header logo image


Page 14«..10..13141516..»

Archive for the ‘Longevity Genetics’ Category

Genetics of aging – Wikipedia

Wednesday, September 26th, 2018

Genetics of aging is generally concerned with life extension associated with genetic alterations, rather than with accelerated aging diseases leading to reduction in lifespan.

The first mutation found to increase longevity in an animal was the age-1 gene in Caenorhabditis elegans. Michael Klass discovered that lifespan of C.elegans could be altered by mutations, but Klass believed that the effect was due to reduced food consumption (calorie restriction).[1] Thomas Johnson later showed that life extension of up to 65% was due to the mutation itself rather than due to calorie restriction,[2] and he named the gene age-1 in the expectation that other genes that control aging would be found. The age-1 gene encodes the catalytic subunit of class-I phosphatidylinositol 3-kinase(PI3K).

A decade after Johnson's discovery daf-2, one of the two genes that are essential for dauer larva formation,[3] was shown by Cynthia Kenyon to double C.elegans lifespan.[4] Kenyon showed that the daf-2 mutants, which would form dauers above 25C (298K; 77F) would bypass the dauer state below 20C (293K; 68F) with a doubling of lifespan.[4] Prior to Kenyon's study it was commonly believed that lifespan could only be increased at the cost of a loss of reproductive capacity, but Kenyon's nematodes maintained youthful reproductive capacity as well as extended youth in general. Subsequent genetic modification (PI3K-null mutation) to C.elegans was shown to extend maximum life span tenfold.[5][6]

Genetic modifications in other species have not achieved as great a lifespan extension as have been seen for C.elegans. Drosophila melanogaster lifespan has been doubled.[7] Genetic mutations in mice can increase maximum lifespan to 1.5times normal, and up to 1.7times normal when combined with calorie restriction.[8]

In yeast, NAD+-dependent histone deacetylase Sir2 is required for genomic silencing at three loci: the yeast mating loci, the telomeres and the ribosomal DNA (rDNA). In some species of yeast, replicative aging may be partially caused by homologous recombination between rDNA repeats; excision of rDNA repeats results in the formation of extrachromosomal rDNA circles (ERCs). These ERCs replicate and preferentially segregate to the mother cell during cell division, and are believed to result in cellular senescence by titrating away (competing for) essential nuclear factors. ERCs have not been observed in other species (nor even all strains of the same yeast species) of yeast (which also display replicative senescence), and ERCs are not believed to contribute to aging in higher organisms such as humans (they have not been shown to accumulate in mammals in a similar manner to yeast). Extrachromosomal circular DNA (eccDNA) has been found in worms, flies, and humans. The origin and role of eccDNA in aging, if any, is unknown.

Despite the lack of a connection between circular DNA and aging in higher organisms, extra copies of Sir2 are capable of extending the lifespan of both worms and flies (though, in flies, this finding has not been replicated by other investigators, and the activator of Sir2 resveratrol does not reproducibly increase lifespan in either species.[9]) Whether the Sir2 homologues in higher organisms have any role in lifespan is unclear, but the human SIRT1 protein has been demonstrated to deacetylate p53, Ku70, and the forkhead family of transcription factors. SIRT1 can also regulate acetylates such as CBP/p300, and has been shown to deacetylate specific histone residues.

RAS1 and RAS2 also affect aging in yeast and have a human homologue. RAS2 overexpression has been shown to extend lifespan in yeast.

Other genes regulate aging in yeast by increasing the resistance to oxidative stress. Superoxide dismutase, a protein that protects against the effects of mitochondrial free radicals, can extend yeast lifespan in stationary phase when overexpressed.

In higher organisms, aging is likely to be regulated in part through the insulin/IGF-1 pathway. Mutations that affect insulin-like signaling in worms, flies, and the growth hormone/IGF1 axis in mice are associated with extended lifespan. In yeast, Sir2 activity is regulated by the nicotinamidase PNC1. PNC1 is transcriptionally upregulated under stressful conditions such as caloric restriction, heat shock, and osmotic shock. By converting nicotinamide to niacin, nicotinamide is removed, inhibiting the activity of Sir2. A nicotinamidase found in humans, known as PBEF, may serve a similar function, and a secreted form of PBEF known as visfatin may help to regulate serum insulin levels. It is not known, however, whether these mechanisms also exist in humans, since there are obvious differences in biology between humans and model organisms.

Sir2 activity has been shown to increase under calorie restriction. Due to the lack of available glucose in the cells, more NAD+ is available and can activate Sir2. Resveratrol, a stilbenoid found in the skin of red grapes, was reported to extend the lifespan of yeast, worms, and flies (the lifespan extension in flies and worms have proved to be irreproducible by independent investigators[9]). It has been shown to activate Sir2 and therefore mimics the effects of calorie restriction, if one accepts that caloric restriction is indeed dependent on Sir2.

According to the GenAge database of aging-related genes, there are over 1800 genes altering lifespan in model organisms: 838 in the soil roundworm (Caenorhabditis elegans), 883 in the bakers' yeast (Saccharomyces cerevisiae), 170 in the fruit fly (Drosophila melanogaster) and 126 in the mouse (Mus musculus).[10]

The following is a list of genes connected to longevity through research [10] on model organisms:

Ned Sharpless and collaborators demonstrated the first in vivo link between p16-expression and lifespan.[11] They found reduced p16 expression in some tissues of mice with mutations that extend lifespan, as well as in mice that had their lifespan extended by food restriction. Jan van Deursen and Darren Baker in collaboration with Andre Terzic at the Mayo Clinic in Rochester, Minn., provided the first in vivo evidence for a causal link between cellular senescence and aging by preventing the accumulation of senescent cells in BubR1 progeroid mice.[12] In the absence of senescent cells, the mices tissues showed a major improvement in the usual burden of age-related disorders. They did not develop cataracts, avoided the usual wasting of muscle with age. They retained the fat layers in the skin that usually thin out with age and, in people, cause wrinkling. A second study led by Jan van Deursen in collaboration with a team of collaborators at the Mayo Clinic and Groningen University, provided the first direct in vivo evidence that cellular senescence causes signs of aging by eliminating senescent cells from progeroid mice by introducing a drug-inducible suicide gene and then treating the mice with the drug to kill senescent cells selectively, as opposed to decreasing whole body p16.[13] Another Mayo study led by James Kirkland in collaboration with Scripps and other groups demonstrated that senolytics, drugs that target senescent cells, enhance cardiac function and improve vascular reactivity in old mice, alleviate gait disturbance caused by radiation in mice, and delay frailty, neurological dysfunction, and osteoporosis in progeroid mice. Discovery of senolytic drugs was based on a hypothesis-driven approach: the investigators leveraged the observation that senescent cells are resistant to apoptosis to discover that pro-survival pathways are up-regulated in these cells. They demonstrated that these survival pathways are the "Achilles heel" of senescent cells using RNA interference approaches, including Bcl-2-, AKT-, p21-, and tyrosine kinase-related pathways. They then used drugs known to target the identified pathways and showed these drugs kill senescent cells by apoptosis in culture and decrease senescent cell burden in multiple tissues in vivo. Importantly, these drugs had long term effects after a single dose, consistent with removal of senescent cells, rather than a temporary effect requiring continued presence of the drugs. This was the first study to show that clearing senescent cells enhances function in chronologically aged mice.[14]

See the original post:
Genetics of aging - Wikipedia

Read More...

Genetics and Genomics of Human Longevity

Monday, September 24th, 2018

References

Abdelmohsen K, Panda A, Kang MJ et al. (2013) Senescenceassociated lncRNAs: senescenceassociated long noncoding RNAs. Aging Cell 12: 890900.

Anselmi CV, Malovini A, Roncarati R et al. (2009) Association of the FOXO3A locus with extreme longevity in a southern Italian centenarian study. Rejuvenation Research 12: 95104.

Atzmon G, Cho M, Cawthon RM et al. (2010) Evolution in health and medicine Sackler colloquium: genetic variation in human telomerase is associated with telomere length in Ashkenazi centenarians. Proceedings of the National Academy of Sciences of the USA 107 (Suppl 1): 17101717.

Barzilai N, Atzmon G, Schechter C et al. (2003) Unique lipoprotein phenotype and genotype associated with exceptional longevity. JAMA: The Journal of the American Medical Association 290: 20302040.

Beekman M, Blanche H, Perola M et al. (2013) Genomewide linkage analysis for human longevity: genetics of Healthy Aging Study. Aging Cell 12: 184193.

Beekman M, Nederstigt C, Suchiman HE et al. (2010) Genomewide association study (GWAS)identified disease risk alleles do not compromise human longevity. Proceedings of the National Academy of Sciences of the USA 107: 1804618049.

Bell JT, Tsai PC, Yang TP et al. (2012) Epigenomewide scans identify differentially methylated regions for age and agerelated phenotypes in a healthy ageing population. PLoS Genetics 8: e1002629.

Bonafe M, Barbieri M, Marchegiani F et al. (2003) Polymorphic variants of insulinlike growth factor I (IGFI) receptor and phosphoinositide 3kinase genes affect IGFI plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control. Journal of Clinical Endocrinology and Metabolism 88: 32993304.

Bonafe M, Marchegiani F, Cardelli M et al. (2002) Genetic analysis of Paraoxonase (PON1) locus reveals an increased frequency of Arg192 allele in centenarians. European Journal of Human Genetics: EJHG 10: 292296.

Boyden SE and Kunkel LM (2010) Highdensity genomewide linkage analysis of exceptional human longevity identifies multiple novel loci. PLoS One 5: e12432.

Campo S, Sardo MA, Trimarchi G et al. (2004) Association between serum paraoxonase (PON1) gene promoter T(107)C polymorphism, PON1 activity and HDL levels in healthy Sicilian octogenarians. Experimental Gerontology 39: 10891094.

Carrieri G, Marzi E, Olivieri F et al. (2004) The G/C915 polymorphism of transforming growth factor beta1 is associated with human longevity: a study in Italian centenarians. Aging Cell 3: 443448.

Deelen J, Beekman M, Uh HW et al. (2011) Genomewide association study identifies a single major locus contributing to survival into old age; the APOE locus revisited. Aging Cell 10: 686698.

Deelen J, Beekman M, Uh HW et al. (2014) Genomewide association metaanalysis of human longevity identifies a novel locus conferring survival beyond 90 years of age. Human Molecular Genetics 23: 44204432.

Deelen J, Uh HW, Monajemi R et al. (2013) Gene set analysis of GWAS data for human longevity highlights the relevance of the insulin/IGF1 signaling and telomere maintenance pathways. Age 35: 235249.

Eggertsen G, Tegelman R, Ericsson S, Angelin B and Berglund L (1993) Apolipoprotein E polymorphism in a healthy Swedish population: variation of allele frequency with age and relation to serum lipid concentrations. Clinical Chemistry 39: 21252129.

Flachsbart F, Caliebe A, Kleindorp R et al. (2009) Association of FOXO3A variation with human longevity confirmed in German centenarians. Proceedings of the National Academy of Sciences of the USA 106: 27002705.

Garatachea N, Emanuele E, Calero M et al. (2014) ApoE gene and exceptional longevity: insights from three independent cohorts. Experimental Gerontology 53: 1623.

Gaspari L, Pedotti P, Bonafe M et al. (2003) Metabolic gene polymorphisms and p53 mutations in healthy centenarians and younger controls. Biomarkers: Biochemical Indicators of Exposure, Response, and Susceptibility to Chemicals 8: 522528.

Gombar S, Jung HJ, Dong F et al. (2012) Comprehensive microRNA profiling in Bcells of human centenarians by massively parallel sequencing. BMC Genomics 13: 353364.

Gro S, UtaDorothee I, Klintschar M and Bartel F (2014) Germline genetics of the p53 pathway affect longevity in a gender specific manner. Current Aging Science. doi:10.2174/1874609807666140321150751 (Epub ahead of print).

HalaschekWiener J, Vulto I, Fornika D et al. (2008) Reduced telomere length variation in healthy oldest old. Mechanisms of Ageing and Development 129: 638641.

Hannum G, Guinney J, Zhao L et al. (2013) Genomewide methylation profiles reveal quantitative views of human aging rates. Molecular Cell 49: 359367.

van Heemst D, Beekman M, Mooijaart SP et al. (2005a) Reduced insulin/IGF1 signalling and human longevity. Aging Cell 4: 7985.

van Heemst D, Mooijaart SP, Beekman M et al. and Long Life study g (2005b) Variation in the human TP53 gene affects old age survival and cancer mortality. Experimental Gerontology 40: 1115.

vB Hjelmborg J, Iachine I, Skytthe A et al. (2006) Genetic influence on human lifespan and longevity. Human Genetics 119: 312321.

Hurme M, Lehtimaki T, Jylha M, Karhunen PJ and Hervonen A (2005) Interleukin6 174G/C polymorphism and longevity: a followup study. Mechanisms of Ageing and Development 126: 417418.

Kerber RA, O'Brien E, Boucher KM, Smith KR and Cawthon RM (2012) A genomewide study replicates linkage of 3p2224 to extreme longevity in humans and identifies possible additional loci. PLoS One 7: e34746.

Kojima T, Kamei H, Aizu T et al. (2004) Association analysis between longevity in the Japanese population and polymorphic variants of genes involved in insulin and insulinlike growth factor 1 signaling pathways. Experimental Gerontology 39: 15951598.

Lee JH, Cheng R, Honig LS et al. (2014) Genome wide association and linkage analyses identified three loci4q25, 17q23.2, and 10q11.21associated with variation in leukocyte telomere length: the Long Life Family Study. Frontiers in Genetics 4: 310.

Li Y, Wang WJ, Cao H et al. (2009) Genetic association of FOXO1A and FOXO3A with longevity trait in Han Chinese populations. Human Molecular Genetics 18: 48974904.

Lio D, Scola L, Crivello A et al. (2002) Genderspecific association between 1082 IL10 promoter polymorphism and longevity. Genes and Immunity 3: 3033.

Lunetta KL, D'Agostino RB Sr., Karasik D et al. (2007) Genetic correlates of longevity and selected agerelated phenotypes: a genomewide association study in the Framingham Study. BMC Medical Genetics 8 (Suppl 1): S13.

Moskalev AA, Aliper AM, SmitMcBride Z, Buzdin A and Zhavoronkov A (2014) Genetics and epigenetics of aging and longevity. Cell Cycle 13: 10631077.

Murabito JM, Yuan R and Lunetta KL (2012) The search for longevity and healthy aging genes: insights from epidemiological studies and samples of longlived individuals. Journals of Gerontology Series A, Biological Sciences and Medical Sciences 67: 470479.

Nebel A, Kleindorp R, Caliebe A et al. (2011) A genomewide association study confirms APOE as the major gene influencing survival in longlived individuals. Mechanisms of Ageing and Development 132: 324330.

Newman AB, Glynn NW, Taylor CA et al. (2011) Health and function of participants in the Long Life Family Study: a comparison with other cohorts. Aging 3: 6376.

Noren Hooten N, Fitzpatrick M, Wood WH III et al. (2013) Agerelated changes in microRNA levels in serum. Aging 5: 725740.

Pawlikowska L, Hu D, Huntsman S et al. (2009) Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell 8: 460472.

Schachter F, FaureDelanef L, Guenot F et al. (1994) Genetic associations with human longevity at the APOE and ACE loci. Nature Genetics 6: 2932.

Sebastiani P, Bae H, Sun FX et al. (2013) Metaanalysis of genetic variants associated with human exceptional longevity. Aging 5: 653661.

Sebastiani P, Riva A, Montano M et al. (2011) Whole genome sequences of a male and female supercentenarian, ages greater than 114 years. Frontiers in Genetics 2: 90.

Sebastiani P, Solovieff N, Dewan AT et al. (2012) Genetic signatures of exceptional longevity in humans. PLoS One 7: e29848.

Soerensen M, Dato S, Tan Q et al. (2012a) Human longevity and variation in GH/IGF1/insulin signaling, DNA damage signaling and repair and pro/antioxidant pathway genes: cross sectional and longitudinal studies. Experimental Gerontology 47: 379387.

Soerensen M, Dato S, Tan Q et al. (2013) Evidence from casecontrol and longitudinal studies supports associations of genetic variation in APOE, CETP, and IL6 with human longevity. Age 35: 487500.

Soerensen M, Thinggaard M, Nygaard M et al. (2012b) Genetic variation in TERT and TERC and human leukocyte telomere length and longevity: a crosssectional and longitudinal analysis. Aging Cell 11: 223227.

Stessman J, Maaravi Y, HammermanRozenberg R et al. (2005) Candidate genes associated with ageing and life expectancy in the Jerusalem longitudinal study. Mechanisms of Ageing and Development 126: 333339.

Willcox BJ, Donlon TA, He Q et al. (2008) FOXO3A genotype is strongly associated with human longevity. Proceedings of the National Academy of Sciences of the USA 105: 1398713992.

Willcox BJ, He Q, Chen R et al. (2006) Midlife risk factors and healthy survival in men. JAMA: the Journal of the American Medical Association 296: 23432350.

Xia Y, Gueguen R, VincentViry M, Siest G and Visvikis S (2003) Effect of six candidate genes on early aging in a French population. Aging Clinical and Experimental Research 15: 111116.

Further Reading

Andersen SL, Sebastiani P, Dworkis DA, Feldman L and Perls TT (2012) Health span approximates life span among many supercentenarians: compression of morbidity at the approximate limit of life span. Journals of Gerontology Series A, Biological Sciences and Medical Sciences 67: 395405.

Chang AL, Bitter PH Jr., Qu K et al. (2013) Rejuvenation of gene expression pattern of aged human skin by broadband light treatment: a pilot study. Journal of Investigative Dermatology 133: 394402.

Chung WH, Dao RL, Chen LK and Hung SI (2010) The role of genetic variants in human longevity. Ageing Research Reviews 9 (Suppl 1): S67S78.

Kahn AJ (2014) FOXO3 and related transcription factors in development, aging, and exceptional longevity. Journals of Gerontology Series A, Biological Sciences and Medical Sciences. doi: 10.1093/gerona/glu044 (Epub ahead of print).

LabatRobert J and Robert L (2014) Longevity and aging. Role of free radicals and xanthine oxidase. A review. PathologieBiologie 62: 6166.

Raichlen DA and Alexander GE (2014) Exercise, APOE genotype, and the evolution of the human lifespan. Trends in Neurosciences 37: 247255.

Rajpathak SN, Liu Y, BenDavid O et al. (2011) Lifestyle factors of people with exceptional longevity. Journal of the American Geriatrics Society 59: 15091512.

Seripa D, D'Onofrio G, Panza F et al. (2011) The genetics of the human APOE polymorphism. Rejuvenation Research 14: 491500.

Sevini F, Giuliani C, Vianello D et al. (2014) mtDNA mutations in human aging and longevity: controversies and new perspectives opened by highthroughput technologies. Experimental Gerontology 56: 234244.

Zhu H, Belcher M and van der Harst P (2011) Healthy aging and disease: role for telomere biology? Clinical Science 120: 427440.

Read this article:
Genetics and Genomics of Human Longevity

Read More...

Longevity FAQ Laura Deming

Wednesday, August 22nd, 2018

Senescent cell removal135%2016Does not affect rotarod performance, object discrimination. Slight delay in wound closure.1Rapamycin110%2009Late-life rapamyicn treatment extends lifespan (pooled females from multiple-site NIA study)2NR105%2016Claim an increase in running distance3Catalase117%2005Mitochondrially-targeted catalase expression extended mouse lifespan compared to control4Sirt6 overexpression115%2012Sirt6-overexpression increases male mouse lifespan5Metformin106%2013In males, small but significant lifespan extension after metformin application6DN-IB110%2013Dominant negative to downregulate IKK-beta activity, delivered to hypothalamus of middle-aged mice7Klotho120%2005Overexpression under human elongation factor 1 promoter increases lifespan, slight fertility loss8S6K1118%2009KO of S6K1 extends lifspan compared to wildtype mice9p66128%1999Mutation of a p66shc, member of proto-oncogene locus SHC, extends lifespan. May be just due to cancer effect.10Lowering protein:carbohydrate ratio128%2014Varied protein, carbohydrate, and total energy levels.11Fat-specific insulin receptor knockout mice111%2003Fat-specific insulin receptor knockout mice show a significant increase in lifespan12C57BL/6 mice with NZB/OlaHsd mitochondrial mutations120%2016Same nuclear, different mitochondrial DNA.13Fasting mimicking diet112%2015FMD followed by 10 days of normal, then repeat14Rapamycin127%2014Rapamycin from 9 months of age, weight decreased ~30% at highest dose15Brain-specific Sirt1 expression116%2013Brain-specific Sirt1 expression in female mice increases lifespan over wildtype16SRT1720104%2014Start diet at 28 weeks of age, very small increase on lifespan17Spermidine111%2016Polyamine, administered in drinking water18Atg5 overexpression117%2013Transgenic mice ubiquitously expressing Atg5 (crucial for autophagasome confirmation) live longer.19Telomerase124%2012Paper showing telomerase therapy increasing life20Insulin receptor substrate null132%2008Insulin receptor substrate 1 null mouse lifespan extension in females21Snell Dwarf Mice142%2001Snell dwarf mouse paper showing life extension22Ames Dwarf Mice168%1996Original Ames dwarf mouse paper showing life extension23s-Arf/p53113%2007An extra copy of p53 and upstream regulator Arf/p16Ink4a increases lifespan24Slow growth during lactation106%2004Male mice suckled by dams fed a low-protein diet lived longer than their control cohort25Methionine restriction111%2005Methionine restriction increases mouse lifespan, here median lifespan increase in mice that survived at least 1 yr.26Rapamycin (3 months)114%2016Lifespan given from time of treatment which was 23-24 mo, used 24 mo to get percentage so this is an estimate27GHR-BP138%2000Mice deficient in growth hormone receptor / binding protein live longer (female mean, not median, lifespan shown here)28mTOR116%2013mTOR depletion extends lifespan29PTEN overexpression112%2012Overexpression of PTEN, a tumor suppressor which counteracts PI3K, extends mouse lifespan30Myc (+/-)121%2015Claim no correlation between weight and lifespan31FGF-21139%2012Hepatic-specific expression of FGF-21 (which suppresses growth hormone and reduces the production of IGF) increases lifespan, female lifespan shown here32BubR1 overexpression114%2012Kinase which localizes to kinetochore, overexpression increases lifespan33AC5 KO132%2007AC5 knockount mice lived longer than control, potentially linked to effects on cAMP production and beta-adrenergic receptor signaling.3417-alpha-estradiol112%201317-alpha-estradiol extended lifespan in males, but not females (as expected)35Acarbose122%2013Acarbose extended male more than female lifespan36TRPV1 -/-114%2014Resting exchange ratio similar at 16 mo to 3 mo37SRT2104106%2014Start diet at 28 weeks of age, very small increase if there38Hcrt-UCP2128%2006UCP2 under hypocretin promoter lowers core body temp, increases lifespan39G6PD overexpression114%2016Reduces NADP+40IGF-1 Receptor Brain KO (+/-)109%2008Brain-specific IGF-1 Receptor +/- mice live longer than WT41SURF-1 KO121%2007Mutations in SURF1, a cytochrome c oxidase assembly factor, extend lifespan. Mitochondrial.42Litter enlargemnet (CR)118%200950% enlargement of litter in first 20 days, to induce caloric restriction43mclk-1 heterozygous115%2005A heterozygous knockout of mclk1 (important in mitochondrial respiration) results in mouse lifespan extension compared to wildtype44Nordihydroguairaitic acid112%2008NDGA and aspirin extend lifespan by a little bit. Small molecule.45Aspirin108%2008NDGA and aspirin extend lifespan by a little bit. Small molecule.46SOD mimetic carboxyfullerene115%2008Carboxyfullerene, described as an SOD mimetic, increased the lifespan of treated mice compared to wildtype control47Removal of visceral fat tissue108%2008Removal of visceral fat tissue increases lifespan over control48Low glycotoxin diet112%2007Low glycotoxin (low levels of AGE's) shown to extend lifespan49Per2 (-/-)118%2016Lifespan study incomplete50Neonatal metformin120%2015Animals recieved on 3, 5, 7th day after birth - bad for females, good for males.51GHRH KO146%2013GHRH (Growth-Hormone Releasing Hormone) disruption extends lifespan, presumably through the insulin/IGF pathway axis52Sod-2 overexpresion104%2007Overexpression of SOD-2 targeted to the mitochondrion increases mouse lifespan relative to wildtype53Metallothionein cardiac-specific expression114%2006Cardiac-specific expression of antioxidant metallothionein extended the lifespan of wildtype mice compared to WT FVB control.54IGF1R(+/-)121%2013Tyrosine kinase receptor activated by IGF1/255Ink4a/Arf/Ink4b116%2009Encodes 2 CDKs (p16 and p15), and Arf (upstream of p53)56Adult-onset Ghr (-/-)100%2016Male mice have >2x higher insulin than female mice57Ovary Transplantation117%2003Original paper showing that transplantation of young ovaries into old animals could result in lifespan increase58UCP-1 transgenic111%2007Transgenic mice with skeletal muscle-specific UCP1 had increased longevity. Small increase if there.59PAPP131%2010Knockout of PAPP-A (which enhances IGF-1 activity by degrading the inhibitory IGF-binding protein) increases lifespan over wildtype, female lifespan shown here60CR diet with lard132%201540% decrease starting at 4 months61loss of function of Riib (PKA subunit)114%2009Knockout of RIIbeta, a subunit of PKA, increased lifespan in mice compared to wildtype62Myostatin (+/-)109%2015Knockout induces double-muscle mice63Akt1 +/-113%2013Haploinsufficiency of Akt1 increases mouse lifespan relative to wildtype. Insulin/IGF-1 pathway.64miR-17117%2014Not clear if there is a main function for miR-1765NDGA111%2015Makes up ~12.5% of the dry weight of leaves66FAT10ko119%2014Ubiquitin-like protein which can signal for protein to go to proteasome.67Intranasal Hsp70116%2015Seemed to extend lifespan when started at 17 months68RasGRF1(-/-)120%2011Ras-guanine nucleotide exchange factor (Ras-GRF1) -/- mice displayed increased lifespan compared to wildtype.69Lmna-Lcs (Lamin C alone)113%2014Body weight and tumor incidence increase in mice expressing only Lamin-C70Cisd2 overexpression119%2011Cisd2 transgenic mice (expressing more of it) lived longer than wildtype. Cisd2 is a transmembrane protein expressed on the mitochondrial outer membrane and associated with a human longevity locus.71metoprolol110%2013Administration of the beta-adrenerginc receptor blocker metoprolol to mice increased lifespan compared to wildtype72nebivolol106%2013Administration of the beta-adrenerginc receptor blocker nebivolol to mice increased lifespan compared to wildtype73uPA (in ocular lens/CNS nerve cells)118%1997uPA expression under alpha-crystallin promoter increases lifespan, small/eat less74MIF-1 KO116%2010MIF-1 knockout mutant (T-cell derived cytokine) extends lifespan75mGsta4-null113%2009Enzyme protects against lipid peroxidation, weird that less of its activity might increase lifespan76Muscle-specific GHRKO109%2015Knockout under muscle creatinine kinase promoter77CAM-(1A)AR mice110%2011Mice with a constitutively active mutant form of the alpha1-adrenergic receptor (CAM-alpha1aAR) lived longer than wildtype control78Cardiac-specific catalase overexpression113%2007Overexpression of catalase specifically in the heart in mice79Icariin108%2015Flavonoid80miR-29 brain-specific KO112%2016miR-29 highly expresed in brain during development81Bi-maternal mice128%2010Mice prepared to be bi-maternal were found longer-lived than their normal cohort82RNase-L(-/-)127%2007Knockout of RNase-L, which accelerates cell senescence when expressed, increases lifespan in mice compared to wildtype83hMTH1-Tg116%2013Express high levels of hMTH1 hydrolase, thought to degrade 8-oxodGTP and 8-oxoGTP. Oxidative stress.84DGAT-1 -/-126%2012Knockout of DGAT1, which catalyzes triglyceride synthesis, extends mouse lifespan relative to wildtype85IGFBP-2 overexpression105%2016Proteins bind IGF1/2, degraded during pregnancy, delay in sexual maturity86PAPP-A on high-fat diet105%2015Males chosen so no adverse developmental effect on fat depots87clk-1(-/-) with clk-1 transgene128%2014clk-1 functions in ubiquinone synthesis, but levels weren't very affected.88AgRP -/-110%2006Neuropeptide that is appetite stimulator, overexpression leads to hyperphagia and obesity.89Bone marrow transplantation106%2013Bone marrow transplantation from young to old mice was claimed to extend lifespan90Young blood injections94%2014Resulted in decreased lifespan91Nas(-/-) mice125%2011Hyposulfatemic NaS1 null mice (Nas1 -/-) had an increased lifespan compared to wildtype control.92Cyclophilin D (+/-)119%2017Decrease in maximum lifespan93PAPP-A in adults120%2017Tamoxifen-induced knockdown94Mtbp (+/-)120%2016Rotarod, open field, blood glucose, insulin, IGF-1 were the same.95

Go here to see the original:
Longevity FAQ Laura Deming

Read More...

Rhonda Patrick, Ph.D.: the performance and longevity …

Monday, August 13th, 2018

Rhonda Patrick, Ph.D. and I go on a Nerd Safari into the jungle of health, nutrition, fitness, performance, and longevity. We visit IGF-1 and whether theres a tradeoff between having high or low levels. We discuss the PPARs (receptor proteins) and genetic polymorphisms. Does Rhonda think theres any benefit in a NAD+ booster for health and longevity? Can saunas lower the risk of heart disease, dementia, and all-cause mortality? We dig into those questionsand a lot more.

Subscribe on:APPLE PODCASTS | RSS | GOOGLE | OVERCAST | STITCHER

Rhonda is a wealth of knowledge and was the perfect companion to explore several interesting topics in this episode. She puts a great deal of thought and effort into her research, and it really shows in this conversation.

What Rhonda believes differently today versus 5 years ago? [5:40]

Calorie restriction and IGF reduction. Is it the best way to boost longevity? [6:30]

Rhondas changing opinion on the ketogenic diet. [9:00]

Peters experience with the ketogenic diet [10:30]

Exogenous ketones: Rhondas personal experience. [13:00]

Diet-induced ketosis, purported benefits and detriments, limitations of current studies, and what Peter would like to see in a future study. [15:30]

The practicality of the ketogenic diet and how to get your nutrients. [16:45]

The IGF-1 paradox, is it good or bad? [21:00]

Misconceptions about protein levels in the ketogenic diet. [22:00]

Intro to PPAR alpha and PPAR gamma, polymorphisms that impact fatty acid metabolism, ketogenesis, and how we react to saturated fat. [23:00]

Saturated fat and genetic variants that may affect how we respond to consuming it. [25:30]

How certain genes variants may affect certain peoples reaction to saturated fat. [29:00]

Rhonda has developed a genetic testing tool available to the public. [30:00]

Why some people have trouble producing ketones and how exercise and fasting may be the crucial piece for getting over the hump. [31:00]

Rhondas approach to eating/fasting/exercise and using exogenous ketones by HVMN. [34:45]

Can ketone esters be used to reduce blood glucose levels? [41:15]

Acarbose for controlling blood glucose. [41:45]

Peter and Rhonda share their evolving understanding of the IGF-1 literature. [42:15]

Only the germ cells in C. elegans divide, which may make cancer in this organism fundamentally different than humans.Nematodes have a fixed, genetically determined number of cells, a phenomenon known as eutely. The adult hermaphrodite has exactly 959 cells. The male C. elegans has 1031 cells. The number of cells does not change after cell division ceases at the end of the larval period, and subsequent growth is due solely to an increase in the size of individual cells. [Wikipedia]

Do we want low or high IGF-1? Or is cycling the key? [48:00]

Figure. Predicted HR for the association between IGF-I and all-cause mortality. [Burgers et al., 2011]

Image credit: Meta-analysis and dose-response metaregression: circulating insulin-like growth factor I (IGF-I) and mortality (Burgers et al., 2011)

Figure. Relationship between serum IGF-1 levels and risk of (A) all-cause mortality (B) cancer mortality and (C) cardiovascular disease (CVD) mortality. [Svensson et al., 2012]

Image credit: Both Low and High Serum IGF-I Levels Associate with Cancer Mortality in Older Men (Svensson et al., 2012)

The important role that IGF-1 plays in muscle and brain tissue through exercise. [50:00]

Efficacy of prolonged fasting for cancer, autoimmune diseases, and other afflictions. [51:00]

Prolonged fasting and cancer, how it could maybe be apart of standard of care in the near future. [53:00]

Can chemo patients benefit from fasting and certain dietary protocols? [54:15]

Can fasting help with the response to, and recovery from, invasive operations? [55:00]

Importance of exercise for brain health. [59:30]

VO2 max, cardiorespiratory fitness, strength training, and how it affects our health. [1:03:15]

Can lowering inflammation be a key to extending life? [1:06:30]

Peter shares his hope/vision for the future of personalized health protocols. [1:11:45]

Sauna, and the growing evidence for the benefits of heat therapy. [1:12:30]

Does sauna have an impact on sleep? [1:13:30]

Saunas and the healthy-user bias, a critical look at the literature. [1:14:15]

The overlapping physiological responses of heat therapy and exercise. [1:16:15]

Saunas as an antidepressant? [1:17:15]

Different types of saunas and which one Rhonda likes best. [1:18:15]

Can saunas act asan anti-inflammatory and improve insulin sensitivity? [1:20:15]

Can saunas help prevent neurodegeneration? [1:21:45]

What kind of disease is dementia? And how might ketones and saunas help? [1:22:15]

Cold therapy vs heat therapy, similarities, and differences. [1:24:30]

Can we stack hot and cold therapy to maximize the benefits? [1:28:30]

Cold therapy and mitochondrial biogenesis. [1:29:00]

How cold therapy can blunt hypertrophy from strength training. [1:31:15]

A primer on NAD+/NADH, its effect on lifespan/healthspan, and a review of the supplements. [1:32:45]

PARP, an important enzyme for DNA repair, needs NAD+ for fuel. [1:34:30]

What causes NAD+ to decrease as we age? [1:35:00]

Could metformin negatively affect the NAD+ to NADH ratio? [1:36:15]

Evidence for NAD+ supplements. [1:37:00]

Can we increase NAD+ levels with fasting? [1:38:00]

Peter asks Rhonda, What is the most interesting question you dont yet know the answer to but you feel like is knowable? [1:39:15]

Rapamycin, the most promising life-extension drug? [1:42:30]

The next medical frontier: specificity and selectivity of drugs. [1:45:00]

Where you can find Rhonda and her work. [1:46:45]

Rhonda Perciavalle Patrick has a Ph.D. in biomedical science from the University of Tennessee Health Science Center, Memphis TN and St. Jude Childrens Research Hospital, Memphis TN. She also has a Bachelors of Science degree in biochemistry/chemistry from the University of California, San Diego. She has done extensive research on aging, cancer, and nutrition. She did her graduate research at St. Jude Childrens Research Hospital where she investigated the link between mitochondrial metabolism, apoptosis, and cancer. Her groundbreaking work discovered that a protein that is critical for cell survival has two distinct mitochondrial localizations with disparate functions, linking its anti-apoptotic role to a previously unrecognized role in mitochondrial respiration and maintenance of mitochondrial structure. Her dissertation findings were published in the 2012 issue of Nature Cell Biology.

Dr. Patrick trained as a postdoctoral fellow at Childrens Hospital Oakland Research Institute with Dr. Bruce Ames. She investigated the effects of micronutrient (vitamins and minerals) inadequacies on metabolism, inflammation, DNA damage, and aging and whether supplementation can reverse the damage. In addition, she also investigated the role of vitamin D in brain function, behavior, and other physiological functions. In February of 2014 she published a paper in FASEB on how vitamin D regulates serotonin synthesis and how this relates to autism.

Dr. Patrick has also done research on aging at the Salk Institute for Biological Sciences. At the Salk she investigated what role insulin signaling played in protein misfolding, which is commonly found in neurodegenerative diseases.

She frequently engages the public on topics including the role micronutrient deficiencies play in diseases of aging, the role of genetics in determining the effects of nutrients on a persons health status, benefits of exposing the body to hormetic stressors, such as through exercise, fasting, sauna use or heat stress, or various forms of cold exposure, and the importance of mindfulness, stress reduction, and sleep. It is Dr. Patricks goal to challenge the status quo and encourage the wider public to think about health and longevity using a proactive, preventative approach. [ FoundMyFitness.com]

Rhonda on Facebook: FoundMyFitness

Rhonda on Instagram: @foundmyfitness

Rhonda on Twitter: @foundmyfitness

Rhondas website: FoundMyFitness.com

Rhondas podcast: FoundMyFitness

See the original post here:
Rhonda Patrick, Ph.D.: the performance and longevity ...

Read More...

Picea abies – Wikipedia

Tuesday, July 31st, 2018

Picea abies, the Norway spruce,[3] is a species of spruce native to Northern, Central and Eastern Europe.[4] It has branchlets that typically hang downwards, and the largest cones of any spruce, 917cm (312634in) long. It is very closely related to the Siberian spruce (Picea obovata), which replaces it east of the Ural Mountains, and with which it hybridises freely. The Norway spruce is widely planted for its wood, and is the species used as the main Christmas tree in several cities around the world. It was the first gymnosperm to have its genome sequenced, and one clone has been measured as 9,550 years old.

The Latin specific epithet abies means fir-like.[5]

Norway spruce is a large, fast-growing evergreen coniferous tree growing 3555m (115180ft) tall and with a trunk diameter of 1 to 1.5m (39 to 59in). It can grow fast when young, up to 1m (3ft) per year for the first 25 years under good conditions, but becomes slower once over 20m (65ft) tall.[6] The shoots are orange-brown and glabrous (hairless). The leaves are needle-like with blunt tips,[7] 1224mm (15321516in) long, quadrangular in cross-section (not flattened), and dark green on all four sides with inconspicuous stomatal lines. The seed cones are 917cm (312634in) long (the longest of any spruce), and have bluntly to sharply triangular-pointed scale tips. They are green or reddish, maturing brown 57 months after pollination. The seeds are black, 45mm (532316in) long, with a pale brown 15-millimetre (58-inch) wing.[1][8][9][10][11]

The tallest measured Norway spruce is 62.26m (204ft) tall and grows near Ribnica na Pohorju, Slovenia.[12]

It can often be observed that the roots of spruces pushed over in a storm form a relatively flat disc. This is usually due to the rocky subsurface, a high clay content or oxygen-depletion of the subsoil and not to a preference of the trees to form a flat root system.[citation needed]

The Norway spruce grows throughout Europe from Norway in the northwest and Poland eastward, and also in the mountains of central Europe, southwest to the western end of the Alps, and southeast in the Carpathians and Balkans to the extreme north of Greece. The northern limit is in the arctic, just north of 70N in Norway. Its eastern limit in Russia is hard to define, due to extensive hybridisation and intergradation with the Siberian spruce, but is usually given as the Ural Mountains. However, trees showing some Siberian spruce characters extend as far west as much of northern Finland, with a few records in northeast Norway. The hybrid is known as Picea fennica (or P.abies subsp. fennica, if the two taxa are considered subspecies), and can be distinguished by a tendency towards having hairy shoots and cones with smoothly rounded scales.[8][9][10]

Norway spruce cone scales are used as food by the caterpillars of the tortrix moth Cydia illutana, whereas Cydia duplicana feeds on the bark around injuries or canker.

Cones of P.obovata and Picea abies

Cones of P.obovata are short and have rounded scales.

Cones of P.abies are longer and have pointed scales.

Populations in southeast Europe tend to have on average longer cones with more pointed scales; these are sometimes distinguished as Picea abies var. acuminata (Beck) Dallim. & A.B. Jacks., but there is extensive overlap in variation with trees from other parts of the range.[8][9][10]

Some botanists treat Siberian spruce as a subspecies of Norway spruce, though in their typical forms, they are very distinct, the Siberian spruce having cones only 510cm long, with smoothly rounded scales, and pubescent (hairy) shoots.[8][9][10] Genetically Norway and Siberian spruces have turned out to be extremely similar and may be considered as two closely related subspecies of P.abies.[13]

Another spruce with smoothly rounded cone scales and hairy shoots occurs rarely in the Central Alps in eastern Switzerland. It is also distinct in having thicker, blue-green leaves. Many texts treat this as a variant of Norway spruce, but it is as distinct as many other spruces, and appears to be more closely related to Siberian spruce (Picea obovata), Schrenk's spruce (Picea schrenkiana) from central Asia and Morinda spruce (Picea smithiana) in the Himalaya. Treated as a distinct species, it takes the name Alpine spruce (Picea alpestris (Brgger) Stein). As with Siberian spruce, it hybridises extensively with Norway spruce; pure specimens are rare. Hybrids are commonly known as Norwegian spruce, which should not be confused with the pure species Norway spruce.[8][9][10]

The Norway spruce is one of the most widely planted spruces, both in and outside of its native range, and one of the most economically important coniferous species in Europe.[14] It is used as an ornamental tree in parks and gardens. It is also widely planted for use as a Christmas tree. Every Christmas, the Norwegian capital city, Oslo, provides the cities of London (the Trafalgar Square Christmas tree), Edinburgh and Washington D.C. with a Norway spruce, which is placed at the most central square of each city. This is mainly a sign of gratitude for the aid these countries gave during the Second World War.[15]In North America, Norway spruce is widely planted, specifically in the northeastern, Pacific Coast, and Rocky Mountain states, as well as in southeastern Canada. It is naturalised in some parts of North America. There are naturalised populations occurring from Connecticut to Michigan, and it is probable that they occur elsewhere.[14] Norway spruces are more tolerant of hot, humid weather than many conifers which do not thrive except in cool-summer areas and they will grow up to USDA Growing Zone 8.

In the northern US and Canada, Norway spruce is reported as invasive in some locations, however it does not pose a problem in Zones 6 and up as the seeds have a significantly reduced germination rate in areas with hot, humid summers.

The Norway spruce tolerates acidic soils well, but does not do well on dry or deficient soils. From 1928 until the 1960s it was planted on surface mine spoils in Indiana.[14]

Several cultivars have been selected as ornamentals (Barrya, Capitata, Decumbens, Dumosa, Clanbrassiliana, Gregoryana, Inversa, Microsperma, Nidiformis, Ohlendorffii, Repens, Tabuliformis, Maxwellii, 'Virgata', 'Inversa', Pendula), with a wide variety of sizes and shapes, from full-sized forest trees to extremely slow-growing, prostrate forms. They are occasionally traded under the obsolete scientific name Picea excelsa (an illegitimate name). The following cultivars have gained the Royal Horticultural Society's Award of Garden Merit:[16]

The Norway spruce is used in forestry for (softwood) timber,[22] and paper production.

The tree is the source of spruce beer, which was once used to prevent and even cure scurvy.[23] This high vitamin C content can be consumed as a tea from the shoot tips or even eaten straight from the tree when light green and new in spring.

It is esteemed as a source of tonewood by stringed-instrument makers.[24] One form of the tree called Haselfichte(de) (Hazel-spruce) grows in the European Alps and has been recognized by UNESCO as intangible cultural heritage. This form was used by Stradivarius for instruments.[25] (see German Wikipedia for details).

Norway spruce shoot tips have been used in traditional Austrian medicine internally (as syrup or tea) and externally (as baths, for inhalation, as ointments, as resin application or as tea) for treatment of disorders of the respiratory tract, skin, locomotor system, gastrointestinal tract and infections.[26]

A press release from Ume University says that a Norway spruce clone named Old Tjikko, carbon dated as 9,550 years old, is the "oldest living tree".[27]

However, Pando, a stand of 47,000 quaking aspen clones, is estimated to be between 80,000 and one million years old.[28][29][30]

The stress is on the difference between the singular "oldest tree" and the multiple "oldest trees", and between "oldest clone" and "oldest non-clone". The oldest known individual tree (that has not taken advantage of vegetative cloning) is a Great Basin bristlecone pine over 5,000 years old (germination in 3051 BC).[31]

The genome of Picea abies was sequenced in 2013, the first gymnosperm genome to be completely sequenced.[32] The genome contains approximately 20 billion base pairs and is about six times the size of the human genome, despite possessing a similar number of genes. A large proportion of the spruce genome consists of repetitive DNA sequences, including long terminal repeat transposable elements. Despite recent advances in massively parallel DNA sequencing, the assembly of such a large and repetitive genome is a particularly challenging task, mainly from a computational perspective.[33]

Within populations of Picea abies there is great genetic variability, which most likely reflect populations' post-glacial evolutionary history. Genetic diversity can in particular be detected when looking at how the populations respond to climatic conditions. E.g. variations in timing and length of the annual growth period as well as differences in frost-hardiness in spring and autumn. These annual growth patterns are important to recognise in order to choose the proper reforestation material of Picea abies.[34]

p-Hydroxybenzoic acid glucoside, picein, piceatannol and its glucoside (astringin), isorhapontin (the isorhapontigenin glucoside), catechin and ferulic acid are phenolic compounds found in mycorrhizal and non-mycorrhizal roots of Norway spruces.[35] Piceol[36] and astringin[37] are also found in P.abies.

Extracts from Picea abies have shown inhibitory activity on porcine pancreatic lipase in vitro.[38]

Picea abies (L.) H.Karst is the accepted name of this species. More than 150 synonyms of Picea abies have been published.[39]

Homotypic synonyms of Picea abies are:[40]

Some heterotypic synonyms of Picea abies are:

Read more here:
Picea abies - Wikipedia

Read More...

Genetics of healthy aging and longevity | SpringerLink

Monday, October 16th, 2017

The basis of human longevity and healthy aging, and how to achieve these desirable phenotypes, remain among the principal challenges of biology and medicine. While an understanding of lifestyle and environmental factors will maximize our ability to prevent disease and maximize health in the general population, studying the genetic basis of longevity and healthy aging in exceptional individuals is providing important biological insights. In model organisms it has been possible to demonstrate effects of mutations in genes that can extend lifespan nearly tenfold (Ayyadevara et al. 2008). Studies of inbred lab strains and of natural genetic variants in model organisms including yeast and worms (Tissenbaum and Guarente 2002), flies (Paaby and Schmidt 2009) and mice (Yuan et al. 2011) have clearly implicated many specific genes in the lifespan of these organisms. Our understanding of human lifespan stands in contrast to this, with only one consistently replicable genetic association, APOE, observed to date in several genome-wide association scans (GWAS) of longevity-related traits. This may be because healthy aging and longevity are particularly complex traits, involving not only maintenance of long-term function but also absence or reduction of disease and other morbidities. It has been proposed that human lifespan is influenced not only by longevity assurance mechanisms and disease susceptibility loci but also by the environment, geneenvironment interactions, and chance (Cournil and Kirkwood 2001). It will be important to understand the effects of environment (lifestyle) and of genetics, as well as how they interact to affect health and lifespan.

The importance of age and aging is underscored by the recognition that all common complex diseases increase with age. Questions remain about whether aging is the cause or effect of such diseases (Hekimi 2006). The study of desirable phenotypes like longevity and healthy aging has been referred to as positive biology (Farrelly 2012). Its premise is that understanding the basis for such desirable traits may allow us to design interventions to improve human health.

This review was intended to summarize our current understanding of genetic factors affecting the phenotypes of longevity and healthy aging in humans, including the definition and heritability of these traits, and linkage, association, and sequencing studies. The surprising and novel findings that centenarians do not appear to have a relative lack of common complex disease risk alleles, and that some genetic variants appear to buffer or protect against specific risk alleles, are discussed in detail. Shorter summaries of the findings related to somatic mosaicism and the promising study of epigenetics of aging are included for completeness.

The phenotypes used in studies of the genetics of human aging are usually lifespan (age at death), longevity (long life, usually defined as being a specific advanced age or older at the time of study), exceptional longevity (defined as attaining or exceeding a specific exceptional age), or healthy aging (a combination of old age and health, often defined as freedom from specific disorders or desirable performance levels on functional tests). Longevity studies focus on long-lived individuals (LLI), often centenarians aged 100 or more years. One advantage of such studies is the simplicity of phenotype definition. Healthy aging can be defined in various ways, usually with regard to reaching an at least moderately old age in the absence of certain diseases or disabilities, and/or in the presence of desirable traits such as intact cognition or mobility. Both types of studies should be differentiated from the study of the fundamental biological processes of aging (for example, cellular senescence).

A major difference between longevity and healthy aging studies is that the former focuses on lifespan, whereas the latter is focused on healthspan. Lifespan and healthspan are intimately related, however, and individuals who live exceptionally long also tend to be healthy for much of their lives. A landmark study of the health of supercentenarians (aged 110119), semisupercentenarians (aged 105109), centenarians (in this context aged 100104), nonagenarians, and younger controls found that the older the age group, the greater the delay in onset of major disease (Andersen et al. 2012). Remarkably, for every category of increasing age, the hazard ratio for each of six disorders (cancer, cardiovascular disease (CVD), dementia, hypertension, osteoporosis, and stroke) was <1.0 relative to the next oldest group. This delay in disease development and postponement of cognitive and physical decline in the oldest group amounted to a compression of morbidity (Fries 1980). Based on these findings, Andersen et al. (2012) suggest that a realistic and practical limit of human lifespan is 110115years, close to that of the oldest documented person in the world to date, who lived to 122 (Robine and Allard 1998).

Women have a lower mortality rate than men at every age, and women live longer than men in most human populations. Any given exceptional age, therefore, is more exceptional for men than for women. As noted by Sebastiani and Perls (2012) 1% of US women (but only 0.1% of men) born circa the turn of the last century lived to be 100. Potential explanations for this difference include hormonal and immune differences, hemizygosity of the X-chromosome in men (which may allow manifestation of unfavorable sex-linked variants), and unrecognized confounders [reviewed in Newman and Murabito (2013)].

Age at death in adulthood has a heritability of approximately 25% (summarized in Murabito et al. 2012). A population-based study of 2,872 Danish twin pairs born between 1870 and 1900 found that the heritability of adult lifespan was 0.26 in men and 0.23 in women (Herskind et al. 1996). This cohort was not only population-based but nearly non-censored and, with follow-up for 94years, encompassed essentially the entire human lifespan. Importantly, the heritability of longevity increases with greater age. The heritability of living to at least 100 has been estimated at 0.33 in women and 0.48 in men (Sebastiani and Perls 2012). Male and female siblings of US centenarians were 17-fold and eightfold more likely (compared with US Social Security data) to reach the age of 100, respectively (Perls et al. 2002). The increase in heritability of longevity at greater age is consistent between several studies. In over 20,000 Scandanavian twins, heritability of longevity was negligible from age 660, but increased with age thereafter (Hjelmborg et al. 2006). Long life was heritable in Icelanders aged over 70years (Gudmundsson et al. 2000). The siblings of Okinawan centenarians show increased adult survival probability that starts at age 55 and increases with age (Willcox et al. 2006); the authors speculate based in part on absence of many age-related diseases from Okinawan (Bernstein et al. 2004) and other centenarians (Evert et al. 2003), that these individuals have genetic factors that confer resistance to such diseases and increase the likelihood of reaching exceptional old age. The estimation of heritability also depends on how it is studied; Murabito et al. (2012) note that the Framingham heart study cohorts give much greater estimates of heritability when longevity is studied as a dichotomous trait (36% heritability for survival to 65 and 40% for survival to 85), compared with 16% heritability when age at death is treated as a continuous trait.

Clustering of longevity and healthy aging is observed in families. Parents of centenarians born in approximately 1870 were sevenfold more likely than their contemporaries to have lived to age 9099; offspring of centenarian parents showed lower prevalence of age-related disease than age-matched control groups (Atzmon et al. 2004). Exceptional familial clusters of extreme longevity have also been reported (Perls et al. 2000). Healthy aging is also heritable. Reed and Dick (2003) defined wellness in male twins as achieving the age of 70 free of heart attack, coronary surgery, stroke, diabetes or prostate cancer, and showed that this trait had a heritability exceeding 50%.

Environment and lifestyle likely constitute much of the remaining influence on human lifespan and healthspan. These factors have varied greatly over time and may not reflect the extrinsic factors that will affect the lifespan of babies born today. Many members of the elderly and centenarian cohorts under study today lived through times of caloric restriction (e.g., the Great Depression) and grew up before the use of antibiotics and vaccines became commonplace. The selective pressures that influenced their mortality are not identical to those experienced by later generations, and this is an important consideration for study design.

Phenotype definition is particularly important in genetic studies; it affects the interpretation and meaning of results, and the ability to compare to the results of other studies. Studies of longevity can include extreme longevity (defined as living beyond a specific extreme age) or age at death. Studies of healthy aging may use age to disease onset, successful aging or wellness (which can also have a variety of definitions), or other phenotypes (Manolio 2007). Linkage or family-based association study designs, longitudinal cohort studies, or case/control designs have been used. Family-based designs have the advantage of being robust to population stratification. Longitudinal cohorts have the advantage of limiting sampling bias, but take time and due to practical limits of size may not contain many individuals of extreme age. Sample size is a consideration for all these study designs. To date, the largest studies of LLI are in the low thousands of subjects; this is much smaller than the largest studies of common complex diseases (which now include over 100,000 subjects), despite the likely similar modest size of many of the genetic factors being sought.

Choice of a comparison group to contrast with exceptionally long-lived or exceptionally healthy elderly individuals is also critical. Health data for LLI can be compared with archived data for deceased individuals of the same birth cohort, but DNA samples from an ideal comparison group (such as their birth cohort) are not available. Case/control molecular genetic studies of long-lived or healthy aged individuals often compare elderly cases to younger controls. Potential pitfalls of such studies include inadequate detection and control for population stratification, particularly for populations that have experienced immigration of different ethnicities over time (Nebel and Schreiber 2005). The use of principal components analysis (Price et al. 2006) or genomic controls (Devlin and Roeder 1999) can mitigate this problem, as can the conduct of studies within specific ethnic groups (Barzilai et al. 2001). In the case/control design, the control group is also expected to contain individuals who will go on to become equivalent to cases; their presence in the control group reduces power. Environmental factors must be acknowledged in such studies as potential confounders; inevitably, the cases and controls have lived in different times and experienced different lifestyles. A way to mitigate some of these problems; however, is to choose controls that are no older than 50 (Halaschek-Wiener et al. 2008) because in modern day developed countries, mortality before age 50 is minimal. Choosing a comparison group <50years of age makes the control group essentially an unselected group with regard to mortality from age-related diseases. Choosing a control group in their 70 or 80, however, would exacerbate this issue, as the control group would fail to include individuals who died in their 50s or 60s.

Several studies, such as the Longevity Gene Study (Barzilai et al. 2001), the Leiden Longevity Study (LLS) (Mooijaart et al. 2011), the New England Centenarian Study (NECS) (Terry et al. 2004), and the Long Life Family Study (LLFS) (Newman et al. 2011) include comparisons of the offspring of LLI (who are assumed to have inherited some longevity factors) to contemporary age-matched controls. They have observed that the offspring of LLI have more favorable blood lipid profiles (Barzilai et al. 2001; Newman et al. 2011) and lower prevalence of hypertension and metabolic and cardiovascular disease (Atzmon et al. 2004; Westendorp et al. 2009; Newman et al. 2011) and all-cause mortality (Terry et al. 2004) than age-matched controls. Comparison of the offspring of LLI with their contemporaries controls for cohort effects such as variation in BMI in human populations over time; it has the limitation, however, of under-estimating the difference in phenotypes and genotypes that would presumably be observed if the LLI could be compared with their largely long-deceased birth cohort.

Linkage studies of long-lived sibships or extended pedigrees with exceptionally long-lived individuals have identified several putative and one replicated longevity linkage. In 2001, the NECS (Puca et al. 2001) reported a 10-cM sib-pair based linkage scan of 308 individuals in 137 sibships with exceptional longevity (defined as having a proband of at least 98 and a 91-year-old male or 95-year-old female sib). They found significant evidence for linkage of longevity to a region around D4S1564. Suggestive support for this region was obtained through analysis of 95 concordant pairs of fraternal male twins with a wellness phenotype (age at least 70 with no overt CVD or prostate cancer) (Reed et al. 2004). Initial convergence of two linkage studies with very different phenotypes led to excitement about this region and its suspected role in longevity and health. Subsequent study of the region focused in part on a regional biological candidate gene, microsomal triglyceride transfer protein (MTP), identified through haplotype analysis (Geesaman et al. 2003).

In 2010 a larger and higher density linkage study (Boyden and Kunkel 2010) expanded on the initial NECS resource, with a genome-wide linkage study of 279 families with multiple long-lived sibs 90years and older, including 129/137 of those previously described (Puca et al. 2001). A limitation of this study was the use of expected life span (estimated from age- and gender-specific life expectancies) for the 70% of subjects who were still living. This analysis of 9,751 SNPs found just-significant LOD scores at 3p22-24 and 9q31-34, as well as modest evidence for linkage at the original site, 4q22-25 and possibly at 12q. A larger study (Kerber et al. 2012) replicated the linkage of 3p22-24 to extreme longevity and identified possible additional loci. Working with 732 subjects from the Utah population database and database and population controls, including 433 Caucasian individuals aged 86109 who showed a phenotype including both excess individual longevity (the difference between observed and expected lifespan) and excess familial longevity (a weighted average of excess longevity for all family members), they used a linkage screen with 1,100 microsatellite markers to identify a strongly suggestive peak at 3p at the same position as Boyden and Kunkel. Meta-analysis of linkage in the Utah and New England data sets supported linkage at the chromosome 3 locus. Other linkage peaks were observed in the Utah data at 18q23-24, 8q23, and 17q21; meta-analysis provided additional support but not outright replication for 8q, 9q, and 17q. The new data; however, did not support linkage to chromosome 4 or chromosome 12. Larger sample sets and denser and more informative linkage analyses were pointing away from the original chromosome 4 linkage observation, and converging instead most strongly at 3q24-22.

Two linkage studies of successful aging in Amish individuals over 80years of age within a single 13-generation pedigree showed linkage to chromosomes 6, 7, and 14, different regions than those found in the longevity linkage studies. Successful aging was defined as cognitively intact and without depression, high functioning, and satisfied with life. These studies (Edwards et al. 2011; Edwards 2012) analyzed 263 cognitively intact Amish over 80years old (74 successfully aged and 189 normally aged) within 12 sub-pedigrees using 630,309 autosomal SNPs. Linkage was found at 6q25-27, as well as association of a SNP, rs205990, in the interval linked to the successfully aged phenotype. The chromosome 6 linkage identified in the Amish is different from those identified in the Utah and New England studies; this may reflect the different phenotype, or may be due to genetic factors specific to the Amish founder population.

The largest linkage study to date was done in the multi-site European Genetics of Healthy Aging (GEHA) Study, which looked at 2118 European full sib-pairs over 90years old (Beekman et al. 2013). GEHA found linkage at 4 regions: 14q11.2, 17q12-q22, 19p13.3-p13.11, and 19q13.11-q13.32. The chromosome 14 linkage is at a different site from that observed in the Amish study; the large chromosome 17 region overlaps the 17q21 locus observed by Kerber et al. Fine mapping of these linkage regions using GWAS data in a subset of 1228 unrelated nonagenarians and 1907 controls identified a SNP near APOE at the 19q locus as significantly associated with longevity. Apolipoprotein E (apoE) isoforms are known risk factors for cardiovascular disease (CVD) and Alzheimer disease (AD), likely due to their involvement in inflammation, elevated lipid levels, and oxidative stress (Huebbe et al. 2011). ApoE has three main isoforms: apoE2, apoE3 and apoE4. Combined modeling in the GEHA study showed that APOE4 (p=0.02) and APOE2 (p=1.0105) account for the linkage at 19q. The APOE linkage was characterized by absence of APOE4, but enrichment for APOE2 among the nonagenarians. In this study the APOE2 allele is the stronger association, and the authors refer to APOE as a longevity gene.

The multiple linkage signals observed in these studies likely indicate genetic heterogeneity of longevity and healthy aging in human populations. Interestingly, the GEHA study observed heterogeneity among its multiple geographic regions; Northern European subjects contribute most to some of the linkage peaks they observe, including the APOE locus. Gender-specific effects were also observed, with a male-specific linkage peak at 8p and female-specific ones at 15q and the 19q APOE locus (Beekman et al. 2013). While the lack of association at the other linkage regions in the GEHA study may be due to power limitations, it could also imply that multiple rare or private variants contribute to linkage but not association at these loci.

Candidate genes examined for association with longevity or healthy aging or related phenotypes fall into several categories. They include genes nominated based on observations of lifespan extension in model organisms; and genes involved in lipid metabolism, immune response and inflammation, stress response, and others. Candidate genes tested for association with longevity and related phenotypes have been the subject of several excellent reviews (Christensen et al. 2006; Wheeler and Kim 2011; Ferrario et al. 2012; Newman and Murabito 2013); an exhaustive listing is beyond the scope of this review.

Of the candidate genes assessed for association with longevity, variants in APOE and FOXO3A have been most consistently replicated, though some candidate genes have been associated with longevity phenotypes in more than one population but not in all populations tested; many more have been associated in a single study but failed to replicate in others (reviewed by (Christensen et al. 2006)). In a study of 1,344 healthy Italians aged 2290, APOE4 was found at lower frequency and APOE2 at higher frequency in elderly and centenarians than in younger individuals (e.g., Seripa et al. 2006); APOE2 is a putative protective factor in this context and APOE4 can be considered a frailty allele (Gerdes et al. 2000). FOXO3A is a homologue of the C. elegans Daf-16 gene that is important in control of lifespan in the worm (Hsin and Kenyon 1999); it is part of the insulin/IGF1 signaling pathway. FOXO3A variants have been associated with longevity in many populations (reviewed in Wheeler and Kim 2011).

Additional genes show promise of great relevance to healthy aging. A variant at CETP, for example, though inconsistently associated with longevity in different populations (reviewed by (Christensen et al. 2006)), in 213 Ashkenazi Jewish individuals of average age 98 is associated not only with longevity but also with additional aging-related phenotypes including a desirable lipid profile (Barzilai et al. 2003) and preservation of cognitive function (Barzilai et al. 2006). Other recent studies with extensive replication data are also encouraging. Association of a SNP in a heat shock factor gene, HSF2 with all-cause mortality was seen in the longitudinal Rotterdam Study (5,974 participants and 3,174 deaths), with replication in eight population-based cohorts (Broer 2012).

Other candidate genes have been associated with longevity or healthy aging phenotypes in some but not all studies. MTP, identified as a regional candidate at the 4q25 locus, failed to show replication of association with longevity in larger studies of approximately 1500 LLI each (Beekman et al. 2006; Bathum et al. 2005; Nebel et al. 2005). Progeria genes have shown association with longevity in some studies. A haplotype of SNPs at LMNA, the gene that is mutated in Hutchinson-Gilford progeria, was associated with long life (age >95years) in 873 LLI and 443 controls, and remained significant upon meta-analysis of 3,619 subjects from four independent samples (Conneely et al. 2012). Polymorphisms at WRN have shown inconsistent associations with age (Castro et al. 2000; Kuningas et al. 2006). Sirtuins mediate the effects of caloric restriction, a non-genetic factor known to increase life span in many organisms. The effect of polymorphisms in sirtuin genes (SIRT1-7) on longevity and age-related diseases was reviewed by Polito et al. (2010). There is evidence that variants in SIRT3 (Rose et al. 2003) are associated with longevity. A functional promoter variant at DNA repair gene EXO1 was associated with longevity in female centenarians (Nebel et al. 2009), but tagSNPs in the gene showed no association with longevity in men (Morris 2013).

Given the multifactorial nature and likely genetic heterogeneity of healthy aging and longevity, as well as environmental influences on these complex traits, it may not be reasonable to expect that replication of candidate gene studies would be uniform between populations. Reasons for lack of replication include limitations of sample size, rarity (low minor allele frequency) of actual variants, and small true effect sizes. Poorly designed or under-powered studies will result in false positives that legitimately fail to replicate. For studies of longevity and healthy aging, in particular, differences in phenotype or type of study will also result in findings that are non-uniform between studies. While larger case/control studies are frequently suggested as a solution to the limitations of present-day association studies, combining data from populations with different lifestyles and genetic backgrounds, even if well-matched for ethnicity, may obscure true association signals.

To date, SNPs in or near APOE are the only ones to achieve genome-wide significance (GWS, generally p5108) in genome-wide association studies (GWAS) of lifespan-related traits. In three GWAS of long-lived individuals vs. younger controls, APOE was significantly associated with longevity at the genome-wide level. The first of these included 763 long lived (94110years) and 1,085 control (4577years) from German biobanks and replication in an independent set of German samples (754 cases aged 95108, 860 controls aged 6075) (Nebel et al. 2011). Only rs4420638 near APOC1 and in linkage disequilibrium (LD) with APOE achieved GWS. GWAS of 403 unrelated nonagenarians (average age 94) from longevity families in the LLS vs. 1,670 controls (average age 58) showed similar results (Deelen et al. 2011a). Only one of 62 SNPs carried forward to meta-analysis with 4,149 nonagenarian cases and 7,582 younger controls from the Rotterdam study, the Leiden 85+ study and the Danish 1905 Cohort reached GWS, rs2075650 at TOMM40 near APOE. Meta-analysis of the APOE2 and APOE4 SNPs showed significant associations of both SNPs with longevity, with E2 being protective of long life (OR 1.31, CI 1.171.46, p=1.35106), and E4 being deleterious (OR 0.62, CI 0.560.68, p=1.331023). A third longevity GWAS (Sebastiani et al. 2012) included three phases: a discovery phase with 801 New England centenarians (aged 95119, many with a family history of extreme longevity) vs. 914 controls genetically matched by means of principal components analysis; a first replication in 253 centenarians (89114) vs. 341 genetically matched controls; and a second replication with 60 additional centenarians (100114) and unmatched controls. Of 243,980 SNPs analyzed only one, TOMM40 SNP rs2075650 near APOE, reached GWS. Inverse association of APOE4 with longevity (p=5.3103) was also detectable in the Southern Italian centenarians study (SICS) of 440 LLIs aged 90109 and 553 young controls aged 1845 (Malovini et al. 2011), despite the known lower frequency of the E4 allele in Southern, as compared with Northern, Europe (Haddy 2002).

Other GWAS of lifespan-related phenotypes revealed no associations that were significant at the genome-wide level. A GWAS of the Framingham health study (Lunetta et al. 2007) (258 Original Cohort and 1,087 Offspring individuals, members of the 330 largest families in the study) revealed no GWS SNPs for any of five aging-related phenotypes. Newman et al. (Newman et al. 2010) meta-analyzed four cohort studies in the cohorts for heart and aging research in genomic epidemiology (CHARGE) Consortium for survival to at least 90years of age. Cases were 1,836 people who achieved survival to at least 90; controls were 1,955 participants who died aged 5580. SNPs were genotyped and imputed in subjects of European ancestry, with systematic elimination of outliers and correction for population stratification. Replication was carried out in the LLS (950 long-lived probands and 744 partners of their offspring and 680 blood bank donors) and the Danish 1905 Cohort Survey (2,262 long-lived participants and 2007 Danish twin study controls aged 4668). No SNPs reached genome-wide significance.

Walter et al. (2011) conducted a meta-analysis of GWAS of nine longitudinal cohort studies in the CHARGE Consortium, including 25,000 unselected people of European ancestry. They analyzed two continuous traits, all-cause mortality, and event-free survival (where event was defined as myocardial infarction, heart failure, stroke, dementia, hip fracture, or cancer). No SNPs reached GWS for either phenotype. SNPs near APOE reached only nominal significance in the CHARGE study (Walter et al. 2011), in contrast to the results of GWAS of centenarians, in which APOE has been a significant and replicable finding. The CHARGE meta-analysis contained few extremely old individuals, and so in comparison with centenarian studies or those targeting long-lived healthy individuals, has examined earlier mortality and events, a different phenotype. The Framingham Study GWAS (Lunetta et al. 2007), which also showed no GWS SNPs also represents a much younger group, on average, than studies of oldest old or centenarians. This may mean that different genes and variants may come into play in different phases of aging, with APOE being most relevant at older ages. Earlier mortality is often related to lifestyle as well, and the heritability of aging is lower at younger ages, as described above.

A genome-wide association study of copy number variants (CNVs) in the Rotterdam study RS1 cohort, with replication in the RS2 cohort and the FHS, found that large common deletions are associated with mortality (vs. survival) at old age (Kuningas et al. 2011a). They tested 312 common CNV regions and measures of CNV burden for association with mortality during follow-up. A higher burden of CNVs of 500kb or more in size was associated with mortality. Two specific regions were also associated with mortality, 11p15.5 and 14q21.3. The 11p15.5 association, which would survive Bonferroni correction for 312 tests, includes insertions and deletions which were analyzed together relative to non-carriers; it contains 41 genes including some related to longevity or complex diseases. The 14q21.3 region contains no genes and is characterized only by deletions. Runs of homozygosity, which can indicate presence of recessive loci, were not associated with survival to old age in this cohort (Kuningas et al. 2011b).

Analyses of phenotypes that may influence long-term good health have also been undertaken. Personality traits are associated with healthy aging and longevity (Terracciano et al. 2008). In the LLFS, a GWAS of five personality factors in 583 families with 4,595 individuals and replication in 1,279 other subjects identified a locus associated with agreeableness, and identified several significant ageSNP interactions that may affect longevity through effects on personality (Bae et al. 2013).

In contrast to the results of longevity GWAS, GWAS of common complex diseases have revealed hundreds of SNPs associated with cancers, CVD, diabetes and other age-related diseases, albeit with increasing numbers of associations found with increasing GWAS size. One explanation for this may be that the phenotypes of healthy aging and longevity may be much more complex than those of these complex diseases, in part because they often (depending on phenotype definition) involve absence of specific complex diseases. If GWAS studies of survival to elderly ages are even more confounded by environmental (E) factors than GWAS of diseases, combining studies from different populations in pooled or meta-analyses may complicate the E effects even more. In studies of older individuals, it is particularly hard to control for E factors experienced over many decades of life.

SNPs at only one locus, APOE, have achieved Bonferroni-corrected levels of GWS in GWAS of longevity. By current standards these GWAS, which involved fewer than 1,000 centenarians, or a few 1,000 nonagenarians, are modest in size. Larger GWAS may in theory allow additional SNPs to achieve this threshold. There are other indications, however, that support the idea that SNPs that do not reach this threshold of GWS may be biologically important, either individually or through their joint effects. Several studies used a variety of techniques to analyze collections of nominally longevity-associated SNPs to determine if they act in concert to affect lifespan.

In the Framingham study GWAS of 5 aging-related phenotypes (Lunetta et al. 2007) observed that SNPs in some candidate genes, including SNPs near the Werner syndrome gene WRN and FOXO1A, as well as GAPDH, KL, LEPR, PON1, PSEN1, and SOD2 were associated with age at death. Kulminski and Culminskaya (2011) used Framingham Affymetrix 50K SNP data to perform GWAS of four endophenotypes (CVD, cancer, systolic blood pressure, and total cholesterol) to identify 63 SNPs that were associated at p<106 with at least one endophenotype. 76 genes at or near these SNPs were enriched in terms of Gene Ontology annotations related to aging-relevant processes. Yashin et al. (2010) hypothesized that lifespan depends on the number of small-effect longevity alleles present in individual genomes. They re-analyzed Framingham 550K SNP data and identified 169 SNPs associated at p<106. The number of these SNPs carried by an individual correlated with lifespan and explained 21% of its variance; in contrast, randomly chosen SNPs did not correlate with lifespan.

Gene set analysis of GWAS data from the LLS and Rotterdam studies was used to show that genes in the insulin/IGF-1 signaling (IIS) and telomere maintenance TM pathways are associated with longevity (Deelen 2011b). 1021 and 88 GWAS SNPs were identified within 10kb of 68 IIS and 13 TM genes, respectively. Both pathways were associated with longevity. Nine IIS genes (AKT1, AKT3, FOXO4, IGF2, INS, PIK3CA, SGK, SGK2, and YWHAG) and one TM gene (POT1) were the main determinants of the association.

Sebastiani et al. (2012) constructed a model in which 281 SNPs showed 89% sensitivity and 89% specificity to predict longevity in their GWAS Discovery set, and 5861% specificity and 5885% sensitivity in independent sets. They call this a genetic signature of exceptional longevity. These SNPs explain nearly 20% of the heritability of extreme longevity. They find that the TOMM40 SNP near APOE alone has poor predictive value; removing it from the model reduces specificity and sensitivity by only 1%. The 281 SNPs include 137 in 130 genes, including LMNA, WRN, SOD2, CDKN2A, SORCS1 and SORCS2, and GIP. This set of 130 genes is highly and significantly enriched for those related to Alzheimer disease (38 genes), 42 related to dementia, 38 to tauopathies, 24 to CAD, and several to neoplasms.

GWAS of the SICS Study of 410 LLI and 553 younger controls identified 67 SNPs that reached a permutation-defined level of genome-wide significance of p<104 (Malovini et al. 2011). Among them was rs10491334 at the calcium/calmodulin-dependent protein kinase IV (CAMKIV) that replicated in 116 additional LLI and 160 controls. Malovini et al. demonstrate that CAMK4 phosphorylates and activates survival proteins FOXO3A, AKT, and SIRT1. Homozygous carriers of the minor allele had lower CAMKIV protein expression and were under-represented among LLIs, consistent with a deleterious effect of this allele on longevity.

The biological relevance of other SNPs besides those at APOE is also strongly supported by similarities between the results of human GWAS and mouse lifespan studies. Eight of the ten top CHARGE SNPs detected by GWAS, but which did not achieve GWS, correspond to mouse lifespan quantitative trait loci (QTL) (Murabito et al. 2012). These studies connect GWAS findings that do not reach GWS with many genes that are relevant to aging or age-related diseases. In several cases, this convergence with genes of biological interest is statistically unlikely to be due to chance and is likely to reflect the presence of true association signals that are not consistent enough to be replicated predictably as candidate genes or achieve GWS, or have effects that are too subtle to be detected individually. Such potential true signals may be more affected by E factors than those that have been replicated, i.e., APOE and FOXO3A. As pointed out by Yashin et al., the same sets of variants would not be expected to work in all populations because of differences in environment (Yashin et al. 2010).

Several recent studies have shown that centenarians do not carry smaller numbers of risk alleles for common complex diseases than average people. In an important paper in 2010, Beekman et al. (2010) studied two case/control collections: (1) 723 nonagenarian siblings (mean age 94) from the LLS vs. 721 unrelated younger controls (mean age 52), and (2) 979 long-lived individuals over 85 (mean age 87) from the pop-based Leiden 85+ study vs. 1,167 younger controls (mean age 41) from the Netherlands Twin Register. They looked at 30 SNPs known to be associated with CVD, cancer or type 2 diabetes (T2D). The cases and controls each carried an average of 27 disease risk alleles. The distribution of risk alleles was the same in elderly and young subjects. Beekman et al. note that GWAS-identified disease risk alleles do not compromise human longevity and suggest that a lack of rare disease factors, or the presence of protective factors, is at work in the long-lived individuals. It is important to note, however, that CVD, cancer, and T2D are diseases that have very clear lifestyle components and that part of the effect could be due to lifestyle differences.

Mooijaart et al. (2011) extended this observation the following year, showing that SNPs associated with T2D and identified by GWAS are not major determinants of the beneficial glucose tolerance that characterizes familial longevity. They compared the offspring of the LLS long-lived individuals with the offsprings spouses and other controls. The LLS offspring had a better metabolic profile and better glucose tolerance than same-age controls, although the frequency of 15 known T2D SNPs did not differ between the two groups. When individuals were compared within each group, however, glucose levels did correlate with the number of T2D SNPs. They speculate that the LLS offspring may have protective factors that improve their metabolic profile and glucose tolerance in spite of the presence of T2D GWAS SNPs. This comparison, using same-age groups of individuals, clearly points to protective genetic factors contributing to preservation of a healthy phenotype, rather than lifestyle and environmental factors that should be very similar (at least in adulthood) between the offspring and their spouses.

Sebastiani et al. (2012) also noted that there was not a substantial difference in the numbers of 1,214 known disease-associated SNPs in centenarians and controls. A similar observation was made in their whole genome sequence data from one male and one female supercentenarian (Sebastiani et al. 2011).

These important and perhaps surprising results show that extreme longevity, and the long-term good health that often accompanies it, is not incompatible with the presence of many disease risk alleles. At least for the common SNPs associated with common complex diseases, it is not the absence of bad alleles, but more likely the presence of good alleles that influences longevity, though effects of good environmental factors may also contribute. Protective factors of some kind may allow these risk variants to not be manifest. These results also have implications beyond the study of longevityin an age when substantial effort is being invested in personalized disease risk prediction, the presence of many disease alleles that are non-penetrant in some individuals potentially complicates predictions of disease.

One mechanism for a lack of effect by an undesirable allele is the buffering mechanism explored by Barzilai et al. They propose that some individuals who show exceptional longevity may do so despite the presence of unfavorable alleles because those alleles are buffered by favorable alleles in other genes (Bergman et al. 2007). They suggest that buffering gene variants (longevity variants) will show a monotonic increase in frequency from early old age (65) to later ages; examples of buffering genotypes are CETP VV, APOC3 CC, and a +2019 deletion in ADIPOQ. Buffered alleles, in contrast, should show a U-shaped frequency curve, higher at younger ages, dipping low in early old age, and then increasing in the exceptionally old (who have the buffering protective genotype that allows disease-related variants to accumulate); examples of buffered genotypes are heterozygotes for deleterious alleles of KLOTHO and LPA. Importantly, Bergman et al. use a cross-sectional study design, with 1,200 subjects in their 611th decades of life to show experimental support for the buffering hypothesis; their data support the idea that CETP VV genotype buffers the deleterious effects of an LPA genotype. They show a genetic interaction between CETP genotype and LPA; LPA heterozygotes with the CETP IV/II genotypes monotonically decrease in frequency with age, but those in CETP VV individuals increase from age 70 onward. They argue that case/control analyses are insufficient to reveal this effect because it does not reveal the shape of the allele frequency age curve.

Earlier observations are also explained by a buffering mechanism. De Benedictis et al. (1998) described an age-related convex trajectory of a 3APOB-VNTR genotype that they interpret as consistent with crossing mortality curves relevant to subgroups of individuals with different genotypes. A X-sectional study of 800 healthy aging subjects from 18 to 109years free of clinically apparent disease genotyped variants in APOA1, APOC3, and APOA4 (Garasto et al. 2003). They noted that an allele of APOA1 that correlated with higher serum LDL-C was paradoxically increased in frequency in the oldest old. The authors called it another genetic paradox of centenarians. While this observation could reflect population stratification in the different age groups, it may also be due to the U-shaped curve of a buffered gene.

The buffering mechanism may also explain some of the inconsistency in the findings for MTP. Huffman et al. (2012) find that MTP CC is a deleterious genotype that is buffered by any of three longevity genotypes of CETP, APOC3, or ADIPOQ. MTP CC shows a U-shaped curve, declining ages 5585, and then dramatically increasing in those who live 90 or more years. If this MTP genotype is observed at high frequency in centenarians, but only in the presence of specific protective variants, this may in part explain why the linkage at chromosome 4 was not observed consistently between studies.

Buffering has been described in model organisms. The heat-shock protein Hsp90 is known to buffer genetic variation in Drosophila, allowing it to accumulate under neutral conditions (Rutherford and Lindquist 1998). Such a gene is known as a phenotypic capacitor, and it masks the presence of phenotypic variation. It is interesting to speculate that protective genetic variants carried by centenarians may be capacitors for the disease risk variants we now know they carry at, on average, the same frequency as other people. Identification of buffering/capacitor genes and study of their function will be necessary to understand the longevity phenotype. It will also be important to determine if such capacitors operate in healthy aging as well as extreme longevity. Because such variants are likely rare, intensive study of rare individuals at the upper ends of the human lifespan and healthspan, perhaps by whole genome sequencing and examination of unusual variants they carry, is paramount.

The interaction between buffering and buffered genes and genotypes also has implications for study design. The exquisite studies carried out by Barzilais group are done in a single well-defined ethnicity, Ashkenazi Jewish individuals. Since a buffered gene will only show a distinctive U-shaped curve in the presence of its buffer, and a buffer may only be advantageous in the presence of a deleterious gene that it buffers, this underscores the importance of avoiding population stratification in such studies. If some of the associations detected to date in case/control studies of healthy aging and longevity are actually underlain by genotypes with U-shaped curves, the choice of ages for the cases and controls will greatly affect whether an association is detected, and may explain some failures of associations to replicate. Finally, the concept of buffering genes has implications for the use of centenarians, or exceptionally healthy elderly individuals as super-controls for disease studies; if the exceptional elderly are healthy because of a protective factor rather than lack of a disease allele, their use as an extreme comparison group may not necessarily be helpful.

Given that lifestyle is expected to have a greater impact than genetics on healthy aging, it seems unlikely that differences in lifestyle are not confounding association studies of longevity and healthy aging. It is challenging to quantify lifestyle in an optimal comparison group for, for example, centenarians. Younger control groups inevitably have different lifestyles than the elderly had at their age. For example, the CHARGE consortium (Newman et al. 2010), which compared individuals who survived to at least 90 to those who died aged 5580, found that the younger controls had higher rates of smoking.

The Longevity Gene Study overcame the birth cohort limitation using pre-existing lifestyle data from 3,164 NHANES controls of the same birth cohort as 477 Ashkenazi Jewish individuals aged 96109 (Rajpathak et al. 2011). They found no obvious differences in lifestyle and suggested that the long-lived individuals may interact with lifestyle factors differently than others. This study, however, did note subtle differences between the long-lived and comparison groups. They saw significantly fewer obese men, more overweight women, and fewer obese women in the long-lived group; in addition, more control men smoked. These differences, combined with recall limitations of the long-lived group, imply that this analysis may have missed many small lifestyle differences that could add up to substantial health differences. It will likely be difficult to take into account all but the largest lifestyle factors when planning GxE studies of longevity and healthy aging. Biomarkers of exposures may vary not only with exposure but also over time, complicating the use of such methods for these phenotypes.

Mitochondria are thought to be important to aging due to their key roles in oxidative phosphorylation, cell metabolism, and apoptosis. A relationship of variation in the mitochondrial genome with health and/or longevity is implied by the observation that age at death correlates more closely with the age at death of a persons mother more so than that of the father (Brand et al. 1992). Associations of mitochondrial genome sequence variants or haplogroups (combinations of specific variants that correlate with specific populations) with healthy aging or longevity have been noted in many populations including, for example, Italian (De Benedictis et al. 1999), Japanese (Tanaka et al. 1998), Amish (Courtenay et al. 2012), Chinese Uygur (Ren et al. 2008), Costa Rican (Castri et al. 2009), Ashkenazi Jewish (Iwata et al. 2007), Irish (Ross et al. 2001), and Finnish individuals (Niemi et al. 2003). The associations observed are inconsistent between populations and do not involve the same variant or haplogroup. This lack of consistency may be due in part to the relatively small size of many of these studies. Three common problems have been noted about such studies: inadequate matching of cases and controls, inadequate correction for multiple tests, and undetected population stratification (Shlush et al. 2008).

Interestingly, when the frequencies of different mitochondrial haplogroups are plotted for Italian individuals aged 20 to over 100, the curve shapes observed include monotonic increase for haplogroup J, and a U-shaped curve for haplotype H (de Benedictis et al. 2000), reminiscent of the longevity and buffered variants described earlier. A variant at the origin of replication of the mitochondrial heavy strand, C150T, has been observed at higher frequencies in centenarians, both through inheritance and through somatic increase in frequency, with some individuals achieving homoplasmy for this variant in their lymphocytes and monocytes, but not in granulocytes; a selective advantage of achieving high frequency of this variant in at least some cell types has been suggested (Zhang et al. 2003). Interactions between nuclear genome variants and both inherited and somatic mitochondrial variants have also been suggested to play a role in aging and longevity (Santoro et al. 2006; Tranah 2011).

Sebastiani et al. (2011) recently reported the whole genome sequencing of one male and one female supercentenarian of European ancestry from the NECS. The genomes of these exceptionally long-lived individuals were similar, in terms of the rate of nonsynonymous SNPs and number of indels, to other genomes sequenced to date. They have a similar number of known disease-associated variants to other genomes showing that their exceptional lifespan does not seem to be due to lack of known disease-associated variants. It is possible, though, that they failed to inherit a combination of variants that would have acted together to cause disease. Both supercentenarians lacked APOE4 alleles. They do not carry most of the longevity variants reported previously in the literature, implying that these known variants are not necessary for longevity. It is possible that they carry as yet undiscovered protective variants. One per cent of the variants observed were novel. Interestingly, an excess of coding region variants was seen in genes closest to GWAS-identified longevity variants, an observation that supports the idea that rare variants of these genes may contribute to the longevity phenotype.

Telomeres are indisputably important to aging. Telomeres shorten with age and are considered to be a biomarker of age. The role of telomere biology in healthy aging and disease was recently reviewed (Zhu et al. 2011). Leukocyte telomere length (LTL) has been correlated with measures of health and ability in elderly individuals. In a community-based cohort of 70- to 79-year-olds, LTL was associated with more years of healthy life; LTL was suggested to be a biomarker of healthy aging (Njajou et al. 2009). Louisiana Healthy Aging Study results concurred with this observation; LTL was correlated with measures of healthy aging in an age-dependent way (Kim et al. 2012). LTL was also found to correlate positively with physical ability (but not cognitive function) in Danish twins aged at least 77years (Bendix et al. 2011) and inversely with disability in American seniors (Risques et al. 2010). Ashkenazi centenarians and their offspring also showed longer telomeres, for their age, than controls; longer telomeres correlated with less disease (Atzmon et al. 2010). In contrast, in a study of Canadian Super-Seniors (individuals aged at least 85 and never diagnosed with cancer, cardiovascular disease, Alzheimer disease, major pulmonary disease or diabetes) the healthy oldest-old did not have exceptional telomere length for their age, but showed less variability in telomere length than mid-life controls, implying that they may be selected for optimal rather than extreme telomere length (Halaschek-Wiener et al. 2008).

Variation in genes involved in telomere maintenance has also been associated with longevity. One SNP at SIRT1 (Kim et al. 2012) and one in TERC (Soerensen et al. 2012) are associated with both LTL and longevity. Detailed analysis of TERT and TERC in Ashkenazi centenarians showed an excess of genetic variation in both genes in the centenarians and identified a TERT haplotype associated with extreme longevity (Atzmon et al. 2010). Gene set analysis of GWAS data also supported the relevance of telomere maintenance (Deelen et al. 2013). Overall, the relationship between telomeres, aging, healthy aging, and longevity is multi-layered. Telomere maintenance is an important process in aging, and also a biomarker of it. LTL is a biomarker of aging and of healthy aging. Variation in telomere maintenance genes appears to affect both telomere length, and life span and health span in humans.

Two recent large-scale analyses of data from GWAS studies have established that mosaicism for large genomic alterations increases with age (Laurie et al. 2012; Jacobs et al. 2012). In one study, data for 50,222 subjects found that <0.5% of people aged <50, and 23% of elderly (2.7% in subjects >80years), have detectable mosaicism in peripheral blood. Age was a significant predictor of mosaic status, but sex, ancestry, and smoking status were not. The second study used data from 31,717 cancer cases and 26,136 controls from 13 GWAS studies and found detectable clonal mosaicism in 0.87% of individuals. In the cancer-free controls, they found mosaicism in 0.23% of those <50years old and in 1.91% of those aged 7579, a significant difference (p=4.8108). Somatic mosaicism (heteroplasmy) of the mitochondrial genome also increases over the lifespan (Sondheimer et al. 2011). Of course, telomere shortening is another somatic genomic change that occurs over the human lifespan. Such somatic changes are both a genetic aspect of aging and an aging-related phenotype.

Epigenetics, at the interface between the genome and the environment, is emerging as an important factor in longevity, and has been the subject of recent excellent reviews (Gravina and Vijg 2010; Ben-Avraham et al. 2012). Methylation patterns change with age, and discordance in methylation between MZ twins also increases with age (Talens et al. 2012), an observation consistent with the effect of environment and lifestyle on the epigenome. Studies of DNA methylation support the idea that aging is associated with a relaxation of epigenetic control and that this epigenetic drift may affect the development of aging-related diseases (Gravina and Vijg 2010). An epigenome-wide association scan (EWAS) identified age-related differentially methylated regions as well as differentially methylated regions associated with age-related phenotypes (Bell et al. 2012). Whole genome bisulfite sequencing of DNA from CD4+ T cells of a centenarian and a newborn identified differentially methylated regions that were usually hypomethylated and less correlated with methylation of adjacent CpG dinucleotides in the centenarian (Heyn et al. 2012). These results support the idea that small cumulative DNA methylation changes accumulate over a lifetime. Age-related temporal changes in DNA methylation also show significant familial clustering, indicating that methylation maintenance is a familial trait (Bjornsson et al. 2008). A study of DNA methylation in centenarians and their offspring compared with the offspring of non-long-lived individuals and young individuals showed that the offspring of the centenarians delay age-related methylation changes (Gentilini 2012). A landmark paper by Hannum et al. (2013) offers an explanation for this familiality. They used methylome analysis to compare human aging rates in individuals of age 19101 and identify methylation QTLs (meQTLs) (including one at methyl-CpG binding domain protein 4) that affect it. Indeed, trans-generational epigenetic inheritance of extended lifespan has been demonstrated in C. elegans (Greer et al. 2011).

It is likely that the effects of epigenetic changes manifest in part by effects on gene expression. Longevity-selected lines of Drosophila show gene expression profiles that are similar to younger control flies (Sarup et al. 2011). This type of observation is more difficult to make in humans, however. Several human studies have compared gene expression between LLI and younger individuals. Blood miRNA expression differences between LLI and younger controls identified genes known to be differentially expressed in age-related diseases (ElSharawy et al. 2012). This study design, however, does not allow discrimination between genes that are differentially expressed because they are involved in longevity, related to chronological age, or affected by environmental differences between the old and young groups. A cross-sectional analysis of individuals aged 5090, and centenarians, was used to identify a miRNA, miR-363*, whose expression declined with age but was preserved at youthful levels in the centenarians (Gombar et al. 2012). The Leiden Longevity Study, however, used LLI and their offspring to show that RPTOR in the mTOR pathway is differentially expressed between the offspring of the LLI and their spouses (Passtoors et al. 2013). The study design issues that are important to avoid confounding by lifestyle factors in studies of inherited factors will be even more important in gene expression studies. It seems likely that as yet unidentified genetic factors and lifestyle practices that help us maintain a favorable epigenetic profile and optimal gene expression will be important in longevity and healthy aging.

See the rest here:
Genetics of healthy aging and longevity | SpringerLink

Read More...

National Institutes of Health – SardiNIA

Thursday, September 21st, 2017

In a first survey, the project team recruited over 6,100 subjects from a catchment area including four towns in east-central Sardinia and assessed a first list of >200 traits. The baseline survey has been followed by follow-up visits that collected longitudinal data on the same traits collected at baseline but added assessment of frailty-related traits, namely measures of bone density and geometry, muscle strength, and gait speed, and additional cardiovascular measures (see below). In the course of SardiNIA3, along with the expansion of the cohort and the addition of more traits (see below), increases in testing efficiency and additional cost-sharing funds from Sardinian sources permit the completion of Fourth Visits for the entire cohort. Also in current actions, DNA sequencing has recovered essentially all of the genetic variation in the cohort, and further arrangements for an Outcome Study have also been made to be implemented in SardiNIA4, a further 5-year continuation of the Project that also projects Fifth Visits for the cohort.

The infrastructure for the clinic and phenotypic testing has been stable, with stringent quality control, which is reflected in the high quality of the database. The initial sample cohort included over 62% of the eligible population living in the region (age 14-102 years), and at least 96% of the initial cohort have all grandparents born in the same province. The initial group included 4,933 phenotyped sib pairs, 4,266 phenotyped parent-child pairs, >4,069 phenotyped cousin pairs, and >6,459 phenotyped avuncular pairs. Additional recruitment has increased the cohort substantially, and results have consistently shown that for essentially every trait, most of the associated genes and variants would be involved in determining variance in both young and old and in men and women. Thus, genetic analyses can draw on data from all ages and both genders.

The added value of studying a founder population has also been demonstrated by the extension of DNA analysis to the full range of variation by sequencing. The population has proven to contain the great bulk of variation found in other populations, but during its isolation over many thousands of years, many variants rare elsewhere have risen to relatively high levels on the island by drift or selection and others have newly arisen as Sardinian-specific. These have provided extensive new information about a whole range of traits and pathways (e.g., Nature Genetics November, 2015 articles and Editorial).

Sardinia also offers a special entre to the genetics of specific diseases that are especially prevalent in the founder population. This includes the anomalously high incidence of autoimmune diseases including Multiple Sclerosis and Type 1 diabetes, which interrupt the high to low gradient of incidence from Northern to Southern Europe. Again, this has fostered novel findings in causation and pathophysiology.

The founder population itself also contains within its DNA a record of human demography through history, which has permitted the inference of the timing of human population movements based on mitochondrial and Y chromosome analyses.

Regarding the course and mechanism of aging, the longitudinal study, now in its 15th year, focuses on residents of the cluster of towns to collect longitudinal information on more than 400 age-related quantitative traits ("endophenotypes" or "quantitative risk-related genetic or environmental factors") that can be scored on a continuous scale, as well as >200 dichotomous traits (including major diseases and risk factors such as smoking). The use of quantitative traits permits the study of the entire range of allelic variation in a population, with particular interest in a range of cardiovascular risk factors, anthropometric measurements, blood test values, facets of personality, and bone-density and frailty-related variables.

The longitudinal study of a broad range of phenotypes in a founder population is distinctive in this study, and stable environmental/epidemiological factors combined with the simplification of genetic analyses also aid in proposed joint investigations of relative risk. Furthermore, because we are collecting risk factor data, we can also analyze, in an Outcome Study, the prognostic power and/or pathophysiological relevance of earlier predictors for the onset of serious risk factors [e.g., increases in pulse wave velocity as a function of earlier (predictor) lipid and inflammatory markers].

See the rest here:
National Institutes of Health - SardiNIA

Read More...

Growth Hormone Receptor Gene Mutation Discovered – Anti Aging News

Friday, September 1st, 2017

A new study has discovered a genetic mutation on growth hormone receptors that they believe could expand our lifespan by an average of up to 10 years.

A new study led by Prof. Gil Atzmon head of the Laboratory of Genetics and Epigenetics of Aging and Longevity at the University of Haifa and his team have discovered a genetic mutation on growth hormone receptors that they believe could expand our lifespan by an average of up to 10 years. "We were aware before that variants involved with genetic paths related to the growth hormone are also associated with longevity. Now we have found a specific variant whose presence or absence is directly connected to it," Prof. Atzmon explained.

According to other research its well known that IGF-1 (insulin-like growth factor-1) contributes to longevity as well as associated with HGH and other growth hormones.

From theSciencegroup in the journalScience Advances,scientists have discovered a variation in genes that changes hormones in men that extend their life-span. Professor Nir Barzilai at Einstein, compared 3 groups of 100-year-old men from around the world with 100 American Jewish men with a control group of 70-year olds.

They discovered that men without Exon 3 on growth hormone receptor genes were more likely to live up to 10 years longer. According to Prof. Atzmon, this variant ensured longevity absolutely. "This study nicely wraps up the connection between growth hormone function and longevity. Our goal now is really to understand the mechanism of the variation we found, so that we can implement it and enable longevity while maintaining quality of life," Prof. Atzmon concluded.

Coincidently the research found that men with this genetic variation were also about 3 cm taller than their counter-parts. This is in opposition to other species in nature wherein that smaller of the strains usually live longer. This phenomenon holds true in insects, rodents and other mammals. In human males however the receptor anomaly caused less growth hormone absorption.

This research is very encouraging for World Health and A4M members. It further explains what we have hypothesized for years, that certain genetics allow for longer life. Our entire existence at A4M and World Health is devoted to Anti-Aging and how to enhance life and longevity.

Article by: Dr. Michael J. Koch, Editor withwww.WorldHealth.net and for Dr. Ronald Klatz, DO, MD President of the A4M has 28,000 Physician Members, has trained over 150,000 Physicians, health professionals and scientists in the new specialty of Anti-aging medicine. Estimates of their patients numbering in the 100s of millions World Wide that are living better stronger, healthier and longer lives.

See original here:
Growth Hormone Receptor Gene Mutation Discovered - Anti Aging News

Read More...

The Genetic Theory of Aging – Concepts and Evidence

Wednesday, August 30th, 2017

Your DNA may predict more about you than the way you look. According to the genetic theory of aging, your genes (as well as mutations in those genes) are responsible for how long you'll live. Here's what you should know about genes and longevity, and where genetics fits in among the various theories of aging.

The genetic theory of aging states that lifespan is largely determined by the genes we inherit.

According to the theory, our longevity is primarily determined at the moment of conception, and is largely reliant on our parents and their genes.

The basis behind this theory is that segments of DNA that occur at the end of chromosomes, called telomeres, determine the maximum lifespan of a cell. Telomeres are pieces of "junk" DNA at the end of chromosomes which become shorter every time a cell divides. These telomeres become shorter and shorter and eventually the cells cannot divide without losing important pieces of DNA.

Before delving into the tenets of how genetics affects aging, and the arguments for and against this theory, it's helpful to briefly discuss the primary categories of aging theories and some of the specific theories in these categories. At the current time there is not one theory or even one category of theories which can explain everything we observe in the aging process.

There are two primary categories of aging theories which differ fundamentally in what can be referred to as the "purpose" of aging. In the first category, aging is essentially an accident; an accumulation of damage and wear and tear to the body which eventually leads to death. In contrast, programmed aging theories view aging as an intentional process, controlled in a way that can be likened to other phases of life such as puberty.

Error theories include several separate theories including:

Programmed theories of aging are also broken down into different categories based on the method in which our body's are programmed to age and die.

There is significant overlap between these theories and even categories of aging theories.

Before discussing the key concepts related to aging and genetics, let's review what our DNA is and some of the basic ways in which genes affect our lifespan.

Our genes are contained in our DNA which is present in the nucleus (inner area) of each cell in our bodies. (There is also mitochondrial DNA present in the organelles called mitochondria which are present in the cytoplasm of the cell.) We each have 46 chromosomes making up our DNA, 23 of which come from our mothers and 23 which come from our fathers. Of these, 44 are autosomes, and two are the sex chromosomes, which determine if we are to be male or female.

(Mitochondrial DNA, in contrast, carries much less genetic information and is received from only our mothers.)

Within these chromosomes lie our genes, our genetic bluepirint responsible for carrying the information for every process which will take place in our cells. Our genes can be envisioned as a series of letters which make up words and sentences of instructions. These words and sentences code for the manufacturing of proteins which control every cellular process.

If any of these genes are damaged, for example, by a mutation which alters the series of "letters and words" in the instructions, an abnormal protein may be manufactured, which in turn, performs a defective function.

If a mutation occurs in proteins which regulate the growth of a cell, cancer may result. If these genes are mutated from birth, various hereditary syndromes may occur. For example, cystic fibrosis is a condition in which a child inherits two mutated genes controlling a protein which regulates channels responsible for the movement of chloride across cells in the sweat glands, digestive glands, and more. The result of this single mutation results in a thickening of mucus produced by these glands, and the resultant problems which are associated with this condition.

It doesn't take an elaborate study to determine that our genes play at least some role in longevity. People whose parents and ancestors have lived longer, tend to live longer and vice versa. At the same time, we know that genetics alone are not the sole cause of aging. Studies looking at identical twins reveal that there is clearly something else going on; identical twins who have identical genes do not always live an identical number of years.

Some genes are beneficial and enhance longevity. For example, the gene that helps a person metabolize cholesterolwould reduce a person's risk of heart disease.

Some gene mutations are inherited, and may shorten lifespan. However, mutations also can happen after birth, since exposure to toxins, free radicals and radiation can cause gene changes. (Gene mutations acquired after birth are referred to as acquired or somatic gene mutations.) Most mutations are not bad for you, and some can even be beneficial. That's because genetic mutations create genetic diversity, which keeps populations healthy. Other mutations, called silent mutations, have no effect on the body at all.

Some genes, when mutated are harmful, like those that increase the risk of cancer. Many people are familiar with the BRCA1 and BRCA2 mutations which predispose to breast cancer. These genes are referred to as tumor suppressor genes which code for proteins that control the repair of damaged DNA (or the elimination of the cell with damaged DNA if repair is not possible.)

Various disease and conditions related to heritable gene mutations can directly impact lifespan. These include cystic fibrosis, sickle cell anemia, Tay-Sachs disease and Huntington's disease, to name a few.

The key concepts in genetics and aging include several important concepts and ideas ranging from telomere shortening to theories about the role of stem cells in aging.

Telomeres - At the end of each of our chromosomes lies a piece of "junk" DNA called telomeres. Telomeres do not code for any proteins but appear to have a protective function, keeping the ends of DNA from attaching to other pieces of DNA or forming a circle. Each time a cell divides a little more of a telemore is snipped off. Eventually. there is none of this junk DNA left, and further snipping can damage the chromosomes and genes so that the cell dies.

In general, the average cell is able to divide 50 times before the telomere is used up (the Hayflick limit). Cancer cells have figured out a way to not remove, and sometimes even add to, a section of the telomere. In addition, some cells such as white blood cells do not undergo this process of telomere shortening. It appears that while genes in all of our cells have the code word for the enzyme telomerase which inhibits telomere shortening and possibly even results in lengthening, the gene is only "turned on" or "expressed" as geneticists say, in cells such as white blood cells and cancer cells. Scientists have theorized that if this telomerase could somehow be turned on in other cells (but not so much that their growth would go haywire as in cancer cells) our age limit could be expanded.

Studies have found that some chronic conditions such as high blood pressure are associated with less telomerase activity whereas a healthy diet and exercise are linked with longer telomeres. Being overweight is also associated with shorter telomeres.

Longevity genes - Longevity genes are specific genes which are associated with living longer. Two genes that are directly associated with longevity are SIRT1 (sirtruin 1) and SIRT2. Scientists looking at a group of over 800 people age 100 or older, found three significant differences in genes associated with aging.

Cell senescence - Cell senescence refers to the process by which cells decay over time. This can be related to shortening of the telomeres, or the process of apoptosis (or cell suicide) in which old or damaged cells are removed.

Stem cells - Pluripotent stem cells are immature cells which have the potential to become any type of cell in the body. It is theorized that aging may be related to either the depletion of stem cells or the loss of the ability of stem cells to differentiate or mature into different kinds of cells. It's important to note that this theory refers to adult stem cells, not embryonic stem cells. Unlike embryonic stem cells, adult stem cells cannot mature into any type of cell but rather only a certain number of cell types. Most cells in our bodies are differentiated, or fully mature, and stem cells are only a small number of the cells present in the body.

An example of a tissue type in which regeneration is possible by this method is the liver. This is in contrast to brain tissue which usually lacks this regenerative potential. There is now evidence that stem cells themselves may be affected in the aging process, but these theories are similar to the chicken-and-the-egg issue. It's not certain of aging occurs due to changes in stem cells, or, if instead, changes in stem cells are due to the process of aging.

Epigenetics - Epigenetics refers to the expression of genes. In other words, a gene may be present, but can either be turned on or turned off. We know that there are some genes in the body that are turned on for only a certain period of time. The field of epigenetics is also helping scientists understand how environmental factors may work within the constraints of genetics to either protect or predispose to disease.

As noted above, there is a significant amount of evidence that looks at the importance of genes in expected survival. When looking at genetic theories, these are broken down into three primary schools of thought.

There are several avenues of evidence that support a genetic theory of aging, at least in part.

Perhaps the strongest evidence in support of the genetic theory are the considerable species-specific differences in maximal survival, with some species (such as butterflies) having very short lifespans, and others, such as elephants and whales, being similar to ours. Within a single species, survival is similar, but survival can be very different between two species that are otherwise similar in size..

Twins studies also support a genetic component, as identical twins (monozygotic twins) are much more similar in terms of life expectancy than are non-identical or dizygotic twins. Evaluating identical twins who have been raised together and contrasting this with identical twins who are raised apart can help to separate out behavior factors such as diet and other lifestyle habits as a cause of family trends in longevity.

Further evidence on a broad scale has been found by looking at the effect of genetic mutations in other animals. In some worms as well as some mice, a single gene mutation may lengthen survival by over 50 percent.

In addition, we are finding evidence for some of the specific mechanisms involved in the genetic theory. Direct measurements of telomere length has shown that telomeres are vulnerable to genetic factors that can speed up the rate of aging.

One of the stronger arguments against a genetic theory of aging or a "programmed lifespan" comes from an evolutionary perspective. Why would there by a specified lifespan beyond reproduction? In other words, what "purpose" is there for life after a person has reproduced and been alive long enough to raise their progeny to adulthood?

It's also clear from what we know about lifestyle and disease that there are many other factors in aging. Identical twins may have very different lifespans depending on their exposures, their lifestyle factors (such as smoking) and physical activity patterns.

Its been estimated that genes can explain a maximum of 35 percent of lifespan, but there is still more we do not understand about aging than which we do understand. Overall, it's likely that aging is a multifactorial process, meaning that it is probably a combination of several of the theories. It's also important to note that the theories discussed here are not mutually exclusive. The concept of epigenetics, or whether or not a gene that is present is "expressed" can further muddy our understanding.

In addition to genetics, there are other determinants in aging such as our behaviors, exposures, and just plain luck. You are not doomed if your family members tend to die young, and you can't ignore your health even if your family members tend to live long.

We are taught to eat a healthy diet and be active and these lifestyle factors are likely just as important no matter how much our genetics are involved in aging. The same practices which seem to keep the organs and tissues of our body's healthy may also keep our genes and chromosomes healthy.

Regardless of the particular causes of aging, it can make a difference to:

Sources:

Jin, K. Modern Biological Theories of Aging. Aging and Disease. 2010. 1(2):72-74.

Kasper, Dennis, Anthony Fauci, Stephen Hauser, Dan Longo, and J. Jameson. Harrison's Principles of Internal Medicine. New York: McGraw-Hill Education, 2015. Print.

Kumar, Vinay, Abul K. Abbas, Jon C. Aster, and James A. Perkins. Robbins and Cotran Pathologic Basis of Disease. Philadelphia, PA: Elsevier/Saunders, 2015. Print.

Leung, C., Laraia, B., Needham, B. et al. Soda and Cell Aging: Associations Between Sugar-Sweetened Beverage Consumption and Leukocyte Telomere Length in Healthy Adults From the National Health and Nutrition Examination Surveys. American Journal of Public Health. 2014. 104(12):2425-31.

Smith, J., and R. Daniel. Stem Cells and Aging: A Chicken-Or-The-Egg Issue?. Aging and Disease. 2012. 3(3):260-267.

Go here to see the original:
The Genetic Theory of Aging - Concepts and Evidence

Read More...

The 5 Aging Startups Backed by Longevity Fund – Nanalyze

Wednesday, August 30th, 2017

Nothing is certain except death and taxes, said Benjamin Franklin, andhis words still hold true today. While there are ways to mitigate the effects of taxes like Foreign Earned Income Exclusion, death is coming and our days are numbered. It seems like quite the taboo topic that nobody wants to talk about. One nurse who did an informal survey found that the number one regret from people on their deathbeds was I wish I lived my life the way I wanted to and not the way other people wanted me to live it. If thats not an option for you, thenmaybe you need more time on this earth to sort yourself out. Quite a few startups are addressing theholy grail of cracking aging, an idea which supersedes every other startup idea out there. While extending the lifespan of humans may not be the best idea for this planet, quite a few people would happily slap down some cash for some extra years on this planet so they can live as long as some of the Asians do:

Source Population.sg (data as of 2014)

Weve looked at various aging startups before like Human Longevity, and Calico. When it comes to the ideal leadership profile for a company thats focused on aging, were split between having an old experienced executive who is increasingly aware of theirown morality versus a younger millennial filled with the endless optimism and naive ambition that typically characterizes youth of all generations. One good example of someone in the latter categorytackling theproblem of aging is Laura Deming. While all the news outlets are (appropriately) fawning all overthe profile of Ms. Deming, an MIT dropout who started working on the fund before she was old enough to smoke cigarettes, were more interested in where she has placed her bets so far. According to Longevity Funds website, theyve made investments in 5 startups so far. We didnt see anyone talking about what investments she has made, so were going to sort that out right now.

Unity Biotechnology first came across our radar earlier this year in an article we wrote about life extension science (a label thats much preferred over anti-aging). Since this San Francisco startup was founded in 2009, theyve taken in $154 million in funding with the latest round, a $35 million Series B, closing just 10 days ago (actually a continuation of a $116 Series B that started in Fall of 2016). According to Unity, that $151 million Series B is said to be one of the largest private financing rounds in biotech history. Loads of highprofile investors haveprovided all this funding including Jeff Bezos of Amazon, Peter Thiel of Founders Fund, Mayo Clinic, and Fidelity just to name some.

Healthy cells that age can experience a stress known as cellular senescence. Unity published research in April of this year showing that they can selectively eliminate senescent cells with a molecule (UBX0101) that may delay, prevent, or even reverse osteoarthritis (OA) which causes chronic joint pain in 80% of people over 65.

Precision Biosciences first came across our radar in an article we publisheda few years back on7 Gene Editing Companies Investors Should Watch. Since that article, 3 of those companies have gone publicand Precision Biosciences has taken in $25.65 million in total funding, all of which came in the form of a Series A which saw participation from Amgen (NASDAQ:AMGN) and Baxter (NYSE:BAX). Their platform revolves around ARCUS, a next-generation genome editing platform derived from a natural genome editing enzyme called a homing endonuclease. Heres how this technology can be used:

Genetics certainly comes into play when we think about life expectancy so its easy to see how this company fits the anti aging investment thesis.

Founded in 2015, San Francisco startup Alexo Therapeutics has taken in $61 million in funding so far to develop ALX148, a fusion protein that is supposed to improve the effectiveness of antibody therapies that are used to fight cancer. Since cancer is a leading cause of death, that may be why this company falls into an anti aging portfolio. Of course the same could be said for just about any healthcare company. The ALX148 protein is based off of research conducted at Stanford University which also happens to be an investor in this startup. On April of this year, they initiated dosing in a Phase 1 clinical program that is evaluating the safety of ALX148 in patients with advanced solid tumors and lyphoma.

Founded in 2015 as well, San Diego startup Metacrine has taken in $36 million in funding so far from investors that includevenBio (also an investor in Alexo Therapeutics and Precision Biosciences). The companys focus is on advancing research in nuclear hormone receptors for the treatment of metabolic diseases like type 2 diabetes and non-alcoholic steatohepatitis (NASH). The technology is based on research conducted Salk Institute researcher Ron Evanswho has founded many companies in the past including co-founding Ligand Pharmaceuticals (NASDAQ:LGND). Its safe to say that if metabolic diseases cause you to die early, then curing them amounts to increasing your longevity. Just a few weeks ago, Metacrine announced a collaboration with Novo Nordisk(NYSE:NVO) to develop Fibroblast Growth Factor 1 (FGF1) variants for glucose lowering and insulin sensitization.

Founded in 2014,Massachusetts startup Navitor Pharmaceuticals has taken in $56.5 millionso far from investors that include Sanofi (NYSE:SNY) and Johnson & Johnson (NYSE:JNJ). The startup is working on a new class of medicines for age-related diseases, and an article by FierceBiotech does a good job of explaining what Navitor does for people with no medical background. Cells in your body get hungry like you do, and there is a protein kinase called mTOR that controls their appetites. When something goes wrong with this process, bad isht happens like metabolic, neurological, inflammatory diseases, and even neurodegenerative conditions like Alzheimers disease. Thanks to some discoveries by Whitehead Institute researcher David Sabatini, Navitor can now target mTORC1 which plays the biggest role in cell growth.

Longevity Fund spent 6 months evaluating the merits of each investment, and all these aging startups picked so far have some commonalities. All have raised more than $25 millionand all are backed by high profile investors with venBio backing three of thesefive startups. While we need to wait and see what other investments are raised, its toughto see how any of these investments could have been made without connections (like Peter Thiel). Ms. Deming is the youngest fellow in the history of Peter Thiel Fellowships, and thats what triggered her to put her MIT education on hold. With 3-5 startupsleft for Longevity to invest in yet, it will be interesting to see what other aging startupsthey choose for their portfolio.

One firm that allows you to buy shares in startups before they IPO is Motif Investing. You can open a Motif Investing account for free with no deposit required so you are ready to buy shares of future IPOs before they begin trading.

Read the original:
The 5 Aging Startups Backed by Longevity Fund - Nanalyze

Read More...

Do Low Calorie Diets Help You Live Longer? – Healthline

Wednesday, August 30th, 2017

Cutting calories from the diets of many animals dramatically increases their life span, leading some people to try to extend their own lives in the same way.

If you could live to be 130 years old, what would you willingly give up?

How about 30 to 50 percent of the calories you eat for the rest of your life?

Proponents of the dietary practice known as calorie restriction (CR) happily make this trade-off every day in hopes of having life spans that leave todays centenarians in the dust.

This may sound like a fad diet, but theres quite a bit of research to back up the use of calorie restriction for longevity although most of it has been done in animals other than humans.

So how likely is it that eating child-size portions for every meal will gain you an extra few decades of life?

Studies have shown that calorie restriction can extend the life span and reduce age-related chronic diseases of many species, including mice, fish, worms, and yeast.

But these creatures are not people.

Which is why scientists turn to primates like rhesus monkeys, which age similarly to humans, as well as develop cancer, diabetes, and some traits of Alzheimers disease.

In a study published earlier this year in Nature Communications, researchers found that monkeys who ate a 30 percent calorie restricted diet lived longer than those on a regular diet.

Six of the 20 monkeys on a calorie restricted diet have lived beyond 40 years. The average lifespan for monkeys in captivity is around 26 years. One male is currently 43 years old, a record for the species.

Researchers also found that calorie restriction benefitted older monkeys, but not younger ones. This is in contrast to other studies in mice that showed that starting calorie restriction at a young age gives the best results.

The sex of the monkeys and what they ate not just the number of calories also affected how much monkeys benefitted from calories restriction.

While the results of animal studies are promising, scientists know less about how calorie restriction affects people, especially long-term.

Given that Americans live on average around 78 years, researchers would have to wait decades to see if calorie restriction extended human life span.

To compensate for this, Duke University researchers instead looked at measures of biological age.

In a study published earlier this year in The Journals of Gerontology: Series A, researchers divided volunteers into two groups a calorie restriction group and a regular diet group.

The calorie restriction group aimed to cut their caloric intake by 25 percent although by the end of the two-year study they had only achieved a 12 percent reduction.

After each one-year period, the biological age of people in the calorie restriction group increased by 0.11 years, compared with 0.71 years for people who stuck with their usual diets.

Researchers calculated biological age using chronological age and biomarkers for things such as cardiovascular and immune system function, total cholesterol, and hemoglobin levels.

However, researchers only followed people for two years. Whether these benefits continue after this point, and at what level, is unknown.

No one is certain why calorie restriction increases the life span of so many organisms.

Some scientists think it may have to do with free radicals atoms with an unpaired electron that are released when the body turns food into energy.

Free radicals can damage important parts of the cell, like DNA and the cells membrane. So cutting back on the food you eat may decrease the number of free radicals circulating in the body.

Insulin could also play a role. As we age, our bodies can become resistant to this hormone, leading to excess glucose in the blood that can damage organs, blood vessels, and nerves.

Some researchers, though, think calorie restriction increases longevity by rejuvenating the bodys biological clock.

This clock is actually a set of genes that change activity in order to sync with the cycle of day and night.

In a recent study published in the journal Cell, researchers found that the biological clock activated different genes in liver cells of older mice, compared with younger ones. As a result, cells in older mice processed energy inefficiently.

However, when researchers cut the calorie intake for older mice by 30 percent for six months, the energy processing in the cells resembled that of young mice.

A second research group, in another study published in Cell, saw a similar reboot of the biologic clock of stem cells in older mice fed a calorie-restricted diet.

If signing up for a lifetime of hunger to gain a few extra years of life doesnt sound appealing, you may have other options for breaking the 100-year mark or at least living healthier.

A team led by gerontologist Valter Longo, PhD, director of the University of Southern California Longevity Institute, tested the effects of a fasting-mimicking diet an alternative to only drinking water on the risk of developing major diseases.

The study was published earlier this year in Science Translational Medicine.

People on the fasting-mimicking diet ate about 750 to 1,100 calories per day, for five days per month, over three months.

Adult women usually eat 1,600 to 2,400 calories per day, and adult men generally eat 2,000 to 3,000 calories per day.

Food used in the study contained exact proportions of proteins, fats, and carbohydrates.

People on the fasting-mimicking diet saw a drop in their blood pressure, blood glucose, and markers of inflammation, compared with people eating a regular diet.

People who fasted also lost weight over the three months, but not muscle mass, which is a concern with a calorie-restricted diet.

As with other calorie restriction studies in people, this one doesnt show that cutting back on calories increases lifespan, only that it may reduce certain risk factors for disease.

The CR Society International, an organization that offers resources for people wanting to live longer by cutting out calories, lists some of the potential unwanted effects of long-term calorie restriction.

These include loss of bone mass, sensitivity to cold, and decreased sex drive.

Some experts are also concerned that calorie restriction could cross the line into an eating disorder, such as anorexia nervosa.

Dr. Ovidio Bermudez, chief clinical officer and medical director of child and adolescent services at the Eating Recovery Center, said if someone walked into his office saying that they were going to cut their caloric intake by 30 or 50 percent for the rest of their life, I would raise serious concerns about that.

You may be awakening a monster that you dont want to deal with, said Bermudez.

He emphasized, though, that not everyone who does calorie restriction will develop anorexia.

Like other eating disorders, anorexia has a strong genetic component that puts some people at risk more than others, although scientists dont fully understand the genetics.

However, genetics alone is not enough to trigger the disease.

The genetic predisposition [to eating disorders] is insufficient and needs to interact with some other influences, said Bermudez. It seems that, in this day and age, those other influences are, to a great extent, environmental.

There is no single environmental trigger for anorexia.

Some teenage girls or boys may take a health class in high school and decide to eat less and exercise more. Or a young adult may look around and try to conform with the fast-paced, thin-ideal culture that we live in, said Bermudez.

Or someone wanting to live longer may restrict their calories.

Not everyone in these situations will develop an eating disorder. But the danger is that someone who drastically cuts down on eating will cross a threshold leading to a neurobiological change that seems to both trigger and cement the illness process, said Bermudez.

People doing calorie restriction who end up with pre-anorexia or anorexia may not even realize they are in trouble.

There is a subset that will probably cross the threshold and lose perspective, said Bermudez, and those are the people who are unlikely to have a keen awareness of whats really happening with them.

Bermudez said that even though eating disorders are serious illnesses with the highest mortality rate of any mental illness there is hope, whether the trigger was extreme veganism or calorie restriction.

Treatment, though, works best with early diagnosis and effective intervention.

Since many people who do calorie restriction see a doctor regularly to make sure they arent slipping into malnutrition, these visits could also be a good time to check their mental health.

Asked whether calorie restriction makes sense in people, Bermudez pointed to the lack of long-term studies in humans.

If I owned a rat or a worm, and I wanted them to live for a long time, I would do calorie restriction for them, said Bermudez. But I wouldnt do it for my kids or my family because the data is simply lacking.

See more here:
Do Low Calorie Diets Help You Live Longer? - Healthline

Read More...

Genetic mutation explains why some men live to 100 – Jewish Journal

Friday, August 25th, 2017

Just as smaller animals of a given species generally live longer than their larger cousins, one might expect that taller humans are genetically programmed to sacrifice longevity for height.

But its not that simple.

A major multinational study of 841 men and women from across four populations found lower levels of insulin-like growth factor 1 (IGF-1) in men living to age 100 and yet most of them were taller than men in the younger control group.

The apparent explanation for this head-scratcher is that some long-lived men and only men have a genetic mutation that makes their growth hormone receptors more sensitive to the effects of the hormone. The cells absorb less growth hormone, yet protein expression is increased by several times.

This mutation seems to be responsible for their ability to live about 10 years longer than the control group of 70-year-old men without the mutation, even though they have a lower amount of growth hormone and are about 1.18 inches taller.

The lead author of the study is professor Gil Atzmon of Albert Einstein College of Medicine in New York and head of the Laboratory of Genetics and Epigenetics of Aging and Longevity at the University of Haifa. Since 2001, Atzmon has been studying the human genome and its impact on aging and longevity.

The researchers working with Atzmon looked at four elderly populations: 567 Ashkenazi Jews in theLongevity Genes Projectat Einstein, 152 from a study of Amish centenarians, and the rest from an American cardiovascular health study and a French longevity study.

In 2008, the Longevity Genes Project found a genetic mutation in the IGF-1 receptor of some women, although its not the same as the one affecting mens lifespans.

We knew in the past that genetic pathways associated with growth hormone were also associated with longevity, and now we have found a specific mutation whose presence or absence is directly related to it, Atzmon said.

This study makes it an established fact that there is a relationship between the function of the growth hormone and longevity. Our current goal is to fully understand the mechanism of the mutation we found to express it, so that we can allow longevity while maintaining quality of life, he added.

The 16 researchers involved in the study, published June 16 inScience Advances, are associated with institutions in Israel and France as well as in New York, Maryland, California, Vermont, Massachusetts and Washington.

While more research is needed to understand why the receptor mutation affects longevity and why it happens only in men, the study suggests that making a slight change in this specific piece of DNA could possibly make people live longer.

Although the presence of the mutation almost certainly ensured longevity, Atzmon stressed that many other factors affect longevity and that many men without the mutation also live to 100 and older.

Atzmon is one of the principal researchers in the Longevity Genes Project at Einstein, along with Israeli endocrinology specialist Dr. Nir Barzilai.

Their groundbreaking 10-year study of healthy Ashkenazi Jews between the ages of 95 and 112 and their children attempted to understand why humans dont all age at the same rate, and why only one in 10,000 individuals lives to 100.

The centenarians were found to have genetic protective factors (longevity genes) that overcame factors such as diet and lifestyle.

The rest is here:
Genetic mutation explains why some men live to 100 - Jewish Journal

Read More...

This 23-year-old just closed her second fund which is focused on … – TechCrunch

Friday, August 25th, 2017

Laura Deming is not your typical venture capitalist. Then again, she isnt typical in many ways.

For starters, the 23-year-old, New Zealand native was home schooled, developing along the way a love of math and physics and, perhaps most interestingly, the biology of aging. In fact, she became so preoccupied with the latter that at age 11, Deming wrote to Cynthia Kenyon, a renowned molecular biologist who specializes in the genetics of aging, asking if she could visit Kenyons San Francisco lab during a family trip to the Bay Area. Kenyon said yes. When, soon after the visit, Deming asked if she could work in the lab, Kenyon said yes again.

Demings family moved to the U.S. to make it possible, and its highly doubtful they regret the decision. Indeed, by age 14, Deming was a student at MIT, and two years after that at the tender age of 16 she was a college drop-out, having been accepted into Peter Thiels two-year-old Thiel Fellowship program, which gives $100,000 to young people who want to build new things.

Often, those new things evolve along the way. Not for Deming, who pitched the idea of a venture fund that would support aging-related startups, and has since turned that early concept intoLongevity Fund, an early-stage venture outfit that just closed its second fund with $22 million.

Earlier today we caught up with Deming to learn more about her path and which technologies shes betting on to extend the human lifespan.

TC: Its incredible that this all started with an email to a UCSF professor.

LD: [Cynthia Kenyon] is the most amazing person Ill ever meet.

TC: What did you do in her lab, exactly?

LD: We were working with tiny, see-through worms. You put them on a plate of jelly and you see what happens if you change their genetic material. Do they live longer or die faster? If you starve them, they live longer. If you starve worms and also turn off certain genes, could you get them to live even longer? I was nave, but I really wanted to make the longest-living worms ever. [Laughs.]

TC: What did you study at MIT?

LD: I majored in physics actually, but I continued to work in a couple of labs, including [one overseen by] Lenny Guarente [a biologist known for his research on lifespan extension]. It was a lot of fun. I thought Id be a scientist, but a grad student familiar with the Thiel fellowship told me I should apply and I did. Its funny, one of the directors of the [Thiel] program told me recently that he thought Id fail, even though he was very supportive. After we closed the first fund, he was like, I never thought that would work out.

TC: Why?

LD: In part because not long ago, if you talked with most VCs about aging, they didnt think there was anything there. I think aging is such a young science, they hadnt heard about it. Meanwhile, I care a lot about it, and though we dont know if itll work or not, its not unlike [biotech companies trying to tackle] cancer in that way, and if you believe in cancer companies, you should also care about aging companies.

TC: How much did you raise for that first fund?

LD: A grand total of $4 million, and I was very proud of this. To be honest, Id assumed $100,000 was enough to build a fund until I arrived in San Francisco and realized it was really enough to live on for two years. When I started fundraising, I was 17 too young to legally sign contracts. Id never managed money before. But I could talk to people about the science and got them on board with that. In the end, we had great anchor investors come together, and we invested in five companies that kind of proved out the strategy.

TC: Were one of those anchor investors Peter Thiel?

LD: We dont really talk about our LPs.

TC: You say we, though youre the sole general partner of Longevity. Is that correct?

LD: Yes, but I have a lot of back-office support. The way Longevity is structured, Im also able to pull in the best people who have expertise from different domains, so its not one person who looks at all the deals.

TC: And these advisors get a stake in the company?

LD: Sometimes. Others especially grad students like to be paid up front. Well find the best incentive for that individual and work with that.

TC: One of your portfolio companies is Unity Biotechnology, a company thats trying to reverse aging through therapeutics. Didnt it just raise a giant Series B round this week?

LD: It did. All of the companies in that portfolio have [at least] raised Series A rounds of $30 million or more to get to that proof of concept.

TC: Given the amounts involved, is the plan to form special purpose vehicles, or SPVs, around your break-out winners?

LD:We like to help LPs follow on, so we look to do that in whatever way makes sense for both parties. With Unity, we put in money as early as possible because Ned Davis, who runs the company, is amazing and we thought its aging thesis would succeed.

TC: How many companies do you expect to fund with your newly closed fund?

LD: Eight to 10 companies.

TC: Do you think your work will be harder, given that investors seem to be paying much more attention to aging suddenly?

LD: No. With our first fund, we spent up to six months with each deal, tracking the company before it was even raising. Its something LPs really value from us; they know when they invest in something that they dont need to re-do the diligence, that weve already looked at a bunch of stuff and we know this is the best possible investment in [a particular vertical].

Earlier, our biggest challenge was getting other investors on board and convincing them that aging has become a place to play. Now thats a non-issue, which is great. Our job is to help the companies get other investors on board, so its wonderful to see excitement in the space begin to build.

TC: You look at a lot of technologies. I have to ask: do you find these new blood transfusion startupsas interesting as the writers of HBOs Silicon Valley?

LD: [Laughs.] While scientifically interesting, I think they get a little over-discussed in the press because of that vampirism. Its not as sexy to talk about new genetic regulatory elements that control the aging process. Thats not going to get as many clicks as a story about drinking the blood of your five-year-old.

Excerpt from:
This 23-year-old just closed her second fund which is focused on ... - TechCrunch

Read More...

Kahn Longevity Center

Saturday, August 19th, 2017

Concierge medicine. You have heard about it. Another name for it is direct patient care or DPC. The most common perception about concierge medicine is access to longer appointments and the ability to reach a doctor easier. Well, what if..Kahn-cierge was access to longer appointments, access to Dr. Joel Kahn, Americas Holistic Heart Doc.AND the MOST advanced detection and treatment center for measuring and treating heart disease at ANY stage with a goal of optimal LONGEVITY. Not just extra time but extra expertise from the worlds first physician to be Certified by a major University medical center in Metabolic Cardiology. A Summa Cum Laude medical school graduate, author, TV personality, and speaker. Are you seeking Healthy Aging? More Energy? Fewer Medications? Early Disease Detection? Strategies for Disease Reversal? Natural Cardiology? Nutritional Cardiology? Are these of interest to you? Feeling better, looking better, sleeping better, more Kahn-fident, and enjoying and following a plan for your best health and energy! Science is advancing rapidly, and it is possible to live longer and better free of dis-ease, dis-stress, and dis-comfort. The future of healthy aging is so exciting. Dr. Kahns personalized medicine offers a plan for you, based on your individual health, nutritional profile, genetic profile, preferences, and abilities. One size does not fit all when it comes to living your most enjoyable and healthiest life. So become one of a few select members of the Kahn Center for Cardiac Longevity and lets walk together, preparing for a bright and joyful future without the worry about Americas #1 killer, heart disease.

Read this article:
Kahn Longevity Center

Read More...

Media’s Anti-Aging Agenda without the Benefit of Scientific Evidence, Fact or Common Sense – Anti Aging News

Saturday, August 19th, 2017

Allure Magazine announces it's intention to ban the term "anti-aging," and wipe it from the internet. Another veiled political attack against the successes of anti-aging sciences, but longevity medicine cannot be stopped.

First of all let us applaud the 29 celebrities in the Allure Magazine online article on August 15, 2017 (https://www.allure.com/gallery/celebrities-against-anti-aging) for what appears to be living and eating healthy, adhering to Anti-aging Medical Principles (preventative medicine), getting the proper amount of exercise, possibly buying all natural, Non-GMO foods, and being blessed with great genetics.

Unfortunately, being blessed with great genetics alone does not always help someone stay beautiful as they circle the sun more than 40 times. You have to believe that, all though age is inevitable, it is not the only factor that causes your body to deteriorate and grow older.

WorldHealth.net thinks its horrible that some people misuse terms, or that in many cases the wrong message gets attached to good philosophies. WHN doesnt believe in unnecessary plastic surgery, hormonal therapies that are not physician supervised and medically legal, stem cell therapies at the beauty shop, overzealous and sometimes completely fake claims by advertisers such as anti-aging miracle skin creams, nor does WHN or any of its affiliates believe a buzz word or coined term should be used to influence potential consumers. WHN believes that credible non-commercial information should be used as a tool to combat ignorance and protect consumers.

If you are truly interested in the mission of anti-aging medicine, subscribe to the Longevity Now newsletter here at WorldHealth.net and go to A4M.com and read about this 25-year-old revolution. Anti-aging medicine, said Dr. Ronald Klatz, President of the A4M, who is accredited with coining the Term anti-aging more than 25 years ago, for the most part is all about inner beauty, it isnt about cosmetic surgery, solely enhancing your appearance, or even living forever. Its about living healthy, growing older with a better quality of life, and slowing down or defeating diseases such as Macular Degeneration, Alzheimer's, Diabetes, disease associated with hormone inadequacies, and a host of other ailments that affect our bodies from gravity to normal wear and tear. No matter how new or old your car is you have to change the oil at regular intervals or your car simply wont last. I know this is a simple analogy, but our bodies are basically the same. anti-aging is basically preventative medicine that looks and takes advantage of natural remedies mixed with scientific gains to help the 100,000 plus doctors the A4M has trained, help their patients live happier and healthier lives.

Dr. Ronald Klatz went on further to point out, magazines such as Allure hardly focus on or promote inner beauty. They focus on outward appearance, pushing cosmetic goods, promote unbelievably attractive celebrities and models, and then they have the gall to take shots at Botox for helping many millions of people try to look and feel a little better. Unfortunately, Allure type magazines give young women unrealistic expectations on a daily basis that you need to look like Hale Berry, or Jennifer Anniston to be beautiful. Thats simply not true, but you do need to live the anti-aging lifestyle so as you advance in years youdont suffer, and decay as old folks, but rather enjoy life to your last breath. Thats the true anti-aging message.

WHN would like to recognize bias, and WHN is a proponent of anti-aging medicine so therefore WHN is slightly biased. WHN is an opponent of the abuse of the term anti-aging by advertisers, product peddlers, and financially motivated journalists. For example, WHN can only make guesses as to the intentions of Allure Magazines position on the term anti-aging. Allure could be talking about skin cream. However, WHN can use the quotes actually published by Allure titled 29 Celebrities That Are Against The Term Anti-Aging to let WHNs readers draw their own conclusions as to the intentions of Allure. If you read the quotes exactly as they appear at https://www.allure.com/gallery/celebrities-against-anti-aging, you will notice that only 3 of 29 beautiful celebrities even mentioned anti-aging.

WHN prefers to be honest and avoid Fake News. WHN would like you to know that Jamie Lee-Curtis almost mentioned the term anti-aging. Instead Jamie refers to anti-aging as anti referencing aging in the same sentence before the term anti which is the word that ends the sentence. Jamie starts the next sentence with aging with the sentence before ending in anti. This is simply journalistic dishonesty by a publications editor. This example of lack of journalistic integrity can be seen underlined below in Jamies quote:

I am appalled that the term we use to talk about aging is 'anti' (end of sentence). Aging (Start of new sentence to create anti-aging) is as natural as a baby's softness and scent. Aging is human evolution in its pure form. Death, taxes, and aging." fromThe Huffington Post

Go here to read the rest:
Media's Anti-Aging Agenda without the Benefit of Scientific Evidence, Fact or Common Sense - Anti Aging News

Read More...

Genetic strategies to reduce gilt feed and development costs – National Hog Farmer

Monday, August 14th, 2017

By Mark Knauer, North Carolina State University Assistant Professor and Extension Swine Specialist; and Garrett See, North Carolina State University Graduate StudentIn recent years industry geneticists have chosen to focus on increasing sow output through increases in litter size. At some point we may reach an optimal litter size at the commercial level. Some may debate we are there now. Yet geneticists are working to enhance piglet quality which should make it easier for farmers to wean large, quality litters in the coming years.

So what future opportunities do we have to enhance female reproduction through genetics? Genetically reducing age at puberty offers producers multiple avenues to reduce sow herd costs. Recent research by Garrett See (2017) suggests genetically reducing age at puberty would allow gilts to be mated at younger ages and lighter weights. The author reported that after four generations of selection for young puberty, average age and weight at puberty were 163 days and 224 pounds, respectively.

Hence, in theory, you could farrow your gilts at an average age of 10 or 11 months versus a year of age. Not only would this allow you to substantially reduce gilt feed cost, but also potentially market late puberty gilts as full-value market animals. See (2017) further suggests selection for reduced age at puberty would increase gilt retention, enhance sow longevity and improve piglet quality. Hence the benefits of a genetically young puberty gilt are multiple. Yet more research around early puberty is warranted. Can we consistently breed genetically young puberty gilts to farrow at 10 months of age? What is the true economic value of age at puberty?

Genetic suppliers will tell you age at puberty is a challenging trait to capture at the nucleus level. They are currently correct. Yet I think there are some strategies to reduce the cost of capturing puberty data in the nucleus. At the commercial level Im not sure many changes would be needed to incorporate early puberty females, just start breeding at a younger age.

I would like to acknowledge the North Carolina Department of Agriculture and North Carolina Pork Council for their support of this project. Contact Mark Knauerwith questions.

ReferencesSee, Garrett. 2017. Correlated Responses to Selection for Age at Puberty in Swine. M.S. Thesis. North Carolina State University, Raleigh.

Original post:
Genetic strategies to reduce gilt feed and development costs - National Hog Farmer

Read More...

An extra dose of this longevity hormone helped make mice smarter – Popular Science

Tuesday, August 8th, 2017

Klotho, in ancient Greek mythology, is one of the Fates controlling the span of human destiny tasked as she was with cutting the string that determined the length of a persons life. Klotho is also a naturally occurring hormone in the body. More than two decades ago, Japanese researchers discovered that this hormone plays a role in aging. People with more klotho in their body, tend to live longer and to retain more of their facultiesthat is to stay sharpwell into old age.

There are many elderly people who walk around and have completely normal, sharp vibrant brain function despite the fact that they are much older and the fact that they have a lot of disease toxins in the brain, Dr. Dena Dubal a neurobiologist at the University of California, San Franciscos Memory and Aging Center told Popular Science.

Dubal is the author on a study released today in the journal Cell Reports that looks at what happens to brain function when you inject klotho protein into mice.

Aging researchers like Dubal arent interested in extending life for its own sakethey dont have a Methuselah complexbut rather because aging is the biggest factor for disease. If they can find a way to slow aging, or at least its effects, they might find a way to allow us to avoid the cognitive declines, and diseases such as stroke, heart disease, and diabetes that tend to come as we flip through more pages on the calendar.

A few years ago, we discovered in collaboration with several people that in human populations those people that had higher levels of klotho had better cognitive functions in normal aging, says Dual.

Which is great if you happen to be genetically blessed with higher levels of klotho, less great if youre not.

Earlier studies have looked at the impacts of klotho on longevity and health, including brain health, but they tended to rely on genetically modified mice. The genes of these mice had been modified to allow them to produce more klotho or less.

Those studies found that mice modified to produce more klotho lived longer, which is promising. But despite recent advances, genetically modifying humans is still an ethical minefield away in the future, especially if the modifications are being made to improve wellbeing as opposed to preventing disease. And, genetically altering embryos might help future populations, but it doesnt do much for those of us already living. So, Dubal wanted to see is if mice injected with klothoacting essentially a hormonal supplementwould show cognitive improvements.

The answer, seems to be yes.

To get the result, Dubal and her colleagues injected three types of mice with a portion of the protein. They injected young mice, aged mice, and mice genetically altered to have brains similar to that which we would see in Alzheimers or Parkinsons patients in humans.

Within hours they showed better cognitive function, says Dubal.

Since you cant exactly administer a mouse an IQ test, they assessed brain power based on the mices ability to navigate a series of water mazes, in an experiment that sounds on par with human a trip to Wisconsins famed waterslide park, The Dells.

They found that mice that had daily injections and were better able to navigate the maze (as measured by the distance traveled to find a hidden platform) than their control group peers. In a classic example of work smarter, not harder, the klotho mice were just much more efficient seekers.

We tested them two weeks later in a different cognitive test and they were still smarter, says Dubal, which suggested that getting the klotho protein into their bodies combined with brain training and stimulation had a long-lasting effect in their brain. Because the half-life of the protein is only seven and a half hours long, any of the protein should have been long out of their system.

There are a few caveats.

First, this was an experiment in micenot in humans. While its incredibly promising, the study results are short term and they werent looking for side effects. The pace at which a promising scientific study is turned into a supplement of dubious efficacy is stunning these days, so please dont subject yourself to some back-alley klotho injection. Between resveratrol and superfoods weve been there before.

At the same time, we know that klotho levels can be affected not only by genetics - but also by stress. If you want to help your body keep its klotho levels at your own peak you might want to try proven stress reduction techniques like exercise and chilling out about your own impending mortality.

Finally, the researchers arent sure how klotho seems to be generating this effect because the protein is too big to pass from the body into the brain. For a long time, weve studied the brain in isolation from the body with the brain mostly telling the body what to do, and the body acting like a series of censors that give the brain useful data. But this study like our growing understanding of the connection between our microbiome and brain health, and exercise and brain health, this study touches on our growing understanding of how the body connects to the brain to help make the brain more resilient.

The rest is here:
An extra dose of this longevity hormone helped make mice smarter - Popular Science

Read More...

EDITORIAL: 99%, My Life Sucks, Dogfighting, and Brady’s Brain – GoLocalProv

Tuesday, August 8th, 2017

Email to a friendPermalink

Monday, August 07, 2017

EDITORIAL

Tom Brady - is his brain immune from injury?

It has been nearly eight years since Malcolm Gladwell wrote his piece in The New Yorker about the long-term tragic impact of football on the brain of those that play the game. It outlines the post-football life, There were men with aching knees and backs and hands, from all those years of playing football. But their real problem was with their heads, the one part of their body that got hit over and over again.

Gladwell's article, Offensive Play How different are dogfighting and football? is one of a growing number of looks at the depressing post-football life of the players - viewed by many as American heroes. He wrote it 2009.

My Life Sucks

Now in 2017, Jim Plunkett, who was a Super Bowl hero back in the day and now 69, told the San Jose Mercury News, My life sucks. Plunkett's body is broken and he is in constant pain. Once a man like Tom Brady - overcoming obstacles and realizing NFL quarterback hero status -- he is now devastated physically and his wife says in the same interview that Plunketts mind is being ravaged by the years of head injuries.

Math 99% of NFL

In July, a paper in the JAMA found that Chronic Traumatic Encephalopathy, known as CTE, was found in 99% of deceased NFL players' brains those that were donated for scientific research. And, CTE was found in three of the 14 high school players and 48 of the 53 college players whose brains were tested. CTE is directly linked tomemory loss, confusion, impaired judgment, aggression, depression, anxiety, impulse control issues, and suicidal behavior. A slew of former NFL players have committed suicide in the past few years and their brains were found to have CTE.

Beautiful Brady

While so many love the amazing story of Bradys greatest of all-time title and marvel at his longevity built of his dedication to physical performance, luck, genetics, and kale salads, the reality is none of those things can protect his brain from the impact of years of missed blocks or blindside corner blitzes.

The collective failure of those missed blocks and defensive scheming of opponents may not have stopped the Patriots from being the great team in NFL history, but they have taken their toll on jarring the brain of Brady no less than any other player. The reality is that the longer he plays, the more his brain is impacted.

His longevity is now working against his future the statistics prove it. The list of NFL players who were found to have CTE is long and depressingly catastrophic. The Patriots alone can claim Junior Seau, Most Tatupu, and Kevin Turner -- all tested after death and all suffered from CTE. Seau killed himself at age 43, Tatupu was 54 at the time of his death, and Turner died of ALS believed to be linked to CTE at the age of 46.

Deviant Behavior

Gladwell ends the article with a passage from Dogmen and Dogfights, an academic analysis of deviant behavior by Rhonda Evans and Craig Forsyth about those involved with dogfighting.

The authors write:

When one views a staged dog fight between pit bulls for the first time, the most macabre aspect of the event is that the only sounds you hear from these dogs are those of crunching bones and cartilage. The dogs rip and tear at each other; their blood, urine and saliva splatter the sides of the pit and clothes of the handlers. . . . The emotions of the dogs are conspicuous, but not so striking, even to themselves, are the passions of the owners of the dogs. Whether they hug a winner or in the rare case, destroy a dying loser, whether they walk away from the carcass or lay crying over it, their fondness for these fighters is manifest."

Email to a friendPermalink

Read more:
EDITORIAL: 99%, My Life Sucks, Dogfighting, and Brady's Brain - GoLocalProv

Read More...

23 People That Lived to 100 Spill Their Secrets of Longevity

Sunday, July 30th, 2017

Ever since I was a kid, I was always curious about the secrets to longevity and what made people live to 100. I mean, 100 is kind of a random number, but triple digits?! Thats pretty amazing there must be some kind of secret. Seeing as you usually hear that peoples grandparents die in their 70s or 80s I was totally intrigued by these long-lived peeps.

My great grandma, who I knew until I was about 10 years old, lived to her later 90s, 97 or 98 and I was still hanging out with her at that age talking about life. Unfortunately since I was so young, the only thing I remember was how creepy she was. She was really small. Really thin. And really veiny.

Anyway, as I got older I became more interested in longevity not because I actually wanted to live to 120, but because the kinds of people that live to 120 are the ones who usually enjoy an unprecedented quality of life throughout their lives.

The people who take good enough care of themselves to live to that ripe, old age, also suffer from a fraction (or none) of the health problems that plague the majority of people today.

Interestingly enough, as I researched the secrets to their longevity I found much of the same advice, over and over.

You can find that advice below.

Exclusive Bonus: Download this bonus guidethat tells the story of how a Chinese herbalist lived to 200+ and his 4 pieces of advice.

Based on her Wikipedia entry, at age 85, she took up fencing and continued to ride her bicycle up until her 100th birthday. She was reportedly neither athletic, nor fanatical about her health.

Calment lived on her own until shortly before her 110th birthday, when it was decided that she needed to be moved to a nursing home after a cooking accident (she was having complications with sight) started a small fire in her house.

Calment smoked from the age of 21 to 117, though according to an unspecified source, she smoked no more than two cigarettes per day. (Damn babygirl you smoked for 100 years?!)

Calment ascribed her longevity and relatively youthful appearance for her age to olive oil, which she said she poured on all her food and rubbed onto her skin, as well as a diet of port wine, and ate nearly one kilogram (2.2lb) of chocolate every week.

Not a bad life, eh? Smoke, drink and eat chocolate.

Based on biographical information via Wikipedia:

Her only child, Kathryn Knauss Sullivan, who was 96 at the time of Sarahs death and lived to be 101 herself, once explained Knauss longevity by saying: Shes a very tranquil person and nothing fazes her. Thats why shes living this long.

On his 115th birthday Mortensen gave his advice for a long life: Friends, a good cigar, drinking lots of good water, no alcohol, staying positive and lots of singing will keep you alive for a long time.

He credited his longevity to funche, a boiled corn, codfish and milk cream-like dish, which he ate every day as a habit. Mercado also claimed that his sense of humor was probably responsible for his long life, and he would tell jokes and humorous anecdotes almost to the end of his days.

He would not elaborate on details of his love life, but would humorously hint about them: in one of the many interviews he gave to Puerto Rican media, Mercado claimed to have been at the dancing club (a euphemism for a bordello) owned by Isabel la Negra the day she was assassinated.

He was 82 years old at the time and reportedly hid under a table when Oppenheimers killers started firing gunshots. Asked what he was doing there, he said: praying or at least I was when the bullets started flying!

Mind your own business and dont eat junk food. Treat everyone the way you want to be treated, work hard and love what you do.

Laughter keeps you healthy. You can survive by seeing the humor in everything. Thumb your nose at sadness; turn the tables on tragedy. You cant laugh and be angry, you cant laugh and feel sad, you cant laugh and feel envious.

Do the right thing, dont smoke, dont drink, eat right and dont overdo it. If you need a little extra help, take some vitamins. Going to work is what keeps me going.

Look inside your soul and nd your tools. We all have tools and have to live with the help of them. I have two tools, my words and my images. I used my typewriter, computer and my cameras to ght injustice. Whenever I see a possibility of helping people who are in danger, I want to help them.

Eat right and do what you love. Whatever you love to do is play; doing what you dont like to do is work. I have never worked a day in my life!

Have a good appetite, lots of friends, and keep busy.

Stay active even at 100. Eat in a balanced way Dont stay mad at anythingyou have to get used to the losses, otherwise you cant win. Lastly, stay close with your family, they keep you thinking.

Have a good wife, two scotches a night, and be easy-going.

It is very important to have a widespread curiosity about life.

Take it easy, enjoy life, what will be will be. Sleep well, have a Baileys Irish Cream before bed if you have a coldyou will wake up ne the next morning.

Dont smoke, dont drink, and dont retire!

Take one day at a time and go along with the tide.

Keep busy! Do things that youve never done before.

Dont ght the day, just let it be. Get up and be positive. Avoid any and all drama; I dont get involved with silly minutiae or difcult personalities; people respect me for that.

Be good, dont complain, just get up and do. Keep on working, keep on going, and have a good time.

Get involved. Youll nd pleasure and sometimes disappointment but there is a sense of achievement if you participate in a successful undertaking, whether it is organizational or professional. Work hard, it will pay off.

Whatever is hard, you make hard, but if you take it as it comes, it doesnt come hard. Dont worry, dont want so much, and be satisfied with what youve got. Be willing to share with your friends and those less fortunate.

You must keep active or you will just wither away. Always be involved in some activity.

You have to make the best out of your life and have a good attitude.

When you live for God, talk to him, go to church, have nice people around you; that is the best medicine. God provides for you. Sometimes you dont know when it is coming, but it is coming.

Try to understand the kind of person you are and accept who you are; but if you want to improve your situation, change it. Keep your eye on the stars and try to succeed at what you want to do.

For a long, healthy life, you need a plan and a purpose. It could be family, writing a book, becoming president. Without a purpose, plan or objective, what do you need?

Never run out of responsibility; if you dont have one, nd one. Find a cause and knock yourself out for it. It will enhance your brainpower, interest in life, and keep you alive longer. Im alert because I work. Virtue is its own reward.

My longevity is attributed to my long happy marriage. We did everything together. She was perfect in my eyes.

You know whats cool?

There are definitely a few things repeated over and over.

I went back through the list and wrote down the top 5 things that appeared the most frequently. Do you know what they were?

The most commonly cited things:

[See Also: A 256 Year Old Man Reveals His 4 Secrets to His Longevity]

I think that many of us intuitively know some of the things that contribute to a long life, like relaxing and enjoying life, keeping your mind busy, and obviously eating right and exercising.

One of the craziest things Ive come across in the past few years is that some people have willed themselves to death. In extreme survival situations, people have been found in safe, secure places, with food and water, who simply gave up. Sometimes there was a journal, but other times these people had no verifiable medical reason for their death. They just didnt want to fight.

The external is seriously overrated in our society people seem to neglect the power of the mind to make a person happy or miserable, successful or unsuccessful, lazy or driven.

The oldest woman in the world smoked every day for almost 100 years. Was she lucky? Maybe.

But listen to all of these people talk they just freaking love life. A good cigar, a glass of wine, some chocolate, and good friends. Theyre living the good life no wonder they want to go on living.

So what about you? Do you know anyone who lived to a ripe-old age? What do you think contributed to it?

Yes, Im pretty biased since Im the author of the book.

But it was an instant #1 Amazon bestseller in the health category, and heres why.

Its not about diets.

Its not about stupid eating less and moving more advice.

Its not about willpower, discipline and grinding it out.

It all comes down to habits just a few in particular.

I cover all of them in my bookMaster The Day: Eat, Move and Live Better With The Power of Tiny Habits.

Check it out on Amazon here.

There are over 55 5-star reviews now. What are you waiting for?

Alex

***

Sources: Some adapted from the book Extraordinary Centenarians in America.

Other quotes were compiled from interviews listed in Wikipedia.

Images: Indian Man, Countryside, Empire State Building, Puzzle, Bridge, Sunset, Ice & Sunset, Green Vegetable, Desert

See the rest here:
23 People That Lived to 100 Spill Their Secrets of Longevity

Read More...

16 Genetic Markers Linked to Lifespan | Worldhealth.net Anti-Aging … – Anti Aging News

Sunday, July 30th, 2017

Researchers have identified the largest-ever number of genetic markers most of which are brand new to science that are linked to life expectancy.

A research team based in Switzerland has pinpointed a massive haul of genetic markers. It is the largest group of such genetic markers ever identified. The vast majority of them are new to science. They are directly tied to the life expectancy of human beings. All but two of these SNPs are brand new to science. The research was made possible thanks to the support of the Swiss Initiative in Systems Biology. The findings were recently published in Nature Communications.

The Genome's Role

The length of an individual's life is predominantly determined by his environment. As an example, the place one resides, his level of wealth, dietary intake and whether he smokes all play major roles in how long he will live. Yet between one-quarter and one-third of variations in life expectancy arelikely determined by the genome.

About the Discovery

Scientists think variations at certain locations along DNA sequences, referred to as nucleotide polymorphisms (SNPs), provide clues about the genetic aspect of lifespan. Yet only two of these markers have been identified. The Swiss research team comprised of experts from Switzerland's Institute for Bioinformatics, the University of Lausanne, the Swiss Federal Institute of Technology in Lausanne and the Lausanne University Hospital have utilized an innovative computational means of pinpointing a remarkable 16 SNPs tied to lifespan. It is the largest group of genetic markers associated with lifespan ever discovered.

How the Discovery was Made

The Swiss research team studied a data set comprised of more than 116,000 people derived from a United Kingdom Biobank study. They analyzed about 2.3 million SNPs. Priority was given to DNA variations known to be associated with sickness tied to age in order to scan the genome in a highly effective manner. The research determined one in ten individuals carries a configuration of these newly identified markers that can decrease lifespan by more than a year versus the population at large.

The majority of the newly identified SNPs were linked to several different risk factors or diseases like a predisposition to develop schizophrenia or the likelihood of developing a drug addiction. It is clear that it is not as simple as pinpointing places along DNA molecules that code for a distinct lifespan. The research performed by the Swiss scientists approached the links between longevity and genetics in more of a holistic manner.

Read the original:
16 Genetic Markers Linked to Lifespan | Worldhealth.net Anti-Aging ... - Anti Aging News

Read More...

Page 14«..10..13141516..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick