• Log in with Facebook Log in with Twitter Log In with Google      Sign In    
  • Create Account
  LongeCity
              Advocacy & Research for Unlimited Lifespans

Photo

Aging Theories (cira)


  • Please log in to reply
99 replies to this topic

#61 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 15 May 2003 - 10:37 PM

Cynthia Kenyon, noted anti-aging researcher has determined that small heat shock proteins aid in preventing the clumping of proteins that is seen in many age-related diseases.

The growing roster of diseases thought to be caused by protein clumping or aggregation -- Alzheimer's, Huntington's, Parkinson's, prion diseases -- suggests that the small heat shock proteins may influence the onset of many age-related ailments, the researchers say. The pharmaceutical industry is already exploring ways to increase the activity of heat-shock proteins. The research by Kenyon's laboratory indicates that if these drugs work, they may not only protect protein function, but also extend life.


A reduction in small heat shock protein expression as we age is thought to be related to the susceptibility of older people to age-related illnesses.


Researchers discover common cause for aging and age-related disease

Link to Abstract

Edited by kperrott, 16 May 2003 - 05:29 AM.


#62 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 15 May 2003 - 10:54 PM

EE) Comment on Program Theory

I have to agree with you on all points ocsrazor. I didn't really agree with the conclusions drawn but thought them interesting enough to warrant some refutation/discussion. I knew you in particular would be able to bring some clarity to the post. :)

One thing that wasn't mentioned in the article either was the possibility that the "negative" changes in gene expression may be to compensate in response to other even more "negative" changes.

Edited by kperrott, 15 May 2003 - 10:57 PM.


Click HERE to rent this BIOSCIENCE adspot to support LongeCity (this will replace the google ad above).

#63 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 16 May 2003 - 05:34 AM

A search of PubMed.com using "herbal induction heat shock protein" came up with this...as well as others..

Curcumin, a medicinal herbal compound capable of inducing the heat shock response.

I'm not sure if it's directly applicable to the results in Kenyon's study but it's worth a read.

sponsored ad

  • Advert

#64 Mondey

  • Life Member
  • 69 posts
  • 106
  • Location:UK

Posted 23 June 2003 - 09:44 PM

Dear kperrott,
from personal experience from cell culture experiments I can tell you take vitamin E(alpha- r,r,r-tocopherol). This would help your cells, to restore protein synthesis and protein degradation. The only problem with the damm vitamin, taht there are special protein receptors and protein carriers, which highly regulates the blood concentration, and even more the brain concentration of vitamin E.
As you mentioned the articel from Hipkiss, try his articles about carnosine, which can make protein aggregates, normally non-degradable, again prone for the proteolytic systems. Carnosine can therefore be used for example as a cure for catarats.
And I think the cells try to get rid of the damaged protein material by aggregating these materials and accumulates is somewhere in the cells, so that it doesn't interfere with the normal protein turnover.


Greetings Dey

#65 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 09 July 2003 - 02:09 PM

Carnosine is part of my regimen. The existence of the enzyme 'carnosinase' which breaks down carnosine makes me wonder just how beneficial carnosine supplementation may be. If the body is breaking it down, perhaps higher levels have deleterious effects although this has not been demonstrated by the studies.

Here's link to an abstract by Hipkiss on Carnosine which also mentions the benefits of n-t-butyl-hydroxylamine (NTBHA)
------------------------------------------------------------
Ann N Y Acad Sci. 2002 Apr;959:285-94. Related Articles, Links

Reaction of carnosine with aged proteins: another protective process?

Hipkiss AR, Brownson C, Bertani MF, Ruiz E, Ferro A.

GKT School of Biomedical Sciences, King's College London, Guy's
Campus, London Bridge, London SE1 1UL, United Kingdom.
alan.hipkiss@kcl.ac.uk
--------------------------------
Cellular aging is often associated with an increase in protein carbonyl groups arising from oxidation- and glycation-related phenomena and suppressed proteasome activity. These "aged" polypeptides may either be degraded by 20S proteasomes or cross-link to form structures intractable to proteolysis and inhibitory to proteasome activity.

Carnosine (beta-alanyl-l-histidine) is present at surprisingly high levels (up to 20 mM) in muscle and nervous tissues in many animals, especially long-lived species. Carnosine can delay senescence in cultured human fibroblasts and reverse the senescent phenotype, restoring a more juvenile appearance. As better antioxidants/free-radical scavengers than carnosine do not demonstrate these antisenescent effects, additional properties of carnosine must contribute to its antisenescent activity. Having shown that carnosine can react with protein carbonyls, thereby generating "carnosinylated" polypeptides using model systems, we propose that similar adducts are generated in senescent cells exposed to carnosine. Polypeptide-carnosine adducts have been recently detected in beef products that are relatively rich in carnosine, and carnosine's reaction with carbonyl functions generated during amino acid deamidation has also been described. Growth of cultured human fibroblasts with carnosine stimulated proteolysis of long-labeled proteins as the cells approached their "Hayflick limit," consistent with the idea that carnosine ameliorates the senescence-associated proteolytic decline. We also find that carnosine suppresses induction of heme-oxygenase-1 activity following exposure of human endothelial cells to a glycated protein. The antisenescent activity of the spin-trap agent alpha-phenyl-N-t-butylnitrone (PBN) towards cultured human fibroblasts resides in N-t-butyl-hydroxylamine, its hydrolysis product. As hydroxylamines are reactive towards aldehydes and ketones, the antisenescent activity of N-t-butyl-hydroxylamine and other hydroxylamines may be mediated, at least in part, by reactivity towards macromolecular carbonyls, analogous to that proposed for carnosine.

Publication Types:

* Review
* Review, Tutorial


PMID: 11976203 [PubMed - indexed for MEDLINE]
http://www.ncbi.nlm....3&dopt=Abstract

Edited by kperrott, 09 July 2003 - 02:11 PM.


#66 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 09 July 2003 - 02:28 PM

EE) Comment on Program Theory

The following paper asserts that polymorphism in genes involved in the IGF-1 pathway is associated with human longevity indicating that genetics may have a more active role to play in regulating lifespan. I wonder if more weight needs to be given to the role that genetics plays in aging?

------------------------------------------------------------
J Clin Endocrinol Metab. 2003 Jul;88(7):3299-304.


Polymorphic variants of insulin-like growth factor I (igf-I) receptor and phosphoinositide 3-kinase genes affect igf-I plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control.
Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E, Giampieri C, Mugianesi E, Centurelli M, Franceschi C, Paolisso G.

Department of Experimental Pathology, University of Bologna, 40126 Bologna, Italy.

Current literature indicates that abrogation of the IGF-I response pathway affects longevity in Caenorhabditis elegans, and that the down-regulation of IGF-I gene expression is associated with an extension of the life span in mice. In this paper we tested the hypothesis that polymorphic variants of IGF-I response pathway genes, namely IGF-IR (IGF-I receptor; G/A, codon 1013), PI3KCB (phosphoinositol 3-kinase; T/C, -359 bp; A/G, -303 bp), IRS-1 (insulin receptor substrate-1; G/A, codon 972), and FOXO1A (T/C, +97347 bp), play a role in systemic IGF-I regulation and human longevity. The major finding of this investigation was that subjects carrying at least an A allele at IGF-IR have low levels of free plasma IGF-I and are more represented among long-lived people. Moreover, genotype combinations at IGF-IR and PI3KCB genes affect free IGF-I plasma levels and longevity. These findings represent the first indication that free IGF-I plasma levels and human longevity are coregulated by an overlapping set of genes, contributing to the hypothesis that the impact of the IGF-I/insulin pathway on longevity is a property that has been evolutionarily conserved throughout the animal kingdom.

PMID: 12843179 [PubMed - in process]

Link to Abstract
Link to PDF

Edited by kperrott, 09 July 2003 - 02:30 PM.


Click HERE to rent this BIOSCIENCE adspot to support LongeCity (this will replace the google ad above).

#67 caliban

  • Topic Starter
  • Admin, Advisor, Director
  • 9,152 posts
  • 587
  • Location:UK

Posted 31 July 2003 - 05:25 PM

In about 2 weeks time, starting from the 14th of August - material, information, issues raised and opinions expressed in this thread will be drawn together and - with a lot of additional information- be forged into a single webdynamic document of distilled high- quality information.

Until then contributions are still possible and sought.

Those who wish to contribute to the editing of the final document should send me a message.



#68 DJS

  • Guest
  • 5,798 posts
  • 11
  • Location:Taipei
  • NO

Posted 31 July 2003 - 08:23 PM

EE) EVOLUTION AND AGING

I put this quote on another thread but thought it would also be relevant here.

The Selfish Gene, p.42

We have already asked what are the most general attributes of a "good" gene, and we decided that selfishness was one of them.  But another general quality that genes will have is a tendency to postpone the death of their survival machine at least until after reproduction....A gene that makes its possessors die is called a lethal gene.  A semi-lethal gene has some debilitating effect, such that it makes death from other causes more probable.  Any gene exerts its maximum effect on bodies at some particular stage of life, and lethals and semi-lethals are not exceptions.  Most genes exert their influence during foetal life, others during childhood, others during young adulthood, others in middle age, and yet others in old age.  (Reflect that a caterpillar and a butterfly it turns into have exactly the same set of genes.)  Obviously lethal genes will tend to be removed from the gene pool.  But equally obviously a late acting lethal will be more stable in the gene pool, provided its lethal effect does not show itself until after the body has had time to do at least some reproducing.  For instance, a gene that made old bodies develop cancer could be passed on to numerous offspring because the individual would reproduce before they got cancer.  On the other hand, a gene that made young adult bodies develop cancer would not be passed on to many offspring, and a gene that made young children develop fatal cancer would not be passed on to any offspring at all.  According to this theory then, senile decay is simply a by-product of the accumulation in the gene pool of late acting lethal and semi-lethal genes, which have been allowed to slip through the net of natural selection simply because they are late acting...

As an aside, one of the good features of this theory is that it leads us to some rather interesting speculations.  For instance it follows from it that if we wanted to increase the human life span, there are two general ways in which we could do it.  Firstly, we could ban reproduction before a certain age, say forty.  After some centuries of this the minimum age limit would be raised to fifty, and so on.  It is conceivable that human longevity could be pushed up to several centuries by this means.  I can not imagine that anyone would seriously want to institute such a policy.

Secondly, we could try to "fool" genes into thinking that the body they are sitting in is younger than it really is.  In practice this would mean identifying changes in the internal chemical environment of body that take place during aging.  Any of these could be "cues" that "turn on" late acting lethal genes.  By simulating the superficial chemical properties of a young body it might be possible to prevent the turning on of late acting deleterious genes.


I guess this would qualify as a kind of quasi-programming. It is not a biological clock, but wouldn't this suggest that natural selection does indeed play a part in deterioration later in life? Or do people here not buy into this theory?

(FF) Relationship between two or more theories

So my own pet theory at this point goes as follows:

After sexual maturity oxidation (damage) begins to negatively effect the body.

This results in aging.

Later in life, detrimental genes are activated as a result of this damage.

The activation of these genes result in the further deterioration of the body and are indicative of the later stages of aging.

If the individual still does not die, the eventual shortening of telomeres will finish them off.

#69 Lazarus Long

  • Life Member, Guardian
  • 8,116 posts
  • 242
  • Location:Northern, Western Hemisphere of Earth, Usually of late, New York

Posted 23 August 2003 - 02:55 PM

I have been reviewing this comprehensive article on aging that must be entered here for it addresses many of the same issues we have been. The author's should somehow be reached and invited to join our merry band as they are too well versed in the substantive investigation to be ignored.

I also think this paper written in '01 remarkably anticipates what is being done at Inner-Age as it actually addresses the question openly of how do we better measure "age"?

It is a 25 page PDF so I will only provide the link here but I strongly suggest downloading the full paper for review.

Plastic Omega - A Survey of Aging Research
Page 1. Held_Final 1 / 25 12/5/01 2:55 PM Plastic Omega Gene Held, FSA, MAAA
Vice President, Pricing SCOR Life US Re Insurance Co Addison, Texas Page 2. ...
http://www.soa.org/r.../Held_Final.PDF

Plastic Omega
Gene Held, FSA, MAAA
Vice President, Pricing
SCOR Life US Re Insurance Co
Addison, Texas
Held_Final 2 / 25 12/5/01 2:55 PM

Plastic Omega

A Survey of Aging Research


1. Abstract

The spin-off from the Human Genome Project is resulting in a rapidly escalating base of knowledge about life processes at their most fundamental level. Knowledge gleaned from that endeavor may offer the prospect of slowing the aging process. Dr. Francis Collins, Director of the National Human Genome Research Institute, has predicted, “By 2030, major genes responsible for the aging process in humans will likely have been identified, and clinical trials with drugs to retard the process may well be getting underway.” A growing number of scientists recognize extension of the maximum life span as a possibility.

Our profession cannot lay claim to expertise in the area of mortality while ignoring
important scientific research into the causes of aging. Otherwise, like generals preparing for the last war, we will ignore events that could destroy the assumptions underlying our projections of the future. This paper provides the actuary with a brief overview of the subject, along with references for those interested in conducting their own review.

#70 bacopa

  • Validating/Suspended
  • 2,223 posts
  • 159
  • Location:Boston

Posted 08 September 2003 - 01:22 AM

Excuse my ignorance, but doesn't it all boil down to slowing down the biological clock?

#71 Cyto

  • Guest
  • 1,096 posts
  • 1

Posted 08 September 2003 - 01:51 AM

Genes can fluctuate in how well they can take Chem, Rad, or Error problems and if they have a "backup" then your still ok, etc etc. I think this is called the Reliability Theory and coincides with nucleo-excision repair's impact on dramatic changes in how people can deal with X stresses (recent article on smoking and OGG1). Below is a good example of having little backups and if you don't get the protection you need, your messed.


Stuck on an aging mitochondria

The mitochondrial theory of aging suggests that DNA damage in the mitochondrial genome leads to dysfunction and production of reactive oxygen species implicated in the aging process. This DNA damage partly results from mutagenic base lesions in the form of 7,8-dihydro-8-oxoguanine (8-oxoG) and uracil. Cells normally utilize DNA glycosylases, such an 8-oxoguannine-DNA glycosylase (OGG1) and uracil-DNA glycosylase (UDG), in the first step of repairing such DNA damage. Aging produces high levels of 8-oxoG damage in mitochondrial DNA, but the levels and activity of OGG1 are higher in mitochondrial extracts from older rodents. In the September 1 PNAS, Bartosz Szczesny and colleagues at the University of Texas Medical Branch show that this apparent inconsistency is caused by a large fraction of OGG1 being caught in the mitochondrial membrane.

(Research method is in full text of article)

"Our results indicate an age-dependent decline in the mitochondrial import of proteins needed for DNA repair," conclude the authors. These findings add further weight to the theory that mitochondria are key in the process of aging.

Link to Article Page Link, heh

-------------------------------------------

So, I don't know if you mean entropy on the biological system or what. We do need to find out why the fluid-mosaic membrane of mito's have troubles with transportation. I, of course, think this also has something to do with maintaining the membrane trafficking gene array (ie: lipid, protein, glycosylated forms of the latter, ion channel fidelity.)

#72 Lazarus Long

  • Life Member, Guardian
  • 8,116 posts
  • 242
  • Location:Northern, Western Hemisphere of Earth, Usually of late, New York

Posted 12 September 2003 - 07:01 PM

I have listed the current page from Nature Magazine that features a Web Fous on Aging and the lins are very important on the page as there is a list of current studies consisely presented that we have nicluded in various topics individually.

Here is the link to that post and from there you can go to a series of published articels that will be expanding over the next few days but this is a very well organized begining of our focused database as well.

http://www.imminst.o...4&t=1762&hl=&s=

#73 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 14 September 2003 - 07:58 PM

Link: http://www.ncbi.nlm....6&dopt=Abstract
Date: 10-01-03
Author: Weinert BT, Timiras PS.
Source: Journal of Applied Physiology
Title: Invited Review: Theories of Aging
Comment: Here is a good overview that can be added to the many excellent posts here that can be used as a source in the study of the causes of aging.


J Appl Physiol. 2003 Oct;95(4):1706-16

Invited Review: Theories of aging.

Weinert BT, Timiras PS.

Dept. of Molecular and Cell Biology, 401 Barker Hall, Berkeley, CA 94720-3202. timiras@uclink4.berkeley.edu

Several factors (the lengthening of the average and, to a lesser extent, of the maximum human life span; the increase in percentage of elderly in the population and in the proportion of the national expenditure utilized by the elderly) have stimulated and continue to expand the study of aging. Recently, the view of aging as an extremely complex multifactorial process has replaced the earlier search for a distinct cause such as a single gene or the decline of a key body system. This minireview keeps in mind the multiplicity of mechanisms regulating aging; examines them at the molecular, cellular, and systemic levels; and explores the possibility of interactions at these three levels. The heterogeneity of the aging phenotype among individuals of the same species and differences in longevity among species underline the contribution of both genetic and environmental factors in shaping the life span. Thus, the presence of several trajectories of the life span, from incidence of disease and disability to absence of pathology and persistence of function, suggest that it is possible to experimentally (e.g., by calorie restriction) prolong functional plasticity and life span. In this minireview, several theories are identified only briefly; a few (evolutionary, gene regulation, cellular senescence, free radical, and neuro-endocrineimmuno theories) are discussed in more detail, at molecular, cellular, and systemic levels.

PMID: 12970376 [PubMed - in process]

Link to PDF

#74 Lazarus Long

  • Life Member, Guardian
  • 8,116 posts
  • 242
  • Location:Northern, Western Hemisphere of Earth, Usually of late, New York

Posted 23 October 2003 - 01:16 AM

Normally this article belongs in the section on genetics but it returns us to the debate about factors relating to a "biological clock". Admittedly this is a complex issue but here is support that there is a gene which acts as the reguator controlling the onset of puberty and it is related to secretions of the hypothalamus as we discussed at one point.

If someone can retrieve the full text from the New England Journal of Medicine it belongs in our archives please.

http://content.nejm....ort/349/17/1614

The GPR54 Gene as a Regulator of Puberty

ABSTRACT

Background Puberty, a complex biologic process involving sexual development, accelerated linear growth, and adrenal maturation, is initiated when gonadotropin-releasing hormone begins to be secreted by the hypothalamus. We conducted studies in humans and mice to identify the genetic factors that determine the onset of puberty.

Methods We used complementary genetic approaches in humans and in mice. A consanguineous family with members who lacked pubertal development (idiopathic hypogonadotropic hypogonadism) was examined for mutations in a candidate gene, GPR54, which encodes a G protein–coupled receptor. Functional differences between wild-type and mutant GPR54 were examined in vitro. In parallel, a Gpr54-deficient mouse model was created and phenotyped. Responsiveness to exogenous gonadotropin-releasing hormone was assessed in both the humans and the mice.

Results Affected patients in the index pedigree were homozygous for an L148S mutation in GPR54, and an unrelated proband with idiopathic hypogonadotropic hypogonadism was determined to have two separate mutations, R331X and X399R. The in vitro transfection of COS-7 cells with mutant constructs demonstrated a significantly decreased accumulation of inositol phosphate.

The patient carrying the compound heterozygous mutations (R331X and X399R) had attenuated secretion of endogenous gonadotropin-releasing hormone and a left-shifted dose–response curve for gonadotropin-releasing hormone as compared with six patients who had idiopathic hypogonadotropic hypogonadism without GPR54 mutations. The Gpr54–deficient mice had isolated hypogonadotropic hypogonadism (small testes in male mice and a delay in vaginal opening and an absence of follicular maturation in female mice), but they showed responsiveness to both exogenous gonadotropins and gonadotropin-releasing hormone and had normal levels of gonadotropin-releasing hormone in the hypothalamus.

Conclusions
Mutations in GPR54, a G protein–coupled receptor gene, cause autosomal recessive idiopathic hypogonadotropic hypogonadism in humans and mice, suggesting that this receptor is essential for normal gonadotropin-releasing hormone physiology and for puberty.

BBC Article --- Gene 'decides timing of puberty'

#75 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 20 November 2003 - 06:10 PM

FF) Relationship between Two or More Theories

A very interesting abstract that brings together the various components of aging theories under one whose main tenet is increased oxidative environment in the cell due to leaking mitochondria causes a chronic inflammation response.
------------------------------------------
J Theor Biol. 2003 Dec 21;225(4):531-40.


A unifying view of ageing and disease: the double-agent theory.

Lane N.

Department of Surgery, Royal Free and University College Medical School, Pond Street, NW3 2QG, London, UK

The quest for therapies based on molecular genetics (pharmacogenomics, DNA microarrays, etc.) drives pharmaceutical research into individual diseases of old age, but has failed to deliver an unequivocal clinical breakthrough. Attempts to treat most age-related diseases using antioxidant supplements have been equally disappointing, despite the clear benefits of a healthy diet. The double-agent theory is a new, unifying synthesis that draws on flaws in three leading theories of ageing. It argues that there is a tradeoff between oxidative stress as a critical redox signal that marshals genetic defences against physiological stress (such as infection) and oxidative stress as a cause of ageing and age-related disease.The stress response and ageing are linked by redox-sensitive transcription factors, such as NFkappaB. Ageing is a function of rising intracellular oxidative stress, rather than chronological time, but this relationship is obscured because free-radical leakage from mitochondria also tends to rise with age. Mitochondrial leakage produces a genetic response which mirrors that following infection, but because mitochondrial leakage is continuous the shift in gene expression is persistent, leading to the chronic inflammation characteristic of old age. Age-related diseases are thus the price we pay for redox control of stress-gene expression. Because the selective pressure favouring the stress response in youth is stronger than that penalising degenerative diseases after reproductive decline, we may be homeostatically refractory to antioxidant supplements that 'swamp' the redox switch. Furthermore, because genetic selection takes place predominantly in the reductive homeostatic environment of youth, alleles associated with age-related diseases are not inherently damaging (they do not inevitably express a negative effect over time), but are simply less effective in the oxidising conditions of old age. Gene therapies for age-related diseases are unlikely to succeed unless oxidative stress can be controlled physiologically, thereby altering the activity and function of potentially hundreds of genes.

PMID: 14615212 [PubMed - in process]

Link to PDF

#76 Mind

  • Life Member, Director, Moderator, Treasurer
  • 19,054 posts
  • 2,002
  • Location:Wausau, WI

Posted 29 February 2004 - 05:36 PM

(BB) Present a single theory leaning towards the PROGRAM theory

The Daily Times

Ageing mechanism linked to X-chromosome

Belgian scientists narrowed the search for a gene linked to ageing on Friday and said it is probably located on the X chromosome.

Researchers at the University of Leuven in Belgium pinpointed the location by studying the length of telomeres, tiny bits of DNA at the ends of chromosomes, linked to ageing. Similar to the plastic caps on shoe laces, they wear down as cells divide, which is a natural process of ageing. Shorter telomeres are thought to be associated with age-related illnesses and earlier death.

“We identified a possible genetic mechanism that interferes with longevity and the potential of ageing,” said Dr Jan Staessen, of the University of Leuven. “We think it is an X-linked phenomenon.”

Women have two X chromosomes while men have one X and one Y. The X chromosome is passed from father to daughter and from mother to son and daughter.

“It could means that the potential for longevity is a trait which is inherited from the mother,” Staessen, who reported his findings in The Lancet medical, explained.

He and his colleagues measured the telomere length of white blood cell DNA from people enrolled in a family-based study. They noticed that the lengths were similar between siblings and fathers and daughters and mothers and daughters and sons. But they were different between spouses and fathers and sons. “X-linked inheritance of telomere length is the most probable explanation of our findings,” Staessen added.

The results need to be confirmed with further studies and although the gene has not been identified the researchers believe they have a good candidate.

Earlier research by scientists in the United States showed that elderly people with longer telomeres lived five to six years longer than people with shorter ones.

If the identify of the gene is confirmed, Staessen and his team believe slight changes in it could affect telomere length and longevity.

Research into telomeres is still in its early phases but scientists believe that increased understanding about telomeres and telomerase, an enzyme that strengthens and lengthens them, will improve understanding of age-related diseases and the ageing process itself. —Reuters

#77 Mind

  • Life Member, Director, Moderator, Treasurer
  • 19,054 posts
  • 2,002
  • Location:Wausau, WI

Posted 29 February 2004 - 06:01 PM

(AA) Present a single DAMAGE - based theory

Children's Hospital Boston

Biochemical clues to long lifespan revealed
Findings extend longevity research from yeast and worms to mammals
BOSTON -- Researchers at Children's Hospital Boston have discovered how two key cellular influences on lifespan work together, providing insights that may help reveal aging mechanisms in humans. The findings extend longevity research from yeast and worms into mammals, and suggest that longer life results, at least in part, from biochemical interactions that boost cells' ability to resist environmental stresses while inhibiting them from committing suicide. The study appears in the February 19th Science Express, the online edition of the journal Science.
Previous studies in yeast and worms pinpointed a gene known as Sir2 as a key regulator of lifespan: deleting Sir2 limits lifespan, and extra copies lengthen it. Sir2 has a counterpart in mammals, but until now, very little was known about how it worked or what it had to do with aging. Working with mouse cells, researchers led by Anne Brunet, a postdoctoral fellow in neuroscience at Children's Hospital who is now at Stanford University, discovered that Sir2 works by regulating a group of proteins known as FOXO transcription factors. FOXO proteins have also been linked with longevity; they control the expression of genes that regulate cell suicide, and also enable the cell to resist oxidative stress, or chemical stresses that can disrupt the cell's DNA, or genetic blueprint.

"Aging involves damage to cells," says Dr. Michael E. Greenberg, director of Children's Program in Neurobiology and senior investigator on the study. "If you reduce oxidative stress, you get less aging."

The Children's team found that in the presence of oxidative stress, Sir2 promoted the ability of at least one FOXO protein, FOXO3, to provide stress resistance while suppressing its ability to induce cell death. In mammals, FOXO proteins confer stress resistance by triggering reactions that detoxify the damaging chemicals, known as free radicals. This leads to the repair of DNA damage while putting cell replication on hold, giving cells more time to perform the detoxification and repair process.



#78 kevin

  • Member, Guardian
  • 2,779 posts
  • 822

Posted 31 May 2004 - 07:10 AM

Sorry.. this post originally from "anothergod" and was moved inadvertently.. kevin

EE - Evolution and Aging
I am sorry that I only just found this thread, I should have been reading this from the begining, but better late than never. I haven't read all of the posts yet, but I did notice some posts on this topic which I certainly have to read. I will do so, but it will take time.

As far as I knew, the most prevalent theory of 'why' we age has been covered reasonably well. I know of at least 3 papers which essentially cover the details of it, and which in my mind do so very well in a complimentary way. The three papers are:

1. Medawar PB. 1952. An Unsolved Problem of Biology. Lewis, London
2. Williams GC. 1957. Pleiotropy, Natural Selection, and the Evolution of Senescence. Evolution 11:398-411
3. Kirkwood TBL. 1977. Evolution of Aging. Nature 270:301-304 and/or
Kirkwood TBL. 2002. Evolution of Ageing. Mechanisms of Ageing and Development 123:737-745

Medawar proposes that age is a result of a declining selective pressure as an organism 'ages' (ie: as time passes). Williams then takes it a step further and connects the less vigourous actions of Selection with Pleiotropy, proposing that genes which confer an early advantage but a later disadvantage will still be selected. These later side-effects will be dealt with by selection later as required. Kirkwood then builds on this by proposing his 'Disposable Soma Theory' which says more of the same, but from a different sort of perspective.

In my opinion, Williams hit the nail on the head in 1957. I am posting this partially hoping to be corrected on this point, but since it hasn't happened yet, I firmly believe that Williams' theory explains why we age perfectly, and now all we need to do is apply its logic to the 'how' and solve it.


What Williams Said:
First of all, it makes no sense of all to propose that biological organisms 'decay' over time: they are not static objects, they are dynamic systems in constant exchange with their environment. More importantly, they all produce themselves from morphogenetic processes: Any system which can form itself can surely maintain itself.

Assuming a world where there was no such thing as senescence, death would be determined by accident, predation, starvation etc. As time passes the probability of succumbing to one of these causes of death accumulates. Eventually you would die. For example: In a population of critters in this theoretical world, a graph of their percentage of survival over time would decline as periodically one and then another and then another dies from one incident or another. Evolution would have a keen interest in making their reproductive age earlier, or somehow making the organism more robust so as to survive to reproductive age. Evolution can do little about making them all immune to accident, starvation etc though.

"This would produce a decline in reproductive probability"Senescnce might be regarded as a group of adaptively unfavourable morphogenetic changes that were brought in as side effects of otherwise favourable genes, and which have only been partly expurgated by further selection." (WIllimas 1957)

And that is essentially the theory. Reading the paper itself is well worth it. I beleive that Oxidative Damage theory of aging is a perfect example of antagonistic Pleiotropy, The advantage of being an Aerobic organism beign huge, but the gradual accumulation of damage to important structures being a long term negative. Over time evolution has fought to restrict and stop this damage by using DNA repairs mechanisms, proteins recyclers, free radical scavengers etc. It has not been able to stop the damage, but it has dulled it down to a background level that allows us a life span perfectly suited to our pre-historic lifestyle.

#79 olaf.larsson

  • Guest
  • 583 posts
  • 21
  • Location:Sweden

Posted 15 June 2004 - 01:04 PM

What do you say about this?

Aging is a Specific Biological Function Rather than the Result of a Disorder in Complex Living Systems.

http://www.protein.b...ll/62111394.htm

If aging is induced by our own bodies we are betrayed by our own genes; they kill us to propagete themselves better, but if it is so it could be more easy to stop.

#80

  • Lurker
  • 1

Posted 21 June 2004 - 02:48 PM

Aging is a Specific Biological Function Rather than the Result of a Disorder in Complex Living Systems.


I agree. I believe that complex living systems cannot be overwhelmed by disorder, otherwise they would eventually all perish. I believe that they actually use disorder to create variation.

#81

  • Lurker
  • 1

Posted 04 July 2004 - 01:37 PM

Furthermore, aging is a not a specific biological function but rather a consequence of deliberately inefficient DNA repair that enables mutation and thereby evolution to take place.

#82 apocalypse

  • Guest
  • 134 posts
  • 0
  • Location:Diamond sphere

Posted 15 August 2004 - 04:24 AM

Following on the ideas of aging as an adaption that favors evolution, here are some responses to prior comments.

There is no reason for an aging program to exist, because humans did not live long enough for the processes we call aging to come into play until very recently in the evolutionary time scale, so there was no selective pressure to create it. In addition, all the molecular and cellular evidence supports this line of thinking.

Birds, bats, and tortoises are interesting because they all have extremely robust anti-oxidant defense systems which is probably tied to their developmental timescale.

It is not that anything is stopping the increase of lifespans it is just that there is often a great advantage to rapidly get to breeding age, which is tied to shorter lifespans.

The key idea about pressure for increased lifespan is that the pressure isn't on aging itself, but it is on the reproductive period of a species. There definitely is selection for longer time of fertility in species with low environmental stress, but long life after the reproductive period is most likely just a nice side effect (i.e. increased defense mechanisms which continue to function even past their evolutionary usefulness). Evolution just doesn't care about us once we have left successful offspring, and there doesn't seem to be any strong reason to get rid of us either. The pressure against longer lifespans (and for fast breeding times) in humans really is just about nonexistent.


Hmmm... Yet some organisms reproduce at a particular age but do not age until way after, while others age quite rapidly after rerproducing. There are also the negligible senescense species. The existence of species were time of reproduction is decoupled from lifespan, is one that conflicts with this.

Finally, although it's true that reproduction and life span are deeply correlated, there are numerous exceptions (Finch, 1990, 547-550; Le Bourg, 2001).


There are also species were the member of one sex ages and the other seems not to age. How could this be if they've both practically the same genome? Why is one exponentially longer lived, seemingly negligible senescent while the other is not?
How can closely related species, same genus, be one short lived, the other negligible senescent, how can even members of the same species be negligible senescent and some not? How can age of sexual maturity and lifespan be decoupled in some species?

How can the c-elegans enter a dauer like state, while retaining it's reproductive capability, and extreme lifespan extension? How can it be that some females restart menstruation after a short period of CR(which is later discontinued) post menopause?

Over the past few months, Kenyon's team and several other groups of worm researchers have documented an unexpectedly large number of genes controlled by this hormonal system, including genes involved in stress responses and antimicrobial actions.- august, 2003

aging genes

Long before Kenyon's work, other researchers linked daf-2 and daf-16 to this arrested form of development. The genes' names derive from "dauer formation." Completely knocking out the activity of daf-2 sends a developing worm right into the dauer state, whether or not nutrients are scarce. Kenyon found something more intriguing: Certain subtle mutations in the gene enabled a developing worm to bypass the dauer state but still have an abnormally long life span.

How can this be?(note an abnormal increase in lifespan by altering regulation/expression of existing mechanisms not by addition of massive novel protective mechanisms)

On the issue of the tortoise, the reason why didn't develop mechanisms for longevity like some more primitive species is because these species never stop growing.


There are species that stop growing yet show negligible senescense, IIRC.

The body knows, as you said, when it is in puberty. It knows when to go through menopause (though that may be simply triggered by the end of Eggs.) It knows when to harden your bones (from the softer, more numerous Baby bones), it knows when to make u grow. It knows when to make u stop growing. It knows when to get hairy.


Women continue to produce eggs throughout their lives, it obviously knows when to stop doing so. Other females after experiencing CR post menopause, and returning to a regular diet start menstruating again. How can this be?

Although I think it is likely that senescence has genetic controls, I think that the death of an individual is a combination of damage and program in a kind of one two punch. It may be that with all the genetic controls that increase senescence turned off that we would still accumulate damage that would eventually cause systems to fail and death. I think it is possible for the two theories to coexist and in fact be linked.

I think the program, if there is one, works by throwing wrenches into the gears, altering genetic expression slowly into a diseased state esque one. If this were taken care of negligible senescense might arise, thus exponential lifespan differences in close species and in members of the same species would be possible, as observed. That is repair/maintainance/etc might actually be downregulated or altered in some controlled way, this would cause deterioration and increased rates of diseases, cancer, etc.

We did see changes that look like the changes that you see in the development of age-related diseases. This has been found by other workers conducting micro-array gene expression studies in other tissues as well. I think our results are very consistent with theirs in showing that gene expression profiles in tissues begin to resemble profiles of tissues that have age related disease processes going on in them. Our tissues looked healthy-we could slice them and look at them under the microscope and see no signs of liver fibrosis for instance. But when we looked at gene expression in these tissues with age, we found changes that more and more resemble those that you see in diseased tissues. So, I think that's part of the development of age-related diseases-a drift towards gene expression that resembles the gene expression of diseased tissues. Calorie restriction reverses much of that, short and long-term.

Also, older organisms with high genetic fitness would still be more desirable for a population than less fit younger ones.

Less fit, non-aging organisms could outcompete fitter but very young organism, causing a detrimental effect to the species, and thus compromising long term survival, fitter ageless ones would monopolize resources, reducing variety by starving young ones who're less fit. Aging begins its effect from early on(some organisms appear to show first signs of it very near, even slightly prior to sexual maturity, this negative effect should be selected out unless it proved beneficial) thus its effect is non-negligible in survival from a young age, it's a negative influence from EARLY ON. Prometheus post, shows examples of how causing negligible senescence might slow evolution itself, compromising species survival.

All the data seems to be pointing to the fact that telomeres are protective, but the enzymes that regenerate them are turned off in an effort by complex organisms to stave off cancer, not as an aging mechanism

Yet negligible senescence species have them always turned on, and they seem to have a low incidence of cancer...

Even telomerase expression, the hallmark of immortal cells, has been found at extraordinary high levels in all the cells of
negligibly aging animals )(Klapper, et al, 1998a, 1998b).

How is that explained?

Edited by apocalypse, 16 August 2004 - 06:13 PM.


#83

  • Lurker
  • 1

Posted 15 August 2004 - 06:53 AM

Women continue to produce eggs throughout their lives, it obviously knows when to stop doing so.  Other females after experiencing CR post menopause, and returning to a regular diet start menstruating again.  How can this be?


It suggests that the CR mechanism can induce a fundamental state change in the cell. You have observed one of the patterns of CR being that it can be activated at any time which can be interpreted differently based on your level of consideration:
1. species specific - we are not designed to consume so many carbohydrates and calories. We are meant to endure periods of starvation. Our current nutritional abundance is toxic. When we curtail our caloric consumption we enter into the 'health zone' which is marked by lower incidence of disease.
2. all species - the metabolic by-products required to produce ATP are inherently toxic, accumulating and overwhelming the cell's capability to neutralize them. Any activity that down-modulates the production of toxic by-products or increases the capability to neutralize them allows the cell to survive longer.

I think the program, if there is one, works by throwing wrenches into the gears, altering genetic expression slowly into a diseased state esque one.  If this were taken care of negligible senescense might arise, thus exponential lifespan differences in close species and in members of the same species would be possible, as observed. That is repair/maintainance/etc might actually be downregulated in some controlled way, this would cause deterioration and increased rates of diseases, cancer, etc.


I think you're right. There are two interesting questions that arise from such an observation of a 'deliberate spanner' in the works:
1. What is the mechanism?
2. How such a mechanism be brought to fit into a "unified theory of evolution" that links aging and disease with evolution?

My favorite for (1) is rather simple to arrive at providing one looks for a pattern in the downstream effect of all longevity related genes studied to date. Their function is always associated with preserving DNA structural integrity. This could be either by increasing DNA repair factors, increasing anti-oxidant production, decreasing oxidant production, etc. Always, though, related to maintaining DNA integrity. In fact if we look at genetic disorders of accelerated aging such as Werner's , guess what - related to DNA integrity. If we look at cancer - yep DNA integrity again. But what about cell death - apoptosis is mainly triggered by damage to DNA. How about cell senescence - yep, triggered by chromosomal instability. So we see a profoundly unmistakable pattern emerge: DNA damage = aging & disease = death.

So how could all this relate to evolution and an answer for (2)? The hint comes from what DNA damage implies. Each time DNA is damaged the cell desperately attempts to repair the damage. It has had millions and millions of years of evolution to perfect strategies to repair it's precious DNA cargo from the likes of radiation and other environmental mutagens including pesky DNA inserting viruses and even its own toxic by-products. You would think that the cell has an arsenal of DNA repair tools by now. You're right. There are over one hundred human DNA repair genes that have been identified to date. So DNA repair is pretty serious business. But back to evolution, DNA damage - if it does not result in the destruction of the molecule - results in a mutation! And that's the key, because: DNA damage = mutation. We know that mutation is the very driver of evolution. It's what keeps our genome plastic enough to be able to adapt. Without mutation life would not be able to evolve.

So every cell treads a fine line between keeping the genome stable and unstable. If it makes it's DNA too protected it sentences the cell to evolutionary stasis and eventual extinction. If its DNA is too unstable - well we know the effects of that.

So there you have it:

aging/disease = DNA damage = evolution

We succumb to aging and disease because we must.

Yet negligible senescence species have them always turned on, and they seem to have a low incidence of cancer...

Even telomerase expression, the hallmark of immortal cells, has been found at extraordinary high levels in all the cells of
negligibly aging animals )(Klapper, et al, 1998a, 1998b).

How is that explained?


This is an easy one. It is called laziness - or throw the baby out with the bathwater. In their feverish quest to identify a biomarker for tumorigenic cells, scientists have grasped at a commonly observable (and easily assayable) denominator - the presence of telomerase reverse transcriptase (TERT) activity. Unfortunately other TERT positive cells include stem cells and germline cells. TERT expression does not equal cancer, but some scientists are so hell bent on on following this path that they actually have devised ways of targeting all TERT positive cells. Clinicians of course, have been targeting all TERT positive cells for years, without realizing it, via the treatments of chemotherapy and radiotherapy.

The astute cell biologist knows that there are other more salient markers on the cell surface that enable a cancer cell to be identified. :)

#84 apocalypse

  • Guest
  • 134 posts
  • 0
  • Location:Diamond sphere

Posted 15 August 2004 - 02:17 PM

aging/disease = DNA damage = evolution

We succumb to aging and disease because we must.


I believe this is so, but I'm not entirely sure If we've come to a complete agreement. My belief is that it seems that the organism is not simply allowing this to take place at a constant rate, but that it's purposely accelerating the rate of dna damage that takes place and slowing maintainance/repair. These changes after reproduction maturity would yield more mutation and allow for increased evolution even of sections vital to the organism's survival without compromising it's function to reproduce by allowing these to take place from a very early age, it would also take out competition from fully developed organisms that might outcompete fitter but undeveloped members of the species, and families of fittest organisms that would monopolize resources and yield inbred populations unable to adapt to sudden change.

Why?

Note the changes are eluded in some of the above examples(prior post), and many can be reversed even when they take place with CR, with positive benefits as a result, showing that they're not inevitable. Why are detrimental reversible(CR, alternative fasting with double consumption on alternate days, cell/tissue rejuvanation, cloning, etc) or eludible(c-elegans non-dauer abnormal lifespan, members of a particular sex in some species being seemingly negligible senescent, while the other is not) changes taking place? Why are cells giving up for some but under some identical circumstances/enviroment for others not(non-dauer abnormal lifespan state v.s. regular state or for particular members of a species but not for others) ?

Some say that in times of scarcity there may be problems for the ageless organism(in addition to the evolution problem), based on experiments from mutants. But that's probably due to the rustic simple mutations that are done that keep maintainance up in a non-carefully regulated manner, allowing them to take place even while enduring famine. But just like CR can increase repair in a regulated manner while in famine, an advanced ageless organism could decrease it to CR or even slightly lower levels(they've been better maintained) a temporarily to withstand famine, and retain this for a short while following without being outcompeted, in fact due to its youthfullness it should outcompete others.

Any activity that down-modulates the production of toxic by-products or increases the capability to neutralize them allows the cell to survive longer.


Yes, and if the cell purposely began diminishing such activity it would begin an accelerated decline, showing an exponential increase in mortality, and an exponential rise in cancer incidence that is seen with aging.

Because the telomerase activity in stem cells does not prevent
telomere shortening

stem cells
Even though telomerase positive the stem cell will lose it's telomere length as time goes on, it also becomes less and less effective with time for some reason. Recent research suggest that stress -say against cardiovascular system-, high cholesterol/overweight, smoking, etc, accelerates this and seems to deplete repair capacity sooner. Older stem cells seem less capable than younger ones. Those individuals with longer average telomeres, seem to live longer. Cancer though telomerase positive seems to keep telomeres quite short, from what I've heard.

Telomere length in these cancer cells is extremely short, about 2-4 kb
vs approximately 20 kb in stem cells and embryonic cells.

short telos cancer

stem cell through the different ages

A form of adult stem cells called endothelial progenitor cells in the blood are inversely correlated with arterial damage that leads to heart disease.

stem cells age

Bone Marrow Stem Cell Aging Key In Atherosclerosis

aging stem cells

"But if you compare someone who is over 60 with someone who is 20 with the same risk factors, there is obviously something else going on as well," he continued. "The possibility that stem cells may be involved is a completely new piece of the puzzle that had not been anticipated or appreciated before. These findings could be the clue to help us explain why atherosclerosis complications like heart attacks and strokes are almost exclusively diseases of older people."

"We found that the bone marrow cells from the young mice had a nearly intact ability to prevent atherosclerosis, while the cells from the older mice did not," Goldschmidt explained. "This finding suggests that with aging, cells capable of preventing atherosclerosis that are normally present in the bone marrow became deficient in the older mice that had developed atherosclerosis."


Limitting the maximum repair/maintainance potential of the body, would be one way to accelerate deterioration later on purposely when this limit is reached.

The study looked at the blood level of endothelial progenitor cells, which are made in the bone marrow and may help the body repair damage to blood vessels. Scientists from NHLBI and Emory University Hospital in Atlanta, GA, found that cardiovascular disease risk was higher in persons with fewer endothelial progenitor cells. The cells of those at higher risk also aged faster than those at lower risk, as determined by the Framingham Heart Study risk factor score, a standard measurement of cardiovascular risk. Additionally, the study found that blood vessels were much less likely to dilate and relax appropriately in persons with low levels of the cells.

limited repair and maintainance

This is an interesting twist. If telomere shortening was causing the stem cells to become senescent then one wouldn't expect a drug that lowers cholesterol to boost the levels of these cells.



While this latest report may demonstrate an advantage of stem cells created by cloning it doesn't answer the question of why those cells are more vigorous. If the report really did find that adult skin fibroblast cells, when cloned, became stem cells that were more vigorous than existing adult stem cells in the same animal then why?

cloned stem cells stronger than uncloned

While Randall shows examples of possible reasons for this being so, I believe it might be that the animal itself has its repair/maintainance mechanisms downregulated, causing not just effects at a cellular level's repair/maintainance but in turn at an organismal level. The fact that these negative changes are reversed/eluded by some means, as mentioned, seems to indicate they're purposely being done.

#85 apocalypse

  • Guest
  • 134 posts
  • 0
  • Location:Diamond sphere

Posted 16 August 2004 - 02:45 AM

Continuing ideas of previous post:
(note the following are quotes and links from a recompilation of relevant studies/data from several journals/sources, from a particular site)

When the tiny worm Caenorhabditis elegans cannot sense what is going on in its immediate surroundings, there is a surprising payoff. It lives up to twice as long as normal, according to Javier Apted and Cynthia Kenyon of the University of California at San Francisco. They have found that 'nematode' worms that have defective sensory organs can double their lifespan, without any apparent change in behaviour. This suggests that, in nematodes at least, longevity is not simply programmed into the genes but can be altered by feedback through the nervous system. The role of nervous system in aging could not be remarked upon*.

C-elegans

Healthy centenarians
Note again(as seen in my post on the prize thread) that the difference between the healthy centenarian and the average aging adult, are akin to those of CR.

In primary care practice, it is not unusual to encounter male patients in their 50s or older who report having loss of libido, erectile dysfunction, fatigue, and depression. Such signs and symptoms may signal an age-related decline in androgen levels, which commonly begin after age 40. The term andropause refers to a state of lowered androgen levels. Androgens are a group of hormones that include testosterone, dehydroepiandrosterone, and androstenedione, among others. There is undeniable evidence that aging results in a lowering of androgen levels.

Testosterone is produced by fetal testes as early as the first trimester. Levels in adult males peak at about age 20 years and gradually decline thereafter. Total testosterone declines at the rate of 3.82 nmol/L (110 ng/dL) per year, typically after age 40. Levels of bioavailable testosterone decline much more dramatically.

Because the decline in androgens is gradual, the term androgen decline in aging males (ADAM) has been used to describe this phenomenon. Alternatively, partial androgen decline in aging males has also been suggested because the androgen deficiency in older men is generally moderate and not a complete deficiency. Symptomatic hypogonadism is sometimes referred to as the andropause syndrome.

andropause

Note dramatic sudden decline after a particular age.

The finding that telomeres, the nucleoprotein structures protecting chromosome ends, shorten with every cycle of cell growth and division suggested a molecular mechanism that could record the number of divisions that a lineage of cells has undergone. Erosion of telomeres to a critical length could serve to activate the senescence program. In accord with this mechanistic model, ectopic expression of the catalytic subunit of the telomerase enzyme, hTERT (human telomerase reverse transcriptase), halts the erosion of telomeres in human cells; in some cell types the expression of this gene prevents the entrance into replicative senescence, suggesting that indeed telomere shortening is the cause for senescence.
Normal cells that are exposed to various physiologic stresses rapidly enter into a state of senescence, doing so within a period as short as several days. Such stresses include DNA-damaging agents, oxidative stress, "oncogenic stress" (due to oncogene overexpression), and other metabolic perturbations. Typically, these forms of senescence do not involve significant telomere shortening and cannot be prevented by ectopic hTERT expression. Accordingly, the hypothesized telomere-based mechanism of division counting could not be invoked to explain these acute responses. Moreover, a model proposing that senescence functions exclusively as a barrier to extended growth-and-division cycles could not accommodate these situations of stress-induced senescence.


Clearly, a stress-based clock contributes to the effect of the telomere-based clock in these cells, and the combined effects of the two dictate the onset time of senescence.

This recent study also underscores a little-regarded observation, namely that many normal human fibroblast lines cannot be immortalized by ectopic expression of hTERT. Telomerase activity enables these cells to proliferate longer, but eventually they do undergo senescence, even though their telomeres have been elongated well beyond the lengths observed in early passage cells. Thus, the immortalizing capabilities of telomerase are only limited to a subset of cell types.


This seems interesting.

Yet another recent study indicates that the endogenous telomerase enzyme, operating in presenescent normal cells, in fact performs maintenance of proper telomere structure. It has long been believed that most normal human cells do not express the hTERT gene and therefore do not possess telomerase activity. This recent work has shown, using sensitive detection methods, that hTERT is in fact expressed and active in normal human fibroblasts. This expression is transient, appearing only during S-phase. When this transient endogenous hTERT expression was eliminated in these cells, the cells underwent premature replicative senescence. Strikingly, the rate of overall telomere shortening did not change in these cells. Instead, their telomeric overhangs eroded more rapidly than did those of control cells, so that fibroblasts entering senescence carried significantly eroded overhangs, whereas control cells that underwent the same number of divisions carried overhangs of normal length.


If this is correct It seems that telomerase might be active in regular tissues, just harder to detect for the mentioned reasons, and it's function and nature and that of telomeres might be slightly different from what's previously believed.

telomere and telomerase more complex functions

The following data suggest that cancer itself too has some suggestive behavior.

Several intriguing implications derive from the properties of the SR/CR mouse. First, this model demonstrates the existence of a host resistance gene that can prevent the growth of advanced, MHC-negative cancers. The existence of host cancer resistance genes has been postulated to be one explanation for the existence of individuals in the human population who fail to develop cancers, despite prolonged and intense exposure to carcinogens. The gene(s) responsible for the SR/CR phenotype may well be an example of such a resistance gene that might have a direct human ortholog. Second, the concept of immune surveillance has been debated for decades and has been difficult to prove, although recent studies have lent support to this concept. The SR/CR mouse may also provide a potential example of such a surveillance mechanism. Third, the alteration in the type of response seen with age in these mice suggests an intriguing possibility. The appearance of cancer in older individuals at a much higher frequency may not solely be caused by the accumulation of mutations in individual preneoplastic cells. This mouse model suggests that there may also be host resistance mechanisms that decline with age. Fourth, the rare phenomenon of SR of cancers has been documented in humans, but has been difficult to study because of a lack of an appropriate animal model. The SR/CR mouse may provide such a model and allow identification of the cellular and genetic machinery necessary to reject a fully developed malignancy. The ability of adoptively transferred infiltrating leukocytes from SR/CR mice to protect control mice from S180 cells may suggest a potentially feasible strategy for treatment of advanced cancers that could be translatable into human patients.

intriguing behavior of cancer and host resistance/immunity with age

Cancer even in some of its most malignant states/forms seems beatable by some organisms, this ability seems to be present even in aging species and does not shorten lifespan, but is removed at a late age... maybe it too...

#86 apocalypse

  • Guest
  • 134 posts
  • 0
  • Location:Diamond sphere

Posted 16 August 2004 - 11:34 AM

Some more thoughts:

Reduced expression of genes discussed above[in linked article-apoc] indicates that there is a general loss of negative cell growth control with age. Seventy-eight percent of the mice of this strain and sex fed the control diet used here die of some form of neoplasia, and the death rate from neoplasia accelerates dramatically with age. Approximately 21% of these mice die of hepatoma, mostly late in life. Decreased expression of the negative growth regulators and overexpression of the chaperone genes with age also are consistent with this higher incidence of hepatoma in aged mice.

Aging decreased expression of a second group of genes with antineoplastic potential, xenobiotic metabolism genes. These genes were negatively regulated by age. Decreased expression of such genes is likely responsible in part for the age-related decline in the xenobiotic-metabolizing capacity of the liver. This decline is a recognized source of adverse drug reactions in aged mammals. It may contribute to the increase in neoplasms with age in mice.


Thus, CR rapidly reversed, rather than prevented, many age-related changes in gene expression. ST-CR induced a more youthful gene expression profile associated with longer life and health span.

aging, gene expression, CR

Note several cancer promoting changes, reversible changes...

It is known that senescent human fibroblasts stimulated hyperproliferation and progression of preneoplastic epithelial cells and accelerated tumorigenesis by neoplastic epithelial cells. These results may seem at odds with the tumor suppression function of cellular senescence.

Senescent human fibroblasts stimulate premalignant and malignant, but not normal, epithelial cells to proliferate in culture and form tumors in mice. In culture, the growth stimulation was evident when senescent cells comprised only 10% of the fibroblast population and was equally robust whether senescence was induced by replicative exhaustion, oncogenic RAS, p14ARF, or hydrogen peroxide. Moreover, it was due at least in part to soluble and insoluble factors secreted by senescent cells. In mice, senescent, much more than presenescent, fibroblasts caused premalignant and malignant epithelial cells to form tumors. It could be suggested that, although cellular senescence suppresses tumorigenesis early in life, it may promote cancer in aged organisms....

promote cancer

While this article posits, antagonistic pleiotropy, as a possible explanation, I believe in context with the other data I've posted, it too indicates purpose. In sum it seems cancer promoting changes take place inside the cell, senescent cells promote those malignant/pre-malignant cells to proliferate, and the immune response is impaired at later ages, allowing even resistant organisms to succumb to such.

#87

  • Lurker
  • 1

Posted 17 August 2004 - 01:21 AM

As a side note because you mentioned declining testosterone levels and aging and in support of your deliberate wrench theory, a paradoxical effect in aging males is the rate of androgen related hair loss which increases even though testosterone levels are declining suggesting an increased sensitivity to testosterone. As blocking the action of testosterone decreases hair loss rate it suggests that an increase in the expression of androgen receptor on the hair follicle out of proportion with the decrease in testosterone is occurring.

Such patterns in gene regulation are not just isolated to androgen related hair loss. Increased or decreased sensitivity, due to abnormal receptor expression, could well be promulgating the aging phenomenon throughout the organism, as one aging cell signals another to follow. Consequently due to a 'deliberate' but abnormal interpretation of its environment the cell is forced to ultimately follow the senescent or apoptotic pathway. These changes, as you said, begin at a very early age.

#88 chubtoad

  • Life Member
  • 976 posts
  • 5
  • Location:Illinois

Posted 20 August 2004 - 07:53 PM

http://www.scienceda...40816002236.htm

A First Glance At The Gene Networks Of Human Aging

Boston, MA – July 29, 2004 – Scientists have rendered the first gene and protein networks of human aging, an important step in understanding the genetic mechanisms of aging. The work led by Joao Pedro de Magalhaes from Harvard Medical School is detailed in the July 30 issue of FEBS Letters.

The work involved the integration of all genes, in both humans and animal models, previously shown to influence aging. By using a combination of bibliographic information and modern high-throughput genomics, employing software developed by the team, each gene was placed in the context of human biology. The putative impact of each gene to human aging was calculated by a combination of manual and computational methods, leading to a new holistic view of the genetics of aging. To organize and catalog all the data pertaining the over 200 genes selected, the first curated database of genes related to human aging was developed: GenAge, part of the Human Ageing Genomic Resources also led by Dr. de Magalhaes. Thanks to the help of many other researchers, the Human Ageing Genomic Resources have become, in months, the landmark online website for research on aging, having been recently featured in Nature Reviews Genetics (volume 5, issue 5, page 330) and SAGE KE (2004 volume, issue 30, nf69), Science Magazine’s website on aging research.

With the collaboration of researchers from the University of Namur in Belgium, scientists also analyzed protein-interaction maps for more specialized pathways previously linked with aging, such as the neuroendocrine regulation of aging and DNA metabolism. These findings and the rendered networks related to aging may prove useful to find novel genes of interest. In fact, several crucial nodes in the networks were identified by way of specialized software: a number of genes so far not linked to aging were chosen by a “guilt-by-association” methodology based on protein-protein interaction maps and data-mining algorithms. One intriguing finding was the apparent overlap between the genetics of aging and development. Aging could then be an indirect result of developmental pathways. The cascade of events that regulates ontogeny would then fade away after sexual maturity resulting in aging. Contrary to other theories of aging that argue aging derives from the accumulation of damage, Dr. de Magalhaes suggests that integrative pathways collaborate during development and then become disrupted during aging.

The GenAge database will be in constant development since several genes involved in aging are certainly yet to be identified. Even so, it provides an overview of the current state-of-the-art knowledge on human aging. While much works remains to fully understand aging, this is a major first step in understanding the genetic mechanisms of human aging and provides a framework for future experimental verification.



#89 apocalypse

  • Guest
  • 134 posts
  • 0
  • Location:Diamond sphere

Posted 26 September 2004 - 02:52 PM

As a side note because you mentioned declining testosterone levels and aging and in support of your deliberate wrench theory, a paradoxical effect in aging males is the rate of androgen related hair loss which increases even though testosterone levels are declining suggesting an increased sensitivity to testosterone. As blocking the action of testosterone decreases hair loss rate it suggests that an increase in the expression of androgen receptor on the hair follicle out of proportion with the decrease in testosterone is occurring.

Such patterns in gene regulation are not just isolated to androgen related hair loss. Increased or decreased sensitivity, due to abnormal receptor expression, could well be promulgating the aging phenomenon throughout the organism, as one aging cell signals another to follow. Consequently due to a 'deliberate' but abnormal interpretation of its environment the cell is forced to ultimately follow the senescent or apoptotic pathway. These changes, as you said, begin at a very early age. -prometheus

"The cascade of events that regulates ontogeny would then fade away after sexual maturity resulting in aging. Contrary to other theories of aging that argue aging derives from the accumulation of damage, Dr. de Magalhaes suggests that integrative pathways collaborate during development and then become disrupted during aging."-sciencedaily

-chubtoad


I've heard of something that could be considered for the disrupting factor, hervs(human endogenous retroviruses) and other mobile elements. They're said to compose between as little as 1-3% up to 8% of the genes, some've said that up to a quarter of the genome is made of hervs and herv related sequences. While supposedly initially inactive, I've read that they may be activated by a myriad of factors, including toxins, viral infections, radiation, etc. IIRC some of'em have been linked to cancer dev., immune system weakening, hormonal changes, changes in gene regulation, and other things . Yet, I believe more information on these is needed to know whether they're what they seem or not, but given the ever greater exposure to environmental factors that may activate them as time goes on, one would guess their activity and effects if any are only likely to increase with time...
.
And another article with more on aging as an adaptation:
http://www.betterhum...ID=2004-09-27-3

Edited by apocalypse, 22 February 2005 - 04:08 PM.


Click HERE to rent this BIOSCIENCE adspot to support LongeCity (this will replace the google ad above).

#90 olaf.larsson

  • Guest
  • 583 posts
  • 21
  • Location:Sweden

Posted 22 November 2004 - 11:22 AM

Considering the salomon which seem to have a inbuild death program which makes it die soon after sex. I suspect that all the accumulation of dead salomon biomass make varius microorganisms grow in the water which the ofspring of the salomon can later eat. Thus death after sex is an evolutionary advantage. Salomons which would not dont die in the place of sex have almost no chance of mating a second time anyway.




0 user(s) are reading this topic

0 members, 0 guests, 0 anonymous users