Exam 2 Flashcards

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

What is gluconeogenesis?

A

making glucose from pyruvate

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

What is glucose?

A

breaking down glucose into pyruvate

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

What is PEPCK

A

regulated by Fox01 which activates it

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

What does Akt do in relation to Fox01?

A

it phosphorylates it not allowing it into the nucleus to transcribe

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

What does act do with glyconeogenesis

A

It inhibits GSK3-B which inhibits glycogen synthase which would inhibit glycogen synthase but with it inhibited more glycogen is created

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is antagonistic pleiotropy?

A
  • genes conveying a benefit for fitness early in life will be selected even though they may be disadvantageous in later life. May contribute to the aging process.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What do the various cyclin molecules do in the cell cycle?

A

o Cdks (Cyclin-Dependent Kinases): Activity changes as progresses through cell cycle
o Cyclins:Regulate cdk activity through direct binding with 4 classes:
➢ G cyclins: promotes passage through Start (Cyclin D)
➢ G1/S cyclins: Bind cdks at end of G1 and commit to DNA replication (Cyclin E)
➢ S-cyclins : Initiate DNA Replication (Cyclin A)
➢ M-cyclins: Triggers entry into Mitosis (Cyclin B)
o CKIs (Cdk inhibitor proteins): Negatively regulate cell cycle
o The progression of the cell cycle occurs through a combination of: transcriptional regulation, cyclical proteolysis, and activity regulation

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

What occurs at the G1 phase?

A

• G1 phase, cell cycle initiates when cell receives an extracellular signal from “mitogens”- G1 phase Cyclin 1 CDK- phosphorylats and

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

What occurs at the G1/S phase?

A

• G1/S phase- DNA damage main driver. p53- is the gatekeeper of the genome is activated in many stressful situations, DNA damage will be sensed by proteins and p53 is phosphorylated and then accumulates in the cell.

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

What occurs at the Sphase?

A

• S phase initiates origin firing prevents pre replication- using cyclin A and cdk 2

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

What occurs at the M phase?

A

entry into M phase begins with the upregulation of cyclin B (M-cyclin) transcription, the activation of cdc25 (polo kinase, cdk1), removes inhibitory strain on cdk1, which triggers mitosis. M-cyclin binds to Cdk1 in inactive form, and then the cdk activating or inhibiting kinase removing the phosphate via a phosphatase which becomes active with a phosphorylation of cdc 25. M-cyclin comes together and becomes activated when they are dephosphorylated. Then the active-M-cdk with only one phosphate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What occurs at the metaphase to anaphase phase?

A

• Metaphase to Anaphase checkpoint- APC- anaphase-promoting completx, with a ubiquitin ligase- which initiates proteasomal degradation, initiates sister-chromatid separation, degradation of securing- releasing cohesions. Leads to the degradation of securing which allows the sister chromatids to actually separate.

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

What happens at the G2/M phase

A

G2/M-DNA damage inactivates cdc25- inhibited if there is DNA damage affecting the phosphatase which inhibits the earlier phosphate adding to the Cdk.

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

What occurs at the G0 phase?

A

allows cell to carry out physiological role without diverting energy to cell cycle.

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

What is cell senescence?

A

o Cells are NOT DEAD!- this isn’t apoptosis so they are irreversibly arrested but not apoptosis
o Alternative to death
o Enlargement of size of nucleus
o Distance between cells becomes greater
o Irrreversibly arrested in G0 (therefore different from quiescent cells)
o Only affects mitotic cells

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

What causes cells to senesce?

A

DNA damage, telomere shortening, and stress

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

What is telomere shortening?

A

evolved as a way of loss of coding sequence, losing telomeres instead of losing genes that code for things. predicts that repeated replication of a chromosome shortens the telomeres to the point at which no further replication can occur without affecting the coding sequences of the DNA.
➢ The repetitive, noncoding base-pair sequences at the ends of each chromosome
➢ Replication of the lagging strand template results in the loss of a short segment of the telomeres
➢ After repeated cell divisions, eventual complete loss of telomeres
➢ Telomerase: telomere elongation, protecting it.
➢ Only in germ line and stem cells: cells which require high fidelity during replication (only care about those germ cells…)
➢ Mitotic Clock Theory: predicts that old cells sense short telomeres, which, in turn, causes cell cycle arrest
➢ telomere shortening activates p53 upregulating p21 inhibit cell cycle from dividing and eventually lead to senescence
➢ Attractive model because: it is well known phenomenon that occurs with age, gametes have a way to overcome this stress (telomerase), but it is hard to decide if telomere shortening is simply a cause of aging (like grey hair) or a diriver of the aging process

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

How does DNA damage lead to senescence/

A

DNA damage via oxygen radicals, UV light, x-rays, errors in replication. Use base-excision repair removing the base, nucleotide excision repair- remove strands, recombination repair, and mismatch repair- repair the base. Try to repair and if you cant you want that cell to die or senesce

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

How does replicative senescence cause aging in organisms/

A

SASP (Senescence-Associated secretory Phenotypes)- with cellular senescence occurring over time in vitro, proliferative capacity decreased with age. Senescent cells secrete interleukins, inflammatory cytokines (IL-6), and growth factors (MPs) that can affecting the surrounding cells. Can secrete things for them to become cancerous
• Cellular Senescnce when you are young is cancer prevention regulation, but with prolonged senescence promoting aging and cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the free radical theory of aging?

A

o Oxidative damage caused by Free radicals drives the aging process, Denham Harman developed hypothesis in 1956 with lots of supporting data, with no direct evidence.
o Theory has evolved: “mitochondrial theory of aging”-oxidatve damage to the mitochondria itself drives the aging process
o Free radicals are Reactive Oxygen species (ROS), defined as reactive chemical species having a single unpaired electron in their outer orbit. O2-, OH-, and H2O2
o ROS is produced in peroxisomes, released by phagocytic cells, and the biggest source is mitochondria producing 95% of them

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

What can these radical oxygen species damage?

A

Biological membranes, DNA and RNA, and proteins cross linking them making them not function

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

how doesradical oxygen damage affect phospholipids and how do u remove it?

A

➢ Oxidative stress causing the double bonds to break as it is susceptible to attack by OH with liquid peroxide (lipid peroxidation) affecting people in diseases such as artherosclerosis, stroke, alzherimer’s disease
➢ Removal of lipid damage: multi-step process that includes Vitamin E, Vitamin C, Glutathione, and NAD+

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

How does radical oxygen damage affect DNA?

A

breaks the base and sugar backbone. Activated DNA repair pathways can cause senescence which can help avoid mutations by shutting down replication, but these same pathways can eventually cause aging

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

What is autophagy?

A

degradation of intracellular components in lysosomes, including damaged organelles and damaged biomolecules, components are then recycled for use in making new proteins, lipid and to synthesize new DNA

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

Why do we have autophagy?

A

alternative source of energy (during starvations. Cellular quality control (degrade damaged proteins) to turnover of cellular components and removal of damage before they interfere with normal cell function.

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

What are the modulators of autophagy?

A

o Modulators of autophagy with nutrient signaling (starvation activates autophagy) such as mTOR, Ras/PKA, Insulin activating autophagy due to starvation. Stress response (damaged biomoleculesactivate autophagy such as ER stress, hypoxia, oxidative stress, pathogen infection.

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

What is mTOR?

A

➢ mTor (mechanistic target of Rapamycin)- when nutrients are high, mTOR is activated to promote protein synthesis and inhibit autophagy through the phosphorylation of ULK1 but when mTOR is inhibited, the “brakes” are relased and autophagy will ensue. Inhibitors of this pathway are rapamycin and analogs, Torin1 PP242. When mTOr is activte inhibiting ULK complex. when reduced in all organisms they have extended life span

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

What proteins are involved in induction that when halted stop autophagy?

A

ULK-1, Atg17, and Atg13

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

What does rapamycin do?

A

rapamycin activates autophagy by inhibiting mTOR

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

What does sirtuin do?

A

o Regulate the acetylation of histones and other proteins:p53, Foxo1

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

What does the TOR pathway do?

A

Controls and regulates protein synthesis and growth in response to nutrients.

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

What does dietary restriction do?

A

with 30-40% less calories it is the o Only nongenetic method to extend life span to date

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

Why is it believed that calorie restriction works?

A

restriction will reduce the rate of metabolism thereby reducing the production of damaging free radical. After prolonged dietary restriction they have increased metabolisms such as oxygen consumption, which is not what we expected and a mix of if oxidative damage is caused by dietary restriction.

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

What would dietary restriction look like with genetic or pharmaceutical?

A

Genetic or Pharmaceutical- Longest life span curve is in the sweet dietary restriction line, if you treated with drug mimetic it would be moved to the right so same amount as normal and the same life span. Drug that inhibited bell curve so gene was regulatory of dietary restriction or deleted the gene. If there was an additive there would be an extension and would be a separate pathways with a lifespan extension and reduction because it is shifting downwards but still the same general format.

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

How would you test to see if genes were in the same or different pathway?

A

delete each, then both and see the results if it is an additive affect in the multiple than it is not in the same pathway, if it is the same then it is in the same pathway

36
Q

What is the sequence of glucose-stimulated insulation excretion?

A

o Glucose enters pancreatic β-cell through GLUT2 (transporter)
o Glucose is metabolized
o Rise in ATP closes the ATP-sensitive K+ channels and prevents K+ ions from leaving the cell
o This causes the inside of the cell become more positive (depolarized)
o Voltage-dependent Ca2+ channels to open
o Ca2+ will diffuse down concentration gradient into the cell
o Ca2+ causes insulin-containing vesicles to fuse with membrane and insulin to be secreted by exocytosis.

37
Q

What happens when insulin binds to cells?

A

• insulan binds causing autophosphoryaltion of its receptor and the other things bind akt is then activated by phosphorylation by PDK1 and this is inhibited by Fox01.

38
Q

what are the 5 main pathways akt activates?

A

glucose uptake and storage, cell proliferation and growth, cell survival and promotes protein synthesis by activating indirectly

39
Q

How is glucose uptake?

A

Akt is a kinase phosphorylating As160 and RGC, which are involved in the translocation and targeting of this Glut4 vesicle moving to the cell membrane. Once Glut 4 on membrane glucose is uptaken by that cell.

40
Q

How does insulin lead to glucose storage? ?

A

Akt inhibits gluconeogenesis in the liver by inhibiting Fox01 getting rid of phosphate which is a transcription factor that would have activated PEPCK (gluconeogenesis).

It then activates glyconeogenesis Glucose storage akt inhibits glycogen synthase kinase (which normally blocks transcription) activating glycogen synthase.

41
Q

What are the different factors that akt affects?

A

Wee1, Myt1, p27, p21, Cyclin D, Cdc25

42
Q

How does akt activate wee1?

A

o Akt phosphorylates and inhibits wee1, which then allows cdk 1 to go forward promiting the cell cycle to go into M phase.

43
Q

How does akt activate myt1?

A

akt phosphorylates and inhibits myt 1 from phosphorylating cdk1 which then allows the cell cycle to go into the m phase

44
Q

How does akt activate p27?

A

Akt inhibits fox01 which inactivates the transcription of p27 allowing activation of cyclin D and E making the cell cycle go through G1 and into the S phase

act also phosphorylates and inhibits p27 or by sequestering it in the cytoplasm and not in the nucleus. this then allows the activation of cyclin D and E making the cell cycle go through G1 and into the cyclin cdk complex through G1 into S phase

45
Q

How does akt activate p21?

A

Akt inhibits fox01 which inactivates the transcription of p21 allowing activation of cyclin D and E making the cell cycle go through G1 and into the S phase
o akt phosphorylates and inhibits p21 c allowing activation of cyclin D and E making the cell cycle go through G1 and into the s phase

46
Q

How does akt activate cyclin d?

A

o akt inhibits Gsk3B1 which then inhibits the degradation of cyclin d allowing it to press into through G1 phase

47
Q

How does akt activate cdc25?

A

o akt inhibits Gsk3B1 which inhibits the degradation of cdc25 which activates cdk1 promoting it past the G2/Mphase

48
Q

how does akt inhibit cell death?

A

it inhibits fox01 which decreases the transcription of BIm a proapoptotic factor

49
Q

What are the human and c. elegans comparison names?

A
  • Daf-2 insulin receptor
  • age-1- Phosphoinositide 3-kinase phosphorylating pip2 to pip3 is inhibited by daf-16
  • daf-16- fox01 need daf16 working for autophagy to extend daf2 lifespan
  • akt is the same
50
Q

How does insulin receptor deletion affect lifespan?

A

when deleted it increases life span by up regulating stress response proteins

51
Q

What is the ribosomal s6 kinase?

A

Activates S6, a component of the 40S ribosome that is required for protein translation

52
Q

What is eukaryotic Initiation Factor 4E-BP1?

A

4EBP1 normally inhibits protein translation by binding to eIF4E. Phosphorylation by mTOR causes dissociation of 4E-BP from eIF4E allowing translation to occur.

53
Q

What is epistasis?

A

is a phenomenon that consists of the effect of one gene being dependent on the presence of one or more ‘modifier genes’. using Microarrays as a way to measure changes in gene expression in the entire genome. Loking at what genes are changing with daf-2 deleted.
• Confirm expression by quantitative RT-PCR, then you can examine the effect of deleting gene on life span on daf-2 mutant,

54
Q

What are the stress response proteins regulated by these response pathways?

A

o Heat shock response (hsf-1)
o Autophagy (bec-1)
o Oxidative stress response (sod-3)

55
Q

What are the proteins necessary for life expansion of age-1, PI-3 kinase deleted worms?

A

need hfs and it is in the same pathway!

56
Q

When deleted daf-2, insulin receptor what other proteins are needed?

A

bec-1 or autophagy but is an independent pathway and it alone does not extend lifespan on its own
needs sod which is unregulated in these and it is required for longevity dependent on daf-16 and is in the same pathway as it is mitochondrial localized and breaks down oxidative stress.

57
Q

What are the similarities between DR and reduced insulin signaling?

A

increased longevity, stress resistance, reduced mTOR signaling, and increased autophagy.

however it is unclear if they are in the same pathway with mixed results

58
Q

What does smelling do to organisms?

A

It increases life span sometimes dietary restriction doesn’t work if they can still smell their food because of the insulin levels rising

59
Q

Summarize results from 1st journal club

A

Here, using heterozygous knockout mice because null mutants are not viable, we report that Igf1r1/2 mice live on average 26% longer than their wild-type littermates(P

60
Q

What is the conclusion of the second journal?

A

As NAD+ is a rate-limiting cosubstrate for the sirtuin enzymes, its modulation is emerging as a valuable tool to regulate sirtuin function and, consequently, oxidative metabolism. In line with this premise, decreased activity of PARP-1 or CD38—both NAD+ consumers—increases NAD+ bioavailability, result- ing in SIRT1 activation and protection against meta- bolic disease. Here we evaluated whether similar effects could be achieved by increasing the supply of nicotinamide riboside (NR), a recently described natural NAD+ precursor with the ability to increase NAD+ levels, Sir2-dependent gene silencing, and replicative life span in yeast. We show that NR supplementation in mammalian cells and mouse tissues increases NAD+ levels and activates SIRT1 and SIRT3, culminating in enhanced oxidative metabolism and protection against high-fat diet- induced metabolic abnormalities. Consequently, our results indicate that the natural vitamin NR could be used as a nutritional supplement to ameliorate metabolic and age-related disorders characterized by defective mitochondrial function.

61
Q

What triggers the Torr pathway?

A

growth factors, amino acids, and insulin

62
Q

What does TORC1 lead to?

A

binds mTor, PRAS and raptor and leads to protein translation (S6K and 4E-BP1), lipid biogenesis (SREBPs), and inhibited autophagy (ULK1)

63
Q

What does TORC2 lead to?

A

binds main, rector and mTor cytoskeletal changes (PKCalpha), cell survival (akt) and stress response (SGK1)

64
Q

What are the two upstream activators of TORC1?

A

akt in high nutrients, and ERk in high nutrients they function to inhibit the tsc complex

65
Q

What are the three upstream inhibitors of TORC1?

A

AMPK (low nutrients), GSK-3B (stress activated protein), and Redd1 (stress activated protein) which activate the tic complex inhibiting TORC1

66
Q

How does TORC1 inhibit autophagy?

A

it binds to ULK1 which is needed in phagophore formation in autophagy inhibiting it

67
Q

Why does reduction in Tor extend lifespan?

A

Not as much proteins made so there are less damaged proteins leading to less ER stress and UPR that lead to autophagy because less S6K means longer life by the same length as reducing 4E-Bp and elf-4E, and autohphagy is no longer inhibited

68
Q

How are tor and insulin interrelated

A

Akt activates Tor downstream so Tor and insulin are in the same pathway

69
Q

What does Fox01 do?

A

leads to gluconeogenesis, cell death, detoxifies, repairs DNA and arrests G1 and G2

70
Q

What does torr need to have extended life span?

A

autophagy/beclin

71
Q

What about TORC2?

A

inhibition of TORC2 is detrimental to longevity giving the mice an inability to tolerate glucose in males only no affect on females

72
Q

How does insulin signaling affect DR?

A

with reduced insulin signaling the results are unclear whether they are connected or not

73
Q

How does mTor connect with DR?

A

There is a very strong linkage in every model tested with reduced mTOR pathway senses nutrients. Delete TOR extend lifespand, the mutant worm displays similar eating phenotypes with dietary restriction. TOR pathway and DR see no double affect suggesting they are in the same pathway

74
Q

what is Activated sirloins affect with DR?

A

there is controversial evidence connecting it but it believed to not cause it

75
Q

What has rapamycin been found to do?

A

lifespan, affecting cancer, cardiovascular disease, neurodegeneration, immune system, and aging. It has negative side affects including aphthous ulcers, edema, hyperlipidemia, immune suppression, glucose intolerance, and reduced male fertility.

76
Q

What does rapamycin bind to and do?

A

Rapamycin binds to FKP12 which directly bind to TORC1 causing it to dissociate with an acute treatment between 15 minutes-1 hour. 24 hours or longer with free mTOR, rapamycin and FKBP12 does not directly bind the complex and so it binds to mTOR removing it from the pool so TORC2 doesn’t form and this happens on multiple cell lines but not all tissues for TORC2

77
Q

What is SIRT?

A

they are cellular stress sensors that modify histones and other proteins modulating diverse cellular processes including metabolism, stress response, and insulin signaling

78
Q

How is NAD+ produced in the cell?

A

from tryptophan through de novo production as well as the salvage pathway with sirtuin signaling leading to the salvage of NAD+ with sirtuins using it to deacetylate histones and proteins
and it decreases with age so supplementing with NMN improves life span

79
Q

How does SIRT1 contribute to aging?

A

It deacetylates Fox01 and p53

80
Q

What does deacetylation of Fox01 do?

A

it promotes oxidative stress resistance and inhibits apoptosis because the life extension by sirloins depends on Fox01

81
Q

What does sirtuins do to p53?

A

it deacetylates it meaning that p53 cannot unwind and transcribe p21 meaning the cell cycle is not stopped as it should be when p53 senses DNA damage

82
Q

What animals does sirtuins work on to extend lifespan?

A

it works on yeast, worms, and flies but sirt1 does not in mice it only affects glucose homeostasis, wound healing, and delayed bone loss because it could not protect against lymphoma
but sort does extend median life span

83
Q

What leads to higher amounts of Sirt expression?

A

nutrient deprivation and endurance exercise enhances SIRT1 mRNA expression

Fox01

resveratrol activates it and improves health and surivial of mice on a high fat diet by inhibiting mitochondria metabolism and increasing NAD+ levels as does supplementing with NR and NMN
PARP reduces NAD+ with age as it is activated by DNA damage

84
Q

What was the largest functional category of long-lived deletions?

A
  1. cytosolic ribosomes supporting the theory that reduced protein synthesis extends life span
  2. then mitochondrial translation with less free radical production
  3. TCA cycle less NADH and FADH2 produced elevating NAD levels extending life span as well as TOrr
85
Q

What was the longest lived single deletion?

A

UBR2 because it leads to the degradation of Rpn4, transcription factor for proteasome and so with less more Rpn4 increasing proteasomal subunits

86
Q

What is los1?

A

a nuclear tRNA exporter that blocks nuclear export or enhances nuclear import of tRNAs enhancing the life span. with its deletion there is more tRNA where they are inactive reducing protein synthesis but no patterns were found and it could be due to dietary restriction because of a similar amount but didn’t work