenase



Alkaptonuria

Defect of homogentisate dioxygenase

Tyrosinemia type I

Defect of fumarylacetoacetase

Albinism

Defect of tyrosinase


Melanine synthesis from tyrosine




DOPA is a precursor of melanine




Similar reaction to tyrosine hydroxylase

Where does biosynthesis of heme happen, and how is it regulated?

Happens in both the mitochondria and in the cytosol, and is regulated by heme as a feedback inhibitor

What does the degradation of heme give?

Heme degradation protects cell from oxidative damage


1. Release of iron


- Fe2+ binds to ferritin


- CO binds to hemoglobin


- CO produced is toxic (high conc.), vasodilator (low conc.) with regulatory effects




2. Formation of bilirubin, via biliverdin


- Bilirubin binds to serum albumin


- Important antioxidant


- Protective effect in developing brain



What is phosphocreatine used for in muscle? When is it produced?

Formed by creatine kinase from creatine


It's an important energy reservoir in skeletal muscle


- Allows for rapid regenerating ATP from ADP, by creatine kinase reaction


Active contraction and glycolysis -> ATP synthesis


Light activity/rest -> Phosphocreatine synthesis

What is glutathione GSH formed from? What is it used for?

From Glutamate, cysteine and glycine


Used 2 ATP


Forms reduced form




GSH - redox buffer


- Maintains -SH group of protein in reduced form


- Maintains the iron of heme in ferrous state


- Serves as reducing agent for glutaredoxin - deoxyribonucleotide synthesis


- Removes toxic peroxides formed during growth or aerobic metabolism during glutathione peroxidase reaction



What neurotransmitters are synthesized from tyrosine? Defects?

Dopa, Dopamine, norepinephrine,




Schizoprenia -> overproduction


Parkinsons ->underproduction

What neurotransmitters are synthesized from glutamate? defects?

GABA


Inhibitory neurotransmitter


Underproduction can lead to epileptic seizures

What neurotransmitters are synthesized from histidine?

Histamine


Vasodilator


Released in allergic response

What neurotransmitters are synthesized from tryptophan?

Serotonin

What neurotransmitters are synthesized from methionine?

Spermine, spermidine


Involved in DNA packing

What neurotransmitters are synthesized from arganine?

Nitric oxide




is a free radical


Important biological messenger

What are the roles of nucelotides in cells?

1. DNA and RNA precursors


2. Carrier of chemical energy


- ATP and GTP


3. Co-factor components


- NAD, FAD, coenzyme A, S-adenosylmethionine


4. Activated intermediates


- UDP-Glucose, CDP-diacylglycerol


5. Cellular second messenger


- cAMP, cGMP

What are the two ways of Nucleotide synthesis?

Salvage pathway


- Activated ribose (PRPP) + base -> nucleotide


- Recycles free bases and nucleotides released during nucleic acid breakdown




De novo pathway


Activated ribose (PRPP) + amino acids + ATP+ CO2 NH3-> Nucleotide

What are the common motives of purine and pyrimidine synthesis?

1. Uses 5- phosphoribosyl 1- pyrophosphate


2. Uses amino acids


- Purine -> glycine


- Pyrmidine -> aspartate


3. N donor


- Glutamine


- aspartate (purine)

Why can nucleotide synthesis limit rate of DNA replication and transcription?

Cellular pool of nucleotides i 1% or less of the amounts required to synthesize the cell's DNA

What is the origin of the different parts of purines?



How is purine nucleotide biosynthesis regulated?

It's regulated by feedback inhibition


It's regulated in 4 steps by endproducts




1. AMP, GMP and IMP will inhibit the first reaction


-Glutamine-PRPP amidotransferase




2. The endproducts will inhibit their own synthesis from IMP


- AMP will inhibit adenylosuccinate synthetase


- GMP will inhibit IMP dehydrogenase




3. Reciprocal regulation


- Balance between AMP and GMP




4. AMP from endproduct will be phosphorylated to ADP, and ADP will inhibit Ribose phosphate pyrophosphokinase (PRPP synthetase)

What are pyrmidine nucleotides made from?

Made from aspartate, PRPP, carbamoyl phosphate



Where does carbamoyl phosphate synthetase II happen?

Happens in the cytosol


Will channel

How is pyrimidine biosynthesis regulated?

Regulated by negative feedback of the endproduct


- Cytidine-5-triphosphate CTP inhibits transcarbamoylase allosterically




ATP is a postive modulator of the same enzyme


- Prevents change induced by CTP

Degradation of purines and pyrimidines gives?

Purine -> uric acid


Pyrimidine -> urea

Adenosine deaminase deficiency

Causes immunodeficiency


- T and B wont properly develop



Salvage pathway

Salvage pathway recycle purine and pyrmidine bases


- Adenine + PRPP -> AMP + PPi


- Hypoxanthine + PRPP -> IMP + PPi


- Guanine + PRPP -> GMP + PPi

Lesch-Nylan syndrome

Prevents salvage pathway of guanin


Causes


- High levels of uric acid


- Brain damage


- Purines overproduce



What are the 3 forms of DNA?


- Helical sense


- Glycosyl bond formation



A form


- Right handed


- Anti glycosyl bond formation


B form


- Right handed


- Anti glycosyl bond formation


Z form


- Left handed


- Anti for pyrimidines, syn for purines

How many base pairs are there between A and T versus G and C

AT -> Double


CG -> triple bond

What is a triple helix?

This a structure where the first half of one strands is separated and folds back on the other half of the repeat to form a triple helix with it's earlier strand

What are some well-characterized non-enzymatic reactions of nucleotides?

Deamination


- Removes a NH2 group for cytosol, 5-methylcytosine, adenine and guanine


- Creates uracil, thymine, hypoxanthine, xanithine




Depurination


- Add water to a guanosine residue -> Guanine and apurinic residue

What introduces a bend or kink into the DNA?

Formation of cyclobutane pyrimidine dimer

What are 3 chemical agents which cause DNA damage? what is their precursor?




What are some alkylating agents which cause damage to DNA?

Sodium nitrite


Sodium Nitrate


Nitrosamine


nitrous acid precursors




Alkylating agents


- S-adenosylmethionine


- Dimethylnitrosamine


- Dimethylsulfate


- nitrogen mustard

Nucleosomes


What are the different histones?




How is the nucleosome positioned?

Histone cores connected with linker DNA


H2A and H2B, H3, H4 are histone cores




H1/H5 is linker DNA




Its positioned in such a way to make optimal use of A=T base pairs where the histone core is in contact with the minor groove of the DNA helix

What is the compaction of DNA in eukaryotic chromosomes?

1. DNA


2. Beads on a string form of chromatin


3. 30 nm fiber


4. One loop


5. one rosette


- Created by nuclear scaffold


6. One coil


- 30 rosettes


7. Two chromatids


- 10 coils each

What are chromatin remodeling factors?

Chromatin remodeling factors are multiprotein complexes with some subunits having helicase activity.


These complexes will dissociate DNA from nucleosome, decondense chromatin, and make DNA more accessible to transcription




May result in transcriptional repression, by exposing the histone tails to deactylases, or by assisting in folding

What are some remodeling complexes, and what is their function?




What does chromatin remodeling facilitate?

SWI/SNF, ISWI, CHD, INO80


- Share similar ATPase domains, but has unique subunits




SWI/SNF are master regulators of gene expression, and they also can modulate alternatic splicing




They can cause eviction of histone core, or sliding of histone cores


- Opens up more DNA




chromatin remodeling facilitates Nucleotide excision repair


- Deletion of lesions


- Recruitment of NER enzymes


- Stimulates repair

What is mammalian homolog of the yeast SWi/SNF complex?

Brm/Brg-associated factor is a chromatin remodeling complex



ISWI family

a chromatin remodeling family


- ACF and CHRAC are reported to promote chromatin assembly -> suppressing transcription


- NURF or nucleosome remodeling factor is another complex in the same family -> it activates RNA polymerase II

CHD family

A chromatin remodeling family


Ejection of nucleosomes -> promoting transcription




Also has complexes with repressive roles

INO80 family

A chromatin remodeling family


transcriptional activation, DNA repair, telomere regulation, chromosome segregation and DNA replication




SWR1, a member, has a unique ability to restructure nucleosomes, by removing H2A-H2B dimers, and replace with H2A.Z-H2B dimers


- INO80 can reverse this process and stabilize chromatin


- These changes inhibit genome stability

what is DNA replication regulated by?

Genetic and epigenetic factors

What is the leading and lagging strand?


In what direction are nucleotides synthesized, and in what direction are they read?

Leading strand -> 3-5 strand


Lagging strand -> 5-3 strand


- Synthesized by polymerase III




Nucleotides are synthesized in a 5-3' direction, and read in a 3-5'


- Creates Okazaki fragments

What are the steps of exonuclease activity of the DNA polymerase I?

DNA polymerase I has 2 sites


- DNA polymerase active site


- 3-5 proofreading exonuclease active site




1. Mismatch is formed


2. Mispaired 3-OH end of the growing strand blocks further elongation


3. DNA polymerase slides back to position the mispaired base in the 3-5 exonuclease active site


4. Mispaired nucleotide is removed


5. DNA polymerase slides forward, and continues it's polymerization activity

what is the exonuclease activity in DNA Pol I, II, III?


- 3-5 exonuclease (proofreading)


- 5-3 exonuclease

DNA pol I


- Has both 3'-5' exonuclease and 5'-3' exonuclease acitivty


- Has two fragments, Large fragment (Klenow) does DNA polymerization and proofreading. The smaller fragment has the 5-3 exonuclease activity -> nick translation


- RNA primers will be removed by the smaller fragment of DNA pol I, and replaced with DNA


- Ligase closes the nick in the DNA strand with ATP or NAD




DNA pol II


- Only 3'-5' exonuclease




DNA pol III


- only 3'-5' exonuclease

Which DNA pol has they highest polymerization and processivity rate?

Polymerization rate


- Nucleotides/s


- DNA pol III has the highest




Processivity


- nucleotides added before polymerase dissociates


- DNA pol III -> above 500,000


- DNA pol II -> 1500


- DNA pol I has only 3-200

What are the ATP dependent stages of replication in E.Coli?

The initiation of replication requires 3 ATP

What are the proteins in the E. Coli replication fork?

SSB


DnaB protein - Helicase


Primase


DNA pol III


DNA pol I


DNA ligase


DNA gyrase/topoisomerase II

What is the mechanism of DNA ligase?

1 and 2nd steps lead to the activation of the 5' phosphate in the nick


- An phosphate group is first transferred to DNA ligase, then to the 5' phosphate in the nick


3. 3'-hydroxygroup will attack the newly attached phosphate and displace AMP


4. Phosphodiesterbond seals the nick

When is it important to distinguish between parent and newly synthesized strands, and how?

Following the replication, the template strand is methylated, but the new strand is not


- Hemimethylated DNA


After a few minutes DAM methylase will methylated the two strands, making them alike




The cell will tag the template, to be able to distinguish the strands.

What are the early steps of the methyl-directed mismatch repair?

ATP-dependent mismatch repair




MutH and MutS proteins will recognize the sequence


- Form a complex


- DNA is threaded through this complex


- Scans until they meet a MutH protein bound to a hemimethylated sequence




MutH will cleave the unmethylated strand


- DNA licase II and several exonucleases will degrade the unmethylated DNA strand from that point towards the mismatch


- Which exonuclease which is used is dependent on the location of the cleavage site relative to the mismatch


- Nick is filled in by DNA polymerase III, and is sealed by DNA ligase

What are the types of DNA repair systems in E.Coli?

Mismatch repair


- DAM methylase


- MutL, MutL, MutS proteins


- DNA helicase II


- SSB


- DNA polymerase III


- Exonuclease I, VII, X


- Recj nuclease


- DNA ligase


Base excision repair


- Used for abnormal bases


- DNA glycosylases


- AP endonucleases


- DNA polymerase I


- DNA ligase


Nucleotide excision repair


- DNA lesions that cause large structural changes


- ABC excinuclease


- DNA pol I


- DNA ligase


Direct repair



What are dimer pyrimidines?

Can be caused when to adjacent thymines are exposed to UV lights


- Causes linkages between the adjacent thymines




Cyclobutane thymine dimer


- C6 - C6 and C5-C5 linkage to adjacent pyrimidine


- causes kinks in the DNA helix




6-4 photoproduct


- Linkage between C6 - C4 between adjacent thymine

Base-excision repair pathway

1. DNA glycosylase will recognize a damaged base, and cleave it between the base and the deoxyribose in the backbone


2. AP endonuclease will cleave the phosphodiester backbone near the AP site


3. DNA pol I will repair with 5-3 exonuclease activity, and replace a portion of the strand


4. DNA ligase will close the remaining nick

Nucleotide excision repair in E. Coli vs humans

Repairs DNA lesions which could cause large damage


The general pathway is similar in all organism


1. An exinuclease will bind to the bulky lesion, and cleave on both sides


- 13 nucleotides in E coli, 29 in humans


2. The DNA segment is removed with helicase


3. GAP is filled by DNA polymerase


- DNA pol I in E.Coli, DNA pol e in humans


4. Nick is sealed by DNA ligase

Direct repair

This a repair mechanism without removing a base or nucleotide




DNA photolyase is an example of an enzyme which uses energy from absorbed light to reverse the damage to pyrimidine dimer


- Folate and FADH- as cofactors




Nucleotides with alkylation damage can also be repaired in a similar way


- Methylguanine nucleotide can be directly repaired with methyltransferase




Alkylated bases can also be reparied by AlkB


- a-ketoglutarate + O2 -> succinate + CO2, OH group is attached and removed by formaldehyde in the next step

What are the two mechanism for double stranded DNA breaks?

Non homologous joining


Homologous recombination DNA repair

What are similatirities and differences in DNA polymerase and RNA polymerase?

Similarities


- Direction of synthesis -> 5'-3'


- Mechanism of elongation


- hydrolysis of pyrophosphate


- Procesivity




Special features of RNA polymerase


- 5'pppG or pppA (tail?)


- No need for primer


- No exonuclease activity


- Template -> one strand of DNA

What are the types of subunits of RNA polymerase

rpo A


- Enzyme assembly


- Promoter recognition


- binds some activators


rpoB


- Catalytic center -> Chain initiation, elongation, binds to DNA template


rpoC


- catalytic center


rpoD


- promoter specificity

How is transcription initiation of RNA regulated? and how is the specificity controlled?

Holoenzyme with a sigma factor will recognize one set of promoters


- A substitution of sigma factor will cause the enzyme to recognize a different set of promoters


- Promoters will therefore control the expression of a set of gene




A sigma factor controls the specificity


- Normally a core enzyme will bind to any DNA, but sigma will destabilize this binding


- The holoenzyme will bind to the promoter

What is the sigma cycle?

RNA polymerase will bind to DNA at promoter sequence guided by a sigma subunit


Once the RNA synthesis has started the sigma subunit will dissociate, and be replaced by NusA


When the RNA polymerases reach the terminator sequence, the RNA synthesis will halt.


- NusA dissociates, and the RNA polymerase dissociates




The free polymerase can bind to any sigma subunit


-

How is RNA transcription regulated?

Strength of promoter


- Close to consensus


Activator/repressor molecules


Sigmoid factor


RNA degradation


Antitermination


attenuatiuon

Antitermination

Normally the RNA polymerase will stop at the terminator, only transcribing the previous region.


But in this case both regions will become transcribed, as the RNA polymerase continues


- RNA will represent region 1+2

What type of RNA synthesis terminations are there?

Rho-dependent


- Moves along the RNA, catches up with the transcription complex when it stops at a termination site


- Rho portein associated with RNA and migrates in 5'-3' direction, reaching the transcription complex.


- At the transcription complex is causes a release of RNA transcript via ATP-depent RNA-DNA helicase activity of Rho




Rho-indepedent or intrinsic


- Formation of a hairpin structure which disrupts many A=U bonds


- Hairpin loop is GC-rich


- Many U's at the end of 3' transcript

What inhibits RNA polymerase?

Actinomycin D


- Both eukaryotic and prokaryotic


Rifampicin


- Prokaryotic RNA polymerase beta unit


A-amanitin


- Pol II, III


- death cap

What an eukaryotic consensus sequence?

TATA- Box: TATAAA at -30

What inhibits only bacterial RNA synthesis?

Tetracycline


- Blocks binding of aminoacylt-RNA and A site of ribosome


Streptomycin


- Prevents transition from initiation complex to elongation


- Causes miscoding


Chloramphenicol


- Block peptidyl transferase on ribosomes


Erythromycin


- Block translocation


Riframpicin


- Block initiation of RNA chains

What inhibits both bacterial and eukaryotic RNA synthesis?

Puromycin


- Causes premature release




Actinomycin D


- Prevents RNA synthesis


- Binds to DNA, and blocks RNA movement

What inhibits only eukaryotic RNA synthesis?

Cycloheximide


- Blocks translocation reaction on ribosomes




Anisomycin


- Blocks peptidyl transferase




a-amanitin


- Blocks mRNA synthesis by binding to RNA pol

Transcription of eukaryotes

Seperated in time and space from translation


- Different from prokaryote


- Transcription happens in nucleus, translation in cytosol


Has 3 RNA pol


Transcription factors


- By binding to promoters, and affect rate of transcription


Enhancers


- Increase rate of transcription


mRNA 5´Cap, Poly A tail


RNA editing and splicing


Basal transcription apparatus, which determines starting point


Upstream elements -> Frequency of initiation


Inducible factors



What are the type of RNA polymerases, and what is their location?


What do they transcribe?


How are they affected by a-amanitin?

RNA pol I


- Nucleolus


- Transcribe rRNA


- Insensitive




RNA pol II


- Nucleoplasm


- Precursors of mRNA and snRNA


- Strongly inhibited




RNa pol III


- Nucleoplasm


- tRNA and 5s rRNA


- Inhibited by high concentrations

How is the transcription termination of RNA pol in eukaryotic cells?

Pol I


- termination factor


Pol II


- Termination can happen at multiple sites




Pol III


- Needs a run of uridins

Ribozymes

Group I introns


Rnase P


rRNA


Spliceosomes


Hammerhead ribozymes

5'cap

7-methylguanosine is joined to the end of most eukaryotic mRNAs


- GTP is methylated


Cap structure is synthesizing complex is attached to RNA pol II, and will synthesize the cap early in transcription


- First 20-30 ntds




Function of 5'cap is to protect mRNA, and for mRNA-ribosome binding

Poly A tail

The addition of a poly a tail to the mRNA transcription of eukaryotes


- 80-250 a residues




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


- Built by adding AMP to RNA


- catalyzed by polyadenylate polymerase




Function of poly A


-Protection


- Transport


- Translation enhancement



What are the types of introns?

Group I

- Self splicing


- G cofactor


- Nuclear, mitochondrial, chloroplast




Group II


- Self splicing


- within an intron -> lariat structure


- Adenosine will have 3 phosphodiester bonds in lariat structure


- Mitochondrial, chloroplast mRNA -> fungi, algea, plants




Group III


- Spliceosome introns


- Nuclear mRNA


- Spliceosomes - snRNPs


- snRNAs




Group IV


- tRNA

What are the steps from transcription to mature mRNA in eukaryotic cells?

1. Transcription


2. 5'capping


2. Splicing


- Introns are removed


4. cleavage


- Extra RNA is remvoed


5. adenylation



On which levels are gene expression regulated?

Nucleus:


1. Transcriptional control


2. RNA processing control




Cytosol:


3. RNA transport and localization control


4. Translation control -> protein


5. mRNA degradation control -> Inactive mRNA


6. Protein activity control

What allows a DNA sequence to code more proteins?

Overlapping genes


- Homologous proteins


Translation in different reading frames


Alternative splicing


-




Poly A site choice


- Alternative cleave and polyadenylation pattern


- Gives variable domains of Ig heavy chains

Processing of tRNA

From the primary transcript -> intermediate


- RNase D is cut (3 end)


- RNase P is cut (5'end)


- Base modifications are done -> methylation, deamination and reduction


- CCA addition - Cytosine-cytosine-adenine sequence at the end of 3'end




Intermediate -> mature tRNA


- Splicing

How can RNA be edited?

Addition


Deletion


alteration or deamination

What are the types of short RNA?

piRNA


rasiRNA


qiRNA


tmRNA


gRNA




miRNA


siRNA


saRNA


snRNA


snoRNA

microRNA or miRNA

Regulate the function of many eukaryotic mRNA - post transcriptional regulators


Mediates the silencing of many genes


Noncoding RNAs, and are complementary in sequence to particular regions of mRNA


Regulate mRNA by binding to complementary sequences in 3'UTR of target mRNA


- Cleaving the mRNA


- Suppressing translation




Protects against invading RNA viruses and to control activity of transposons


Role in formation of heterochromatin



siRNA

Small interfering RNA


Binds to mRNA -> silence it

saRNA

Small activating RNA


induce gene activation that is long lasting

snRNA

Small nuclear RNA


Processing of pre-mRNA in nucleus


Part of spliceosome


- Has different types, which have different roles

snoRNA

Small nucleolar


- Guide chemical modification of other RNAs


- Methylation or psudouridylylation

piRNA

piwi-interacting RNA


Largest class of small RNAs in animal cells


RNA-protein complexes with piwi


Transcriptional gene silencing of retrotransposons in germ line cells

rasiRNA

repeat associated small interfering RNA


- Subclass of piRNA


- in the germline


- Establishing and maintaining heterochromatin structure


- Controlling transcripts form repeat sequences


- Silencing transposons and retrotransposons

qiRNA

tmRNA

Transfer messenger RNA


- Bacterial RNA


- Properties similar to tRNA and mRNA


- rescues ribosomes, recycles stalled


- Facilitates degradation of messenger RNA



qiRNA

DNA damage induces its expression


Role in DNA damage response is to inhibit protein translation



M1 RNA

M1 RNA is the catalytic component for ribonuclease P. It functions in the processing of tRNA molecules in prokaryotes

gRNA

Guided RNA


gRNA will function int he editing of certain mRNA, directing where and what changes can occur

what causes an extension of the central dogma?

Retroviruses will extend the central dogma, to include RNA-dependet synthesis of RNA and DNA




Envelope


integrase


Viral envelope proteins


Virus structural proteins


Capsid


Reverse transcriptase

Transposon/retrotransposon

A DNA sequence which can change it's position within the genome, causing mutations and altering the genome size

rRNA

These form the core structure of the ribosome


The ribosomes will translate the tri-nucleotides to amino acids -> translating proteins from mRNAs

tRNA

Used by the ribosome to translate the trinucleotide


On one end of the loop a trinucleotide is displayed, on the other a corresponding amino acid

What are the types of point mutations?

Silent


Missense


- Wrong amino acid


Nonsense


- Unexpected stop


Frameshift


- Insertion or deletion

What is interferon control?

Virally infected cells will turn of translation to prevent the virus from spreading.


This is done by interferons, a type of cytokine.


The cell response by producing protein kinase R or PKR which phoshporylates elF-2 causing reduced translation


PKR also indirectly induced RNAase L, which causes mRNA degradation

What are some 3 features of the genetic code?

Degenerate


- But unambigous


Conserved


Universal


- only slightly flexible with GMO

What gives the accuracy of the protein synthesis?

Wobble in codon-anticodon pairing


- First two bases in codon -> strong base pairing


- First base in anticodon determines how many codons can be recognized


Proofreading activity of aminoacyl-tRNA synthetases


-Specific interactions between tRNA and aminoacyl-tRNA synthetases


Codon-anticodon pairing check on the ribosome

What components are needed for each stage of the protein synthesis in E.Coli?

1. Activation of amino acids


- 20 amino acids + 20 aminoacyl-tRNA synthetases


- 32 or more tRNA


- ATP, Mg2+


2. Initiation


- mRNA


- fmet


- initiation codon in mRNA


- 30s and 50s ribosomal subunit


- Initiation factor - IF1,2,3


- GTP




3. Elongation


- 70s ribosome - complex


- aminoacyl-tRNA specificed by codons


- Elongation factors


- GTP




4. Termination and ribosome recycling


- Termination coodin in mRNA


- Releasing factors


- EF-G


- IF-3

Proofreading

Aminoacyl-tRNA synthetase has two ends


- Activates the amino acid for peptide bond formation


- Ensure appropriate placement of amino acid in growing polypeptide


The amino acid attached to the tRNA is not checked in on the ribosome, so ensuring this is the correct amino acid is important




The aminoacyl-tRNA synthase will proofread after the formation of aminoacyl-AMP intermediate


- If its the wrong amino acid, it's hydrolyzed to aa and AMP




In addition to this most aminoacyl-tRNA synthetases can hydrolyze the ester linkage between amino acids and tRNAs in the aminoacyl-tRNA


- Greatly increased hydrolysis for incorrectly charged tRNA




The interaction between the aminoacyl-tRNA synthetases and tRNA is the second genetic code


- Important to discriminate between the different tRNA


- To discriminate we need to look at specific nucleotides which are unique


- These are used as recognition site on tRNA for aa-tRNA synthetase

Initiation complex in bacteria

30s


IF-3 -> prevents premature binding of 50s


IF-1 -> A site


mRNA


IF-2 GTP+ fmet-tRNa


50S associates, IF's leave

Initiation in eukaryotes

40s


elF3


PAB -> Poly a binding protein


elF4F


- E -> 5'cap binding


- G -> PAB


- A -> Helicase


elF4B


- scanning mRNA to find first AUG


elF2


- Met-tRNA binding


- Unloading it at the expense of hydrolysis of GTP

Elongation during protein synthesis in bacteria and eukaryotes


- 3 steps

Elongation requires initiation complex, aminoacyl-tRNA , three soluble cytosolic proteins -> elongation factors and GTP


- EF-Tu, EF-Ts, EF-G




1. Binding of incoming aa-tRNA


- EF-Tu +GTP +aa-tRNA enter A site


- GTP is hydrolyzed and EF-Tu-GDP complex is released


- EF-Tu-GDP is regenerated by EF-Ts and GTP




2. Peptide bond formation


- Peptidyl transferase - ribozyme 23s rRNA




3. Translocation


- Ribosomes moves on codon toward 3'end of mRNA


- EF-G is required for movement


- EF-G binds to A site, and mimics peptidyl-tRNA

Proofreading on ribosome

Only the correctness of codon-anticodon pairing is checked


- Mischarged aminoacyl-tRNA can't be checked



What catalyses the peptide bond formation in protein synthesis?

23S rRNA, a ribozyme

Termination of protein synthesis

Termination is signale by presence of termination codons in mRNA


When a termination codon occupies the ribosomal A site 3 termination factors are released


- RF-1, RF-2, RF-3


In eukaryotes eRF recognizes all termination codons




1. These cause hydrolysis of terminal peptidyl-tRNA bond (RF1+2)


2. Release of free polypeptide and last tRNA from P site (RF3)


3. Dissociation of the ribosome complex



What are the difference between prokaryotic and eukaryotic protein synthesis?

Ribosome size and composition


- 70 s -> 50s+30s


- 80s -> 60s + 40s




initiator tRNA


- fMet-tRNA


- Met-tRNA




Initiation


mRNA structure




Elongation and termination is analogous, but through different factors and proteins

What is the energy cost of protein synthesis?

Energy cost


- 2 P /aa-tRNA + 1 P for proofreading


- 1 GTP /First step of elongation


- 1 GTP/Translocation - not necessary


> 4 NTP/peptide bond in prokaryotes




The same in eukaryotes, but + indefinite P for mRNA scranning

What are the optimal regulation points of protein synthesis?

elF2


- Before investing too much energy




elF4E


- the lowest amount of inhibitor cuases the largest relative extent of inhibition




miRNA and siRNA


- partial or total inactivation of mRNA

How is elF-4E regulated?

Transcription


Phosphorylation


Inhibited by interactions with binding proteins



Antibiotics

Molecules or substances used to kill microorganisms


Act against protein synthesis, mostly on the ribosome


Ribosome is highly conserved, making it hard to find a species specific inhibitor




Microorganisms tend to become resistant

Protein targeting

Directs the structures to specific destinations by synthesizing a signal sequence of amino acids on the end


Signal recognition particle binds to ribosome, and direct it to SRP receptor on ER.


- Binds GTP


Ribosome receptor and SRP receptor are found on the outside of ER


On the inside peptide translocase complex is found


The product is guided into the ER, and signal peptidase will cleave the signal sequence

Where does N/O-linked oligosaccharides happen?

N-linked in ER


O-linked in golgi

Transported into mitochondria

Tom receptor + pore


- Outer mitochondrial membrane


Tim receptor + pore


- Inner mitochondrial membrane

Transported into nucleus

Nuclear localization sequence attached to protein


Protein with NLS binds to importin a and B


Translocated to the nucleus through a nuclear pore complex


- RAN

Which chemical bonds participate in stabilizing protein structure?

Disulfide bridges


H-Bonds


Ionic interactions


Hydrophobic interactions

Folding

A spontaneous process, which sometimes proteins cannot do on their own


- Cooperative process -> entire protein is folded or nothing

What are chaperones?

Proteins which assist in the correct folding of polypeptides


- Proteins assisting in the correct folding of polypeptide


- Does this by preventing wrong folding




They catalyze the formation of functionally active proteins, but are not a part of the product




Heat shock proteins


- Chaperons induced by stress, such as heat

What induces heat shock proteins?

Heat shock proteins are induced by stress, such as heat.


During heat-stress the probability of incorrect folding is much higher

What is the role of nucleoplasmins?

Assembly of chromatin




DNA+histone -> aggregate




DNA+nucleoplasmin+histone -> nucleosome + nucleoplasmin



Degradation in bacteria

1. Lon, ATP dependent protease


2. Other proteases

Degradation in eukaryotes

1. Ubiquitination


- Signal death for proteins


- Needs 3 enzymes -> E1, E2, E3


2. Proteasome


- Enzyme where the proteins are degraded


- 26s with a core and regulatory particles(caps)

What can the UPS regulate?

Ubiquitin-proteasome system regulate


- Gene transcription


- cell cycle


- Apoptosis

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

1. Transcription


2. Post-transcriptional processing


3. mRNA degradation


4. Translation


5. Post-translationsal processing


6. Protein degradation


7. Protein targeting and transport

How is gene expression regulated in eukaryotes?

By changing the structure of chromatin we change the access to the eukaryotic promoters.


- Heterochromatin - Euchromatin - Hypersensitive site


This is mostly a positive mechanism

what is the process of gene expression?

Combined process of


- Transcribing a gene into mRNA


- Processing of that mRNA, and it's translation into protein

What are 4 gene regulatory mechanisms?

1. Transcriptional mechanism


- Type of promoters and RNA polymerases


- Control of transcription by transcription factors and TFBS




2. RNA processing


- 5' capping and 3' poly-adenylation


- RNA degradation rates


- Splicing and alternative splicing




3. Translational mechanism


- MicroRNA (miRNA) inhibit translation and degrade mRNA


- silencer RNA or siRNA degrade mRNA




4. Epigenetic mechanism


- DNA methylation


- Histone modifications -> Acetylations, methylations, phosphorylation


- Chromatin remodeling

What are enhancer elements, and how d they affect the transcription?

Enhancer elements are DNA sequences


No promoter activity of their own, but greatly increase activity of many promoters in eukaryotes


Functions as binding site for specific regulatory proteins


Only effect in specific cell type, with appropriate regulatory proteins




Can work both as an upstream and downstream promoter


They can stimulate any any promoter in the vicinity and may act on more than one

HIF-1 alpha

HIF-1 is synthesized during conditions of low oxygen concentration


- Hypoxia inducible factor




HIF-1 regulates gene expression to reduce ROS formation


HIF-1 acts as a transcription factor, and increases synthesis of


- Glucose transporter


- Glycolytic enzymes


- PDH-kinase


- Lactate dehydrogenase


- Protease which degrades cytochrome oxidase




These changes will all counter the formation of ROS, by decreasing the supply of NADH and FADH2, and by making cytochrome oxidase of complex IV more effective.




when electron flow of FADH2 and NADH to respiratory chain decrease, the formation of ROS is reduced.

What activates the HIF-1a domain in the nucleus?

Interaction with p300



Name 4 enzyme that FoxO upregulate?

G6Pase


G6Pase transporter


HMG-CoA synthase


PDK4

What is the function of SIRT1 deacetylase on transcription factors?

It inhibits p53, foxo and PPAR


Stimulates PGC1a and LXR





DNA replication in mitochondria

Happens in matrix


Replication not limited to S-phase


Replication at random


Double stranded ring, nucleoid


Has no histones, no introns


Polycistronic transcription


Attached to inner membrane

Which proteins are encoded into the mtDNA?

Components of the respiratory chain


- Complex I, III, IV, ATP synthase, transfer RNA, ribosomal RNA, control region of DNA

Dense packing in mtDNA

Almost every nucleotide is part of a coding sequence, very limited regulatory sequence

Relaxed codon usage

Only 22 tRNA for protein synthesis


Normal codon-anticodon pairing rules are relaxed

Variable genetic code

4 codons have different meaning

What is required for replication of mtDNA?

Replisome


- mtDNA polymerase y


- Twinkle -> 5'-3' helicase


- SSB protein


- TFAM - transcription activator mitochondria




very reduced accuracy





Transcription of human mtDNA

POLRMT


- mitochondrial RNA polymerase


TFAM, TFB1M, TFB2M


Both mtDNA strands are transcribed at the same rate


Each strand has a single promoter -> Two large polycistronic precursor RNA


- Completely symmetric transcription


- Strand 1: 2 rRNAs, most tRNAs, 10 poly-A-RNAs


- Strand 2: 8 tRNAs, 1 small poly-A-RNA -> no useful info




No cap at 5'end, poly-A at 3'end added post-transcriptionally

How is mtDNA inherited?

It's inherited maternally


Non-mendelian, cytoplasmic inheritance

What are hormones?

Chemical messengers of the endocrine system

What are the 6 steps a hormone goes through as a signal?

1. Synthesis


2. Storage and secretion


3. Transport to target cell


- Endocrine, paracrine and autocrine


4. Recognition by receptor


5. Relay and amplification of hormonal signal


- Leads to cellular response


6. Degradation of hormone

Endocrine vs paracrine vs autocrine

Endocrine action - Hormone diffuses to distant regions of the body carried by blood


Paracrine action - Hormon is released into the extracellular space and diffuses to neighboring cells


Autocrine - Hormone acts on same cell that produced it

What are peptide hormone, how are they made and what do they bind to?

Insulin, glucagon, somatostatin etc


made by proteolytic cleavage of precursor proteins


Bind to receptors in plasma membrane

Catecholamine hormones



Epinephrine, norepinephrine


Plasma membrane receptors



Steroid hormones

Nuclear receptors


Derived from cholesterole


Cortisol, progesterone etc

Vitamin D

Nuclear receptors


Calcitrol

Retinoid

From vit A


Nuclear receptors

Thyroid hormones

Nuclear receptors




T3 from tyr in thyroglobulin

How does epinephrine desensitize B-adrenergic receptors?

The binding to BARK causes it to dissociate


It becomes phosphorylated on the carboxyl terminus of the receptor


The complex enters the cel by endocytosis, and arrestin dissociates


Receptor is then dephosphorylated and returns to surface

JAK

Janus kinase

STAT

Signal transducer and activator of transcription

JAK-STAT pathway

1. Ligands bind to receptor, activating JAK, increasing its kinase activity


2. Activated JAK phosphorylates receptor, creating a binding site for STAT


3. STAT is recruited tor receptor, and phosphorylated by JAK


4. Activated STATs form dinners and move into nucleus where they induce transcription of target genes

How is the JAK-STAT pathway regulated?

Protein tyrosine phosphatase will remove phosphates from receptors and STAT


- Deactivates




SOCS inhibit stat phosphorylation


Protein inhibitors of activated STAT or PIAS works in nucleus to inhibit transcriptional activation by STAT

What 2 mechanism does erythropoietin receptor work on?

JAK-STAT pathway


via GBR2 and MAPK cascade