Genes, ageing and longevity in humans: Problems, advantages and perspectives

22
Genes, ageing and longevity in humans: Problems, advantages and perspectives S. SALVIOLI 1,2,3 , F. OLIVIERI 4 , F. MARCHEGIANI 4 , M. CARDELLI 4 , A. SANTORO 1,2 , E. BELLAVISTA 1 , M. MISHTO 1,2 , L. INVIDIA 1 , M. CAPRI 1,2 , S. VALENSIN 1,2 , F. SEVINI 1,2 , E. CEVENINI 1,2 , L. CELANI 1,2 , F. LESCAI 1,2,3 , E. GONOS 5 , C. CARUSO 6 , G. PAOLISSO 7 , G. DE BENEDICTIS 8 , D. MONTI 9 , & C. FRANCESCHI 1,2,3,4 1 Department of Experimental Pathology, University of Bologna, via S. Giacomo 12, 40126 Bologna, Italy, 2 Centro Interdipartimentale “L. Galvani”, via S. Giacomo 12, 40126 Bologna, Italy, 3 ER-GenTech laboratory, via Saragat 1, 44100 Ferrara, Italy, 4 Department of Gerontological Sciences, I.N.R.C.A., via Birarelli 8, 60121 Ancona, Italy, 5 National Hellenic Research Foundation, Institute of Biological Research and Biotechnology, 48 Vas. Constantinou Avenue, Athens 11635, Greece, 6 Immunosenescence Unit, Department of Pathobiology, University of Palermo, Palermo, Italy, 7 Department of Geriatric Medicine and Metabolic Diseases, University of Naples, Naples, Italy, 8 Department of Cell Biology, University of Calabria, Rende, Italy, and 9 Department of Experimental Pathology and Oncology, University of Florence, viale Morgagni 50, 50134 Florence, Italy Accepted by Dr T. Grune (Received 15 June 2006; in revised form 19 July 2006) Abstract Many epidemiological data indicate the presence of a strong familial component of longevity that is largely determined by genetics, and a number of possible associations between longevity and allelic variants of genes have been described. A breakthrough strategy to get insight into the genetics of longevity is the study of centenarians, the best example of successful ageing. We review the main results regarding nuclear genes as well as the mitochondrial genome, focusing on the investigations performed on Italian centenarians, compared to those from other countries. These studies produced interesting results on many putative “longevity genes”. Nevertheless, many discrepancies are reported, likely due to the population-specific interactions between gene pools and environment. New approaches, including large-scale studies using high-throughput techniques, are urgently needed to overcome the limits of traditional association studies performed on a limited number of polymorphisms in order to make substantial progress to disentangle the genetics of a trait as complex as human longevity. Keywords: Genetic polymorphisms, ageing, longevity, centenarians, association studies, mitochondrial DNA Introduction Several years ago, Harman proposed the free radical theory of ageing [1] that has been last revalued and esteemed to be one of the candidates to explain the biology of ageing. This theory purports that ageing is mainly caused by oxidative stress that develops when the well-regulated balance between pro-oxidants and antioxidants gets out of control in favour of the pro- oxidants. Oxidants are mainly produced as by- products of normal aerobic metabolism, but also by phagocytic cells, and during lipid peroxidation. They induce oxidative damage on several substrates (nucleic acids, lipids and proteins). The cell is equipped with a series of antioxidant systems in order to cope with the ISSN 1071-5762 print/ISSN 1029-2470 online q 2006 Informa UK Ltd. DOI: 10.1080/10715760600917136 Correspondence: C. Franceschi, Department of Experimental Pathology, University of Bologna, via S. Giacomo 12, 40126 Bologna, Italy. Tel: 39 051 2094743. Fax: 39 051 2094747. E-mail: [email protected] Free Radical Research, December 2006; 40(12): 1303–1323

Transcript of Genes, ageing and longevity in humans: Problems, advantages and perspectives

Genes, ageing and longevity in humans: Problems, advantages andperspectives

S. SALVIOLI1,2,3, F. OLIVIERI4, F. MARCHEGIANI4, M. CARDELLI4, A. SANTORO1,2,

E. BELLAVISTA1, M. MISHTO1,2, L. INVIDIA1, M. CAPRI1,2, S. VALENSIN1,2, F. SEVINI1,2,

E. CEVENINI1,2, L. CELANI1,2, F. LESCAI1,2,3, E. GONOS5, C. CARUSO6, G. PAOLISSO7,

G. DE BENEDICTIS8, D. MONTI9, & C. FRANCESCHI1,2,3,4

1Department of Experimental Pathology, University of Bologna, via S. Giacomo 12, 40126 Bologna, Italy, 2Centro

Interdipartimentale “L. Galvani”, via S. Giacomo 12, 40126 Bologna, Italy, 3ER-GenTech laboratory, via Saragat 1, 44100

Ferrara, Italy, 4Department of Gerontological Sciences, I.N.R.C.A., via Birarelli 8, 60121 Ancona, Italy, 5National Hellenic

Research Foundation, Institute of Biological Research and Biotechnology, 48 Vas. Constantinou Avenue, Athens 11635, Greece,6Immunosenescence Unit, Department of Pathobiology, University of Palermo, Palermo, Italy, 7Department of Geriatric

Medicine and Metabolic Diseases, University of Naples, Naples, Italy, 8Department of Cell Biology, University of Calabria,

Rende, Italy, and 9Department of Experimental Pathology and Oncology, University of Florence, viale Morgagni 50, 50134

Florence, Italy

Accepted by Dr T. Grune

(Received 15 June 2006; in revised form 19 July 2006)

AbstractMany epidemiological data indicate the presence of a strong familial component of longevity that is largely determined bygenetics, and a number of possible associations between longevity and allelic variants of genes have been described. Abreakthrough strategy to get insight into the genetics of longevity is the study of centenarians, the best example of successfulageing. We review the main results regarding nuclear genes as well as the mitochondrial genome, focusing on the investigationsperformed on Italian centenarians, compared to those from other countries. These studies produced interesting results onmany putative “longevity genes”. Nevertheless, many discrepancies are reported, likely due to the population-specificinteractions between gene pools and environment. New approaches, including large-scale studies using high-throughputtechniques, are urgently needed to overcome the limits of traditional association studies performed on a limited number ofpolymorphisms in order to make substantial progress to disentangle the genetics of a trait as complex as human longevity.

Keywords: Genetic polymorphisms, ageing, longevity, centenarians, association studies, mitochondrial DNA

Introduction

Several years ago, Harman proposed the free radical

theory of ageing [1] that has been last revalued and

esteemed to be one of the candidates to explain the

biology of ageing. This theory purports that ageing is

mainly caused by oxidative stress that develops when

the well-regulated balance between pro-oxidants and

antioxidants gets out of control in favour of the pro-

oxidants. Oxidants are mainly produced as by-

products of normal aerobic metabolism, but also by

phagocytic cells, and during lipid peroxidation. They

induce oxidative damage on several substrates (nucleic

acids, lipids and proteins). The cell is equipped with a

series of antioxidant systems in order to cope with the

ISSN 1071-5762 print/ISSN 1029-2470 online q 2006 Informa UK Ltd.

DOI: 10.1080/10715760600917136

Correspondence: C. Franceschi, Department of Experimental Pathology, University of Bologna, via S. Giacomo 12, 40126 Bologna, Italy.Tel: 39 051 2094743. Fax: 39 051 2094747. E-mail: [email protected]

Free Radical Research, December 2006; 40(12): 1303–1323

inescapable and long lasting production of radical

oxygen species (ROS). According to the Harman’s

theory, it is postulated that genes whose product are

part of such antioxidant systems deeply impact on

ageing and longevity. In this review, we will focus on

genes involved in antioxidant systems as well as in

biochemical pathways affected by or sensitive to

oxidative damage. We will review the available data

and hypotheses mainly regarding association studies

on polymorphisms of such genes and human ageing

and/or longevity, focusing in particular on studies on

exceptionally long-living subjects such as centenar-

ians, who are expected to maximize the possible

contribution of genetics to longevity.

Paraoxonase1

In western countries, the major cause of morbidity and

premature death is atherosclerotic related disease [2].

ROS have been demonstrated to play a role in

inflammatory responses and lipidic homeostasis that

lead eventually to the development of atherosclerosis.

In particular, the peroxidation of low density

lipoproteins (LDL) and the capability of oxidized

lipoprotein-associated proteins to modulate local

inflammatory response are recognized to be a major

cause of atherosclerosis [3,4]. Human serum Para-

oxonase1 (PON1) is an A-esterase with peroxidase-

like activity present on the surface of high density

lipoprotein (HDL) and it is highly involved in the

prevention of LDL oxidation [5,6]. It is to note that,

under oxidative stress conditions, also the HDL

become a substrate for lipidic peroxidation. The

following reduction of HDL constitutes an indepen-

dent risk factor for atherosclerosis and furthermore

their oxidation remarkably increases the risk. It has

been observed that PON1 is able to maintain the anti-

atherogenic activity of HDL. In vitro assays have shown

that PON1 can inhibit LDL lipid peroxidation and

inactivate LDL-derived oxidized phospholipids. This

could potentially reduce the amount of the oxidized

lipids involved in the initiation of atherosclerosis [7].

HDL obtained from PON1-knock-out mice cannot

prevent oxidation of LDL in a co-culture model

simulating the artery wall, and their macrophages

contain more oxidized lipids and have an increased

capability to oxidize LDL. These mice are more prone

to atherosclerosis than their wild-type counterparts,

probably as a consequence of increased oxidative stress

[8]. The apoE knock-out (apoE2/2) mouse is a well

known model of atherosclerotic lesion development.

The double knock-out apoE2/2 /PON12/2 mouse has

a susceptibility to develop atherosclerosis which is

higher than that of the single knock-out apoE2/2

mouse suggesting a role for PON1 in the prevention of

the disease [9]. On the contrary, mice overexpressing

PON1 are protected against atherosclerosis in both a

wild-type and an apoE2/2 background [10].

Because of its involvement in antioxidant defence,

PON1 has been taken into consideration as a

candidate longevity gene in a series of association

studies [11–14]. The paraoxonase gene family

contains at least three members, named PON1,

PON2 and PON3, which are located on chromosome

7q21.3–22.1 [15]. The most studied PON1 gene

polymorphisms are due to an amino acid substitution

at position 192 (Gln-Arg) and at position 55 (Leu-

Met) in the coding region of the gene. Alleles at

Codon 192 (Q and R alleles) and 55 (L and M alleles)

in PON1 locus have been associated with enzymatic

activity and concentration, respectively [16–18]. In

recent years, PON1 gene variants have been widely

investigated, especially for their possible role in the

development or severity of coronary artery disease

(CAD). Since cardiovascular diseases are common

age-related diseases, it is possible that exceptionally

long-living subjects such as centenarians, who escaped

the major age-related diseases including CAD, are

enriched in particular polymorphic variants of PON1

gene. Thus, we analysed the genetic variability at

PON1 locus in such individuals, and performed an

analysis of PON1 192 and 55 polymorphisms in a

sample of 579 young Italian individuals and 308

Italian centenarians. We found that the frequency of R

allele, and consequently of R þ carriers (QR þ RR

individuals), significantly increased from young

people to centenarians, thus conferring a small

survival advantage for subjects carrying R allele

compared to subjects carrying Q allele (OR 1.3, CI

1.04–1.6; p ¼ 0.02). Furthermore, when the PON1

192 polymorphism was analyzed together with the

polymorphism at position 55, we found that, among

R þ subjects, the phenomenon was due to an increase

of people carrying M allele at codon 55 locus [11].

These results were subsequently confirmed in a large

combined group of Italian centenarians and Northern

Ireland octo/nonagenarians and their respective con-

trols for a total of 1479 subjects. In particular, a

significant increase in the frequency of R þ carriers in

Italian centenarians and Northern Ireland octo/nona-

genarian subjects was found in comparison with their

younger control subjects (53.6 vs. 46.1%, p ¼ 0.004)

[12,13] and a logistic regression analysis on the whole

sample highlighted that, when comparing R and Q

alleles, there was a survival advantage for octo/nona-

genarian/centenarian subjects who carried the R allele

(OR 1.3, CI 1.1–1.5; p ¼ 0.007) [12]. In a recent

study, we have found that in people with high risk to

develop CVD such as hypertensive patients, the

frequency of PON1 192 RR genotype is increased in

comparison to non-hypertensive subjects (14.3 vs.

5%, respectively) [19]. This result is apparently in

contrast with our previous studies in which we

reported that subjects with PON1 192 QR genotype

have a higher probability to attain longevity and that

the frequency of PON1 192 RR genotype was around

S. Salvioli et al.1304

9.4% in long-living people [13]. These conflicting

findings can be explained by the hypothesis that

PON1 192 RR genotype has a biphasic, antagonistic

behaviour: it could be considered a risk factor for

hypertension and CVD in the elderly, but a protective

factor for nonagenarians and centenarians. Only few

other studies have taken into consideration the impact

of PON1 gene variation on survival at extremely

advanced age [20–22]. Heijmans and colleagues

performed a population-based study of PON1 192

and 55 polymorphisms among subjects aged 85 years

and over in a cross-sectional and prospective design

[20]. The study revealed that no difference in

genotype distributions between elderly and young

subjects was present and that the risk of all-cause and

cardiovascular mortality was not increased in elderly

subjects with the PON1 LL or RR genotype.

A following paper published by Christiansen et al.

[21] investigated the impact of PON1 gene variability

on mortality using a sample of 1932 Danish

individuals aged 47–93 years. In this study, no

difference in the genotype and haplotype distributions

between different age groups was found.

More recently, Tan et al. [22] presented a novel

survival analysis model that combined population

survival with individual genotype and phenotype

information in assessing the genetic association with

human longevity in cohort studies. In this study, the

Authors considered three SNPs in the human PON1

gene (the two in the coding region, amino acids M55L

and Q192R, and one additional in the promoter

region, C-107T) and they measured the association of

PON1 gene polymorphisms with human survival at

advanced ages in the Danish female 1905 birth cohort

followed from 1998 to 2005. The application of this

novel model to PON1 genotype data detected a

haplotype (T-Q-L) that significantly reduces the risk

of death and thus reveals and stresses the important

role of PON1 genetic variation in promoting human

survival at advanced ages.

Several authors so far have suggested that the

antiatherogenic role of PON1 enzyme is to reduce

oxidative stress during ageing and counteract the

deleterious effects of atherogenesis. Accordingly, the

measure of paraoxonase and arylesterase activities was

investigated in subjects prone to development of

atherosclerosis such as subjects affected by Type 1 or

Type 2 diabetes, familial hypercholesterolaemia, renal

disease and CAD. For all of these age-related

pathologies, a significant decrease of paraoxonase

activity was observed (reviewed in [23]). The increase

of atherosclerosis with age could be due to the

increased susceptibility of LDL and HDL to oxidation

[24,25]. Because of the tight relationship between

PON1 and atherogenesis, some studies have investi-

gated the involvement of PON1 in the ageing process.

In a sample of 129 healthy subjects aged between 22

and 89 years, Seres et al. found that serum PON1

activity significantly decreased with age, while its

arylesterase activity, as well as its concentration and

HDL concentration, remained unchanged with age.

Moreover, the HDL from elderly subjects resulted

more susceptible to oxidative stress than HDL from

young subjects [24]. The authors concluded that the

development of oxidative stress conditions with ageing

could explain, in part, the reduction in PON1 activity.

They also proposed that factors which could

compensate the atherosclerosis could be altered.

Recently, we have investigated the possible role of

PON1 genotypes and phenotypes in aged people free

of overt diseases, focusing our attention especially on

nonagenarians/centenarians. In a sample of 229

participants from 22 to 104 years of age, divided

into young, elderly and nonagenarians/centenarians,

we have evaluated the role played by PON1 genotypes,

paraoxonase actvity, arylesterase activity and paraox-

onase mass concentration in the chance for young and

elderly people to become extremely old. We have

found that paraoxonase activity, arylesterase activity

and paraoxonase specific activity were significantly

lower in nonagenarians/centenarians compared to

elderly and young individuals. On the contrary,

paraoxonase mass concentration was found decreased

in the elderly group and again increased in the

nonagenarian/centenarian group. These parameters

were also analyzed in relation to the PON1 192 and

PON1 55 polymorphisms. This analysis demonstrated

a genetic control for paraoxonase activity that was

maintained throughout life, also in the oldest old

group. This activity showed different mean values

among R and M carriers, having R þ and M 2

carriers the highest paraoxonase activity levels. Using

a multinomial regression logistic model we also

demonstrated that both paraoxonase activity and

R þ and M 2 carriers contributed significantly to the

explanation of the longevity phenotype [14]. On the

whole, we concluded that PON1 activity and genetics

are deeply interconnected throughout life and that

both play a role in human longevity.

Genes for apolipoproteins and proteins related

to lipid metabolism

Beside proteins associated with HDL particles such as

PON1, a large number of studies have considered the

involvement in ageing and longevity of allelic variants

in genes encoding apolipoproteins (APOE, APOB,

APOC1, APOC2, APOC3, APOA1 and APOa),

transfer proteins [microsomal transfer protein

(MTP), cholesteryl ester transfer protein (CETP)]

and transcription factors involved in lipid metabolism

[peroxisome proliferator-activated receptor g

(PPARg)]. One of the most explored loci is that of

the apolipoprotein E (APOE) gene. This locus

appears to be one of the few for which a worldwide

consensus has been reached in terms of negative

Genes, ageing and longevity in humans 1305

association with longevity. Indeed, a higher frequency

of the APOE4 allele in young subjects compared with

old subjects (octogenarians, nonagenarians and

centenarians) has been observed, concluding that the

presence of the APOE4 allele is associated with

decreased lifespan, likely because of an increased

incidence of cardiovascular as well as neurodegenera-

tive diseases [26,27]. Much less is known about other

APO genes and ageing and longevity (reviewed by

Ordovas and Mooser) [28]. In particular, we

investigated common APOB polymorphisms in a

sample of 143 centenarians from southern Italy and a

control sample of 158 individuals [29]. We found that

the frequency of 30APOB-VNTR alleles with fewer

than 35 repeats (small, S) was significantly lower in

centenarians than in controls. A further analysis of

seven different age cohorts (697 individuals from 10 to

109 years old) revealed that there is an age-related

change in the 30APOB-VNTR SS genotype. In

particular, a convex trajectory of the frequency of SS

homozygotes was found. The frequency of SS in the

genotype pool increased from the group aged 10–19

years (3.06 ^ 1.74%) to that aged 40–49 years

(8.51 ^ 4.07%), then it declined reaching the

minimum value in centenarians (1.58 ^ 0.90%)

[30]. This study was repeated in a sample of Danish

people, and neither genotype nor allele frequencies

differed between centenarians and 20–64-year-old

subjects [31]. Furthermore, the demographic-genetic

approach revealed in Danes a significant gene-sex

interaction relevant to Long alleles (more than 37

repeats). The different findings in Denmark and Italy

suggest that these associations between APOB

polymorphism and longevity are population-specific,

and heavily affected by the population-specific genetic

and environmental history. In further studies, we

found that the S alleles lower the average values of

serum Total Cholesterol and LDL-Cholesterol, while

the alleles M and L have no significant effect on the

lipidemic phenotype [32]. Thus, the S alleles would be

advantageous in adults by protecting from coronary

artheriosclerosis and related diseases, while dangerous

in the elderly, probably by lowering serum cholesterol

below a critical threshold. This could explain the

convex frequency trajectory of SS genotypes pre-

viously observed.

Other studies performed on apolipoprotein A1

(APOA1), apolipoprotein C3 (APOC3) and apolipo-

protein A4 (APOA4) that are tandemly organized

within a short region on chromosome 11q23–q24,

have revealed a MspI-RFLP in APOA1 (275 nt from

the transcription starting site) that modulates the

serum level of LDL. In particular, the A allele reduces

the levels of serum LDL-cholesterol, while the allele P

increases them. Surprisingly, the P allele is more

represented in male centenarians than in 46–80 year

old males [33]. This unexpected finding has been

explained by suggesting that this allele can be risky in

middle-age but protective at very advanced age, and

that the effect of a mutation may depend on the age-

related physiological scenario in which the mutation

acts.

In the case of CETP gene, two different studies have

highlighted the importance of the populations

considered. This gene is characterized by the presence

of a polymorphism at codon 405 (I405V), and the VV

genotype has been reported to be associated with

longevity in Ashkenazi Jewish [34], but not in Italian

population [35]. This seems to indicate that longevity

is not linked to a specific polymorphism of CETP, but

rather that this polymorphism likely interacts with

other factors (gene pool, environment, chance) which

differ from population to population, to determine or

not the “longevity” phenotype.

ApoJ

ApoJ was firstly identified in ram rete testis fluid in

1983 as a secreted glycoprotein enhancing cell

aggregation in vitro (named thus as clusterin) [36].

In humans, it was firstly purified from serum and the

cloned gene was named as CLI (complement cytolysis

inhibitor) [37], SP-40,40 (secreted protein 40,40)

[38] or Apolipoprotein J (ApoJ) [39] due to

similarities with other known apolipoproteins. A

comparison of the ApoJ protein sequences among

mammalian species reveals a high degree of conserva-

tion of ,70–85%, while attempts to clone its

homologues in the worm or the fly by using specific

primers spanning the conserved regions of the gene

appeared negative (our unpublished results). These

two observations along with its wide distribution in

animal tissues and the absence of studies highlighting

functional ApoJ polymorphisms in humans [40,41]

suggest that the protein has evolved in vertebrates to

accomplish a function of fundamental biological

importance.

ApoJ protein is constitutively secreted by a number

of cell types including epithelial and neuronal cells,

while in cells featuring a regulated exocytotic pathway

its secretion depends on the appropriate exogenous

stimulus (reviewed in Trougakos and Gonos 2002)

[42]. However, despite the systematic and combined

effort of several groups, ApoJ function has remained

elusive, the main cause being the intriguingly distinct

and usually opposing functions proposed in an array of

various cell types and tissues. We have developed a

clonal system of rat embryo fibroblast conditionally

immortalized cell lines which undergo senescence

upon SV40 Tantigen inactivation [43,44] and we have

cloned Clusterin/Apolipoprotein J as a senescence

associated gene [45]. We have shown that CLU/ApoJ

is up-regulated during replicative senescence and

stress-induced-premature senescence in various mam-

malian tissues [45,46,47]. Moreover, we have found

that CLU/ApoJ accumulates in human serum during

S. Salvioli et al.1306

in vivo ageing as well as in several age-related diseases,

such diabetes type II, myocardial infarction or

coronary heart [48]. We have also investigated the

effects of CLU/ApoJ on cellular growth and survival

by taking advantage of three human osteosarcoma

(OS) cell lines, since these cells express distinct

endogenous CLU/ApoJ levels and, moreover, they

have distinct and characterized genetic backgrounds.

Following CLU/ApoJ forced over-expression by

means of an artificial transgene we found that

extracellular CLU/ApoJ inhibits cell death in all

three OS cell lines assayed. Interestingly, intracellular

CLU/ApoJ has different effects on cellular prolifer-

ation and survival in these cell lines. Transgenic

KHOS cell lines adapted to moderate intracellular

CLU/ApoJ levels and became resistant to both

genotoxic and oxidative stress, whereas transgenic

SaOS and U2OS cell lines adapted to high intracellu-

lar CLU/ApoJ amounts and were sensitive to the same

cytotoxic agents [49]. To conclude about the primary

CLU/ApoJ function we used small interfering RNAs

(siRNAs) to silence CLU/ApoJ gene expression. Our

data demonstrate that CLU/ApoJ knock down

induces significant reduction of cellular growth,

higher rates of spontaneous endogenous apoptosis

and reduced plating efficiency. These effects are

enhanced in those cell lines expressing high endogen-

ous CLU/ApoJ levels. Moreover, CLU/ApoJ knock

down OS and prostate cancer cells were dramatically

sensitized to various apoptosis-inducing agents. By

assaying the expression levels of several proteins

involved in regulating apoptosis we found that

CLU/ApoJ silencing results in the down-regulation

of the anti-apoptotic molecule bcl-2. In U2OS cells,

which have functional p53, sCLU/ApoJ knock down

apart from bcl-2 down-regulation is also accompanied

by p53 accumulation and up-regulation of its down-

stream pro-apoptotic effector bax and the cyclin-

dependent kinase inhibitor, p21 [50]. Overall, our

results reveal that CLU/ApoJ is a central molecule in

cell homeostasis that exerts a potent cytoprotective

function.

As far as MTP is regarded, it recently attracted great

attention after the finding that a region of chromo-

some 4 appeared to be linked to exceptional longevity

in US Caucasians [51]. In this region (4q25),

containing about 50 genes, a SNPs analysis was

performed and indicated a two-SNPs haplotype of

microsomal transfer protein (MTP) as associated to

longevity [52]. However, further studies were unable

to reproduce these findings on 4q25 and MTP, and a

meta-analysis confirmed this lack of association [53].

IGF-1 and IGF-1 pathway

Hyperglycemia can increase oxidative stress through

a mechanism that promotes the formation of

advanced glycosylation end-products (AGEs) and

PKC activation [54]. Once formed, AGE-protein

adducts are stable and virtually irreversible. More-

over, during oxidative stress, AGEs formation

increases substantially [55]. The glycosilation of

LDL represents the cause leading for the modifi-

cation of the putative LDL receptor binding domain.

These glycated LDL, that are no more recognized by

LDL receptors, become the substrate for macro-

phages that stimulate foam cells formation and

promote atherosclerosis. Monocyte-macrophage

interaction with AGEs also results in the production

of mediators such as interleukin-1, tumor necrosis

factor-a, platelet-derived growth factor, and insulin

growth factor-I [56,57,58], which have a pivotal role

in the pathogenesis of atherosclerosis [59].

As IGF-1 is an important regulator of the insulin

action by stimulating the glucose transport, Paolisso

et al. have investigated this molecule in centenarians

and they have found a decrease in IGF-1 plasma levels

[60,61] together with a well preserved insulin action,

thus indicating that insulin responsiveness is funda-

mental to reach the extreme limits of human life span

[62,63]. In contrast, several papers reported low levels

of IGF-1 to be associated with cardiovascular

pathologies [64,65,66,67]. Also, we have found that

in aged people high levels of this hormone are

beneficial especially for the physical performance and

the maintenance of muscle strength [68]. Apparently,

these last results do not fit with those obtained from

centenarians showing that reduced IGF-1 is associ-

ated with longevity [61]. Indeed, this apparent

paradox could be explained with the evolutionary

theory of ageing which assumes that expression of

particular genes could be beneficial early in life, but

could become detrimental with age [69]. In this case,

high levels of IGF-1 can be helpful to avoid disability

and frailty in the elderly, but later in life (after the age

85), low levels of IGF-1 could promote survival by

avoiding cancer development and growth.

Recent evidences from evolutionary biology show

that, in a variety of model systems from invertebrates

to mammals (yeast, worms, fruit flies and rodents),

mutations in genes that share similarities with the

human genes involved in the insulin/IGF-1 signal

response pathway are responsible for an extension of

life span [70–74]. On the basis of this consideration

and on the basis of the findings that insulin

responsiveness impacts on human longevity, we tested

the hypothesis that human loci which share similarities

with genes that regulate the insulin pathway in

invertebrates could affect human longevity [61]. In

particular, we investigated polymorphisms at IGF-1R

(insulin-like growth factor type 1 receptor), PI3KCB

(phosphoinositide 3-kinase), IRS-1 (insulin receptor

substrate-1) and FOXO1A genes as well as their

possible effects on IGF-1 plasma levels. These genes

represent key molecules of the insulin/IGF-1 pathway

and their sequential activation determines a cascade

Genes, ageing and longevity in humans 1307

activation signal. This study was performed on 496

Caucasian healthy subjects subcategorized into two

groups by splitting the sample at the age of 85 years.

The major findings emerging from the study were that

free IGF-1 plasma levels displayed an age-related

decrease and that these levels were affected by the

polymorphisms at IGF-1R and PI3KCB. The poly-

morphisms considered in the study were the following:

a G to A transition at nucleotide 3174 in exon 16

of the IGF-1R and a T–C transition located

359 bp upstream from the starting codon of

PI3KCB. Again, subjects carrying at least one A allele

at the IGF-1R locus have lower plasma IGF-1 levels

than the rest of the population and these subjects are

more represented in the long-lived individuals in

respect to younger subjects. Accordingly, Holzenber-

ger and colleagues have demonstrated that the

inactivation of IGF-1R prolongs the life span and

increases the resistance to oxidative stress in mice [75].

When the polymorphisms of IGF-1R and PI3KCB

were considered together, another interesting result

emerged, i.e. the proportion of IGF-1R/PI3KCB-

Aþ/Tþ carriers is significantly increased among long-

lived individuals. Intriguingly, in animal models like C.

elegans and M. musculus the down-regulation of IGF-1

pathway is associated with an extension of life span

[76–78], whereas high levels of IGF-1 are associated

with a shortened life span [78]. Moreover, caloric

restriction reduces plasma free IGF-1 levels [80,81]

and cannot be excluded that the peculiar nutritional

regimen of centenarians could resemble that of

subjects who have undergone a moderate caloric

restriction. To date, this study represents the first

indication that also in humans, polymorphisms of

genes regulating the IGF-1/insulin pathway affect

longevity, contributing to the hypothesis that the

impact of the IGF-1/insulin pathway on longevity is a

property that has been evolutionarily conserved

throughout the animal kingdom. Further studies

performed in European and Japanese populations

give results that are in agreement with those previously

reported and add further indications that IGF-1

pathway is involved in human longevity [82,83].

Taking into account that high levels of circulating

IGF-1 are positively associated with muscle strength

in the elderly [68], we can hypothesize that a

phenomenon of antagonist pleiotropy occurs in ageing

and longevity regarding IGF-1. According to such an

interpretation, high level of IGF-1 could be beneficial

in the elderly as a protective factor toward sarcopenia

but the price to pay would be a higher risk of cancer

incidence. Conversely, in extreme ages and in

centenarians, low levels of IGF-1 would give a survival

advantage (low cancer incidence) but in this case the

price to pay would be physical frailty and loss of

muscle mass and strength.

Furthermore, it is interesting to note that YTHDF2,

a gene recently found to be associated with longevity

in a study conducted on 412 Italian participants,

including 137 centenarians, and expressed in a wide

variety of organs but mainly in testis, placenta and

pancreas [84], has been reported to be regulated by

high glucose concentration (see GenBank accession

no. AF192968 [85]). Thus, it could be worthwhile to

test the hypothesis of a possible role of YTHDF2 in

carbohydrate homeostasis and in the insulin/IGF-1

signaling pathway.

Genes for cytokines

ROS are signaling molecules for a wide variety of

inflammatory stimuli and serve as signaling messen-

gers for the evolution and perpetuation of the

inflammatory process, but relatively little is known

about the relationship between oxidative stress and

production of inflammatory cytokines [86,87]. Oxi-

dative stress and pro-inflammatory cytokines trigger

common signal transduction pathways that lead to

amplification of the inflammatory cascade, mainly

through activation of mitogen-activated protein

kinases (MAPK) and nuclear factor kappaB (NF-

kB). ROS could activate ubiquitous transcription

factors, such as NF-kB and AP-1, up-regulating

monocyte chemotactic molecules expression, respon-

sible for the initial inflammatory events and up-

regulating the cytokines gene expression [88,89].

Cytokines, in turn, could cause the formation of nitric

oxide (NO) that combines with superoxide to form the

potent oxidant peroxynitrite, thus generating a vicious

circle [90]. As an example, it was reported that

oxidative stress increases the production of tumor

necrosis factor alpha (TNF-a), a potent pro-inflam-

matory cytokine; TNF-a impairs the production of

glutathione (GSH), an intracellular antioxidant,

leading to a pathogenic “loop” [89]. Moreover, ROS

can modulate apoptosis in a variety of cell types,

including human inflammatory cells [91]. From

invertebrate to humans the cellular response to a

variety of stressors involves the up-regulation of

mediators such as ROS and pro-inflammatory

cytokines, such as interleukin-1 (IL-1), interleukin-6

(IL-6) and TNF-a and this type of cellular response

appears to be highly maintained during the evolution

[69]. On this basis, the stress response and inflam-

mation could be considered an integrated evolutionary

conserved defence network, and oxidative stress could

modulate immune response and inflammation, balan-

cing the pro-inflammatory/anti-inflammatory cyto-

kines production [92,13]. In particular, IL-6, TNF-a

and IL-1 are pro-inflammatory pleiotropic cytokines

capable of regulating proliferation, differentiation and

activity of a variety of cell types [93,94]. TGF-b1 is a

multifunctional cytokine that regulates cell prolifer-

ation, differentiation and migration, and it was

considered an anti-inflammatory molecule [95].

Interleukin-10 (IL-10) is a powerful cytokine that

S. Salvioli et al.1308

inhibits lymphocyte replication and secretion of

inflammatory cytokines [96]. Ageing seems to be

associated with a pro-inflammatory shift in cytokine

expression profile, as suggested by the increased

circulating levels of pro-inflammatory cytokines in

healthy elderly humans, as a consequence of age-

related alterations in the endocrine system, cellular

metabolism, and redox regulation [94,97]. In order to

test whether the model of inflamm-ageing does apply

not only to human ageing but also to human longevity,

we performed a series of studies on such cytokines in

centenarians. Data obtained in these studies suggested

that longevity is associated with the capability of cells

to cope with a variety of stressors, including oxidative

stress, modulating the inflammatory response [13].

Moreover, it has been demonstrated that centenarians

have lower oxidative stress than subjects 70–99 years

old, suggesting that a reduction of protective mechan-

isms against ROS during ageing could induce a greater

toll of oxidative damage, deleterious for longevity

[98,99]. In summary, the capability to maintain a

lower production of pro-inflammatory cytokines and

the capability to be well protected from oxidative stress

appears to be favourable for reaching the extreme limit

of human life span in good health conditions and could

be genetically controlled [69,99].

Moreover, recent data suggest that increased levels

of pro-inflammatory cytokines, such as IL-6, IL-1 and

TNF-alpha and a decreased production of anti-

inflammatory cytokines such as TGF-b1 and IL-10

during ageing, represents the biological background

favouring the susceptibility to the major age-related

diseases, such as cancer, cardiovascular disease,

insulin resistance, type 2 diabetes and Alzheimer’s

disease and can also be strong short-term predictors of

mortality associated with age-related chronic diseases

[100–104,69,13].

On the whole, data obtained from genetic analysis

of candidate genes in centenarians and in sample of

patients suffering by major age-related diseases,

confirmed the hypothesis that genes related to

inflammation are implicated in human longevity and

in the susceptibility to many age-related diseases. A

great amount of papers reported a positive association

between some polymorphic markers of IL-6 gene and

longevity, and the capability of producing low levels of

IL-6 throughout life span appears to be beneficial for

longevity [13,94,105,106]. Nevertheless, it appears

that IL-6 polymorphism does not affect life expect-

ancy neither in the Sardinian population, nor in

people from Southern Italy, suggesting that the effect

of IL-6 polymorphism on longevity might be

population-specific and dependent on gene-environ-

ment interactions [107,108].

Particular attention was devoted to two anti-

inflammatory cytokines, i.e. TGF-b1 and IL-10. We

reported an association between TGF-b1 poly-

morphic markers and human longevity [109]. In this

study, the plasma levels of biologically active TGF-b1

were significantly increased in the elderly group,

independently from TGF-b1 genotypes.

The relation of some IL-10 polymorphisms, such as

T-819C, A-592C and A-1082G promoter SNPs, with

age was evaluated in different population samples,

suggesting that IL-10 may be a promising candidate

ageing-related gene [107,110–114]. In particular, IL-

10 and TNF-a have complex and opposing roles, and

an autoregulatory loop appears to exist [115].

Furthermore, their synergistic role in the control of

immune-inflammatory responses has been hypoth-

esised, and the interaction between TNF-a -308

and -1082 IL-10 polymorphisms promoter was

observed [112].

Instead, no particular polymorphism in the IL-1

gene cluster was reported to confer a survival advantage

in the last decades of life, and the observed age-related

increase in IL-1Ra plasma levels seems not to be

genetically controlled [93]. However, the studies of IL-

6 and TNF-a suggest that the levels of these pro-

inflammatory molecules are, to a large extent,

genetically determined, even in elderly individuals,

and therefore, it is important to elucidate the genetic

factors involved in the regulation of these proteins,

especially in age-related diseases [116]. Recently, many

associations between a great number of polymorphic

markers of cytokine genes and the incidence and/or the

prognosis of age-related diseases, such as cardiovas-

cular disease, type 2 diabetes, cancer and Alzheimer’s

disease have been reported [117–126].

An additive effect of variants of genes of cytokines

(IL-6 G174C) and of other genes involved in

metabolic pathways such as PPARg (Pro/Ala) on the

total variation of obesity-related factors (BMI, insulin

resistance, triglyceride levels and so on) has been

recently reported [126].

In summary, a great number of genetic markers

related to a pro-inflammatory phenotype are associ-

ated with the major age-related diseases, and have

been found to be under-represented in centenarians,

while genetic variants associated with an anti-

inflammatory activity have been found to be more

represented in centenarians, confirming that the

balancing during ageing between pro and anti-

inflammatory mechanisms is largely under genetic

control. Cytokines could be thus an important trait

that links mitochondria and oxidative stress on one

side and inflammation and ageing on the other side

[127].

SOD and PARP genes

Studies performed on animal models [128] and on

octo/nonagenarian subjects [129] evidenced that the

level of nonenzymatic antioxidants declines during

senescence, whereas systemic oxidant load tends to

increase in older adults [129]. For these reasons, the

Genes, ageing and longevity in humans 1309

enzymes involved in the inactivation of ROS

(“enzymatic antioxidants”), such as superoxide dis-

mutase (SOD), and enzymes involved in the repair of

the DNA injuries caused by oxidative damage or other

genotoxic stress, such as PARP (poly (ADP-ribose)

polymerase), are thought to play a key role in

controlling the rate of the ageing process, and have

been extensively studied in relation to longevity.

SOD enzymes catalyze the breakdown of super-

oxide into hydrogen peroxide and water and are

therefore central regulators of ROS levels [130].

Different SODs have evolved to inactivate both

intracellular and extracellular superoxide radicals

produced as by-products of metabolic oxidation.

Mammalian cells, in particular, contain a manganese

superoxide dismutase (Mn SOD/SOD2) localized

within the mitochondrial matrix [131], a copper- and

zinc-containing superoxide dismutase (Cu/Zn SOD/

SOD1) localized predominantly in cytoplasmic and

nuclear compartments [132], and a copper- and zinc-

containing SOD predominantly found in extracellular

compartments (EC SOD/SOD3) [133].

The Cu/ZnSOD and MnSOD overexpression has

been found to increase life-span in Drosophila

[134,135], while null mutations in these genes greatly

decrease viability and life-span in the same species

[136,137], and only MnSOD mutation has a severe

effect in mice [138].

The role of enzymatic antioxidants, SOD in

particular, in human ageing has been addressed by

different studies. The enzyme activities of SOD

(Cu/Zn-SOD), glutathione peroxidase, catalase and

glutathione reductase (GR) in erythrocytes was

analysed in a cross-sectional study [139] conducted

on 41 Danish centenarians and on 52 control subjects

(aged between 60 and 79 years). Cu/Zn-SOD activity

was found to be decreased in centenarians (particu-

larly in centenarians with the lowest cognitive and

physical functional capacity), and the result was

interpreted as a reduced demand for the enzyme at

lower metabolic rate and oxygen consumption

(whereas subjects with high GR activity were more

frequent than expected in centenarians).

In a different study [140], plasma levels of non-

enzymatic antioxidants (vitamin C, uric acid, vitamin

E, vitamin A, carotenoids, total thiol groups), and the

activity of enzymatic antioxidants were assayed in 32

healthy centenarians, 17 elderly subjects aged 80–99

years, 34 elderly subjects aged 60–79 years, and 24

adults aged less than 60 years. In particular, plasma

SOD, plasma glutathione peroxidase (GPX), and red

blood cell (RBC) SOD activities were measured.

A constant decrease with age of the nonenzymatic

antioxidants and a concomitant increase of the

enzymatic antioxidant activities were observed in the

three younger age classes, but not in centenarians,

who on the contrary showed the highest levels of

vitamins A and E, and activities of both RBC and

plasma SOD which were not increased or decreased

with respect to each other age class. From the results

of the above cited studies, it can be hypothesized that

normal ageing is probably characterized by a pro-

oxidant status, accompanied by a decrease of non-

enzymatic antioxidants and by a compensatory

increased activity of enzymatic antioxidants, whereas

healthy centenarians show a completely different

profile, in which high levels of vitamin A and vitamin

E are associated with normal or reduced activities of

enzymatic antyoxidants. A reduced cellular suscepti-

bility to free radicals attack in centenarians was also

suggested in studies [141–143] which analysed the

modifications of the plasma membrane composition in

centenarians. Altogether, these studies suggested that

the erythrocyte membranes from centenarians have

some distinct features in comparison with elderly

subjects, that might act in a protective way against

injuries [144].

Other genes whose products have an antioxidant

action include GSTT1. GSTT1 gene is a member of

the Glutathione-S-Transferase superfamily, involved

not only in protection against ROS, but also against

xenobiotics and exogenous carcinogens [145]. We

studied polymorphisms in CYP1A1, GSTT1 and

GSTM1 genes and found that centenarians have an

increased proportion of a deletion of GSTT1 gene

with respect to younger controls [146]. Accordingly,

the presence of one or two alleles of such a gene has

been observed to be correlated with an increase in

mortality [147,148].

Oxidative stress results in many damaging cellular

effects; among them, the effects on DNA are the most

harmful to the cell function. Oxidative stress induces

DNA single strand breaks and leads to the activation

of the DNA repair enzyme poly (ADP-ribose)

polymerase (PARP). Poly (ADP-ribosyl)ation is a

post-translational modification of proteins which is

catalyzed mostly by PARP-1 [149,150], an abundant

nuclear protein that binds to a DNA single-strand

break (SSB) and catalyses the formation of poly

(ADP-ribose) polymers on itself and other acceptor

proteins (Lindahl et al. 1995). Poly (ADP-ribose)

polymers formation is suggested to be important to

protect DNA from breaks and to attract DNA repair

proteins to the site of damage [151,152]. PARP-1 is

also involved in base excision repair (BER) [153]. Poly

(ADP-ribosyl)ation capacity in mononuclear blood

cells positively correlates with the species-specific

longevity of 13 mammalian species [154], and the

observed differences in the catalytic capacity may be

explained by sequence divergence in the PARP-1

protein [155]. Similarly, poly (ADP-ribosyl)ation

capacity of human limphoblastoid cells correlates

with longevity of the donors [156]. PARP-1 activity

appears to finely regulate the rate of genomic

instability events, caused by DNA-damaging agents

[157,150]. However, excessive activation of PARP-1

S. Salvioli et al.1310

can lead to cell death (suicide hypothesis), and it is

probably implicated in a variety of insults, including

cerebral and cardiac ischemia, 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine-induced Parkinsonism,

traumatic spinal cord injury and streptozotocin-

induced diabetes [158], and has been hypothesised

to be the central mediator in all the mechanisms by

which hyperglycaemia induces diabetic vascular

dysfunction [159]. It has been hypothesised that

PARPs might regulate cell fate as essential modulators

of death and survival transcriptional programs [158].

Recent results suggest that PARP-1 activity might

be impaired in the elderly. In particular, the results of

a recent study [160] suggest that increasing age is

associated with an impaired capacity of PARP-1 in

base excision DNA-repair, which in turn is suggested

to be due to the reduced bioavailability of zinc ions

observed in the elderly. Interestingly, the PARP-1

impairment seems to be enhanced in old infected

patients (acute and remission phases of broncho-

pneumonia infection), but reversed in centenarians,

where good zinc ion bioavailability and higher

capacity of PARP-1 in base excision DNA-repair

were observed [160].

The influence of the genetic variability of PARP and

SOD2 genes on the inter-individual variability of

human longevity has been tested in an association

study [161] in which a (gt)n repeat polymorphism in

exon 1 of the PARP-1 gene, and a T/C substitution

(which changes Valine16 to Alanine16) in the

mitochondrial targeting domain of the SOD2 gene

were analysed in two age groups respectively

composed of centenarians (109 from northern Italy

and 87 from southern Italy) and of younger control

subjects (from 10 to 85 years, 119 from northern Italy

and 239 from southern Italy) matched for sex and

geographic area. The results of this study suggested

that these polymorphisms do not affect inter-

individual variability in life expectancy.

Two other studies obtained negative results about

the possibile association of PARP-1 gene polymorph-

isms and human longevity. In the first [156], 437

DNA samples (239 centenarians and 198 controls)

were analysed for a polymorphic dinucleotide repeat

located in the promoter region of PARP-1, but no

significant enrichment of any of the alleles or

genotypes identified among centenarians or controls

was observed. In the same study, maximal oligonu-

cleotide-stimulated PARP-1 activity in lymphoblas-

toid cell lines was found to be significantly higher in

centenarians (n ¼ 49) than in younger controls

(n ¼ 51). The second study [162] was performed by

analysing two single nucleotide polymorphisms in the

PARP-1 gene in 648 DNA samples from a French

population (324 centenarians and 324 controls) and

even in this case no significant difference in genotype

frequency was found. Furthermore, none of the

genotype combinations at any polymorphic site

studied could be related to a high or low level of

poly(ADP-ribosyl)ation capacity, suggesting that the

longevity-related differences in the poly(ADP-ribosy-

l)ation capacity of human lymphoblastoid cell lines

cannot be explained by the genetic polymorphisms in

the PARP-1 coding sequence studied until now.

Nevertheless, experimental evidence seems to indicate

a link between poly(ADP ribosyl)ation and longevity

in mammals [163].

p53

The p53 tumor suppressor gene (TP53, Trp53) is a

pivotal gene controlling DNA repair, cell cycle,

apoptosis and cell senescence. Due to its extensive

involvement in biological phenomena so crucial for

cell life and death, it is conceivable that p53 gene can

be involved also in ageing and longevity. Accordingly,

it has been described that mice with increased p53

activity display an increased resistance to cancer, a

premature ageing phenotype and a reduced longevity

[164]. The complete deficiency of p53 leads to a

dramatic shortage of life expentancy due in this case

to increased cancer incidence in both mice [165] and

humans (Li-Fraumeni patients, [166]). Thus p53 has

to be considered a longevity-assurance gene in the

sense that it avoids premature death by protecting

from cancers in young age, and at the same time it is

a gene involved in ageing, since it appears that its

expression, while protecting from cancer, induces

ageing of the organism. On the basis of these results,

we looked at possible involvement of p53 gene

polymorphisms in ageing and longevity. In fact, it is

known that p53 gene harbours a number of

polymorphisms, the most studied of which is the

G-to-C transversion at codon 72 in the hexon 4. This

mutation leads to an aminoacidic change at position

72 in the protein from Arginine to Proline [167].

This mutation lies in the proline-rich domain and

confers a number of biochemical and biological

differences to the two resulting p53 isoforms [168]

and in particular as far as apoptosis, the Arg allele is

endowed with a greater apoptotic potential with

respect to Pro one [169–172]. By contrast, the Pro

allele appears to be much more efficient in cell cycle

arrest and cell senescence [173,174]. Interestingly,

these differences appeared more overt when cells

were obtained from aged subjects and centenarians

[173,174]. In the Caucasian population, the fre-

quency of the Arg allele is roughly 70%, while that of

the Pro allele is around 30% [175]. We wondered if

this polymorphism could impinge upon longevity,

and to test this hypothesis, we studied a total of 1086

Italian subjects of different ages (from young to

elderly people, including 307 centenarians). Besides

this G-to-C transversion, we also studied two

additional polymorphisms of p53 gene, that is the

16 bp Insertion/Deletion (Ins/Del) in Intron 3 and

Genes, ageing and longevity in humans 1311

the C to T transition in Intron 6, and found no

difference in the frequency distribution of these

polymorphisms in the age classes considered. We

concluded that p53 polymorphisms do not have an

effect on longevity [176–178]. In contrast, van

Heemst et al. reported that in a study of 1226 people

aged 85 years and over, Pro/Pro homozygous

subjects appear to have an increased survival despite

a 2.54 fold increased proportional mortality from

cancer [179]. The conclusion of this study was that

the survival advantage brought by Pro allele over-

whelms the increased risk of cancer. The discrepancy

with other data [176–178] nevertheless casts some

doubts on this general conclusion. The authors

themselves acknowledge that the study could be

underpowered and should be repeated in other

populations. In fact, Pro/Pro genotype is the more

rare and thus the number of subjects needed to have

a strong statistical power is likely not affordable

by a single study. Moreover, it is possible or even

likely that the effects observed by all these studies

[176–179] are population-specific. In our opinion

this is the reason why these results are apparently

in contrast.

On the basis of our preliminary results, we are

tempted to speculate that p53 codon 72 polymorph-

ism impinges upon not only cancer, but also other

common age-related diseases, and the algebraic sum

of the positive and negative effects on these different

pathologies gets to zero, at least on the Italian

population, so that in studies performed on Italian

subjects, the frequency distribution of p53 codon 72

genotypes does not change over age, but on the other

side it may vary in other (ethnically different)

populations in which the algebraic sum of these

effects is different. To conclude, p53 codon 72

polymorphism appears to impinge upon longevity,

and this effect might be population-specific, likely

depending on the interactions with both gene pools

and environmental conditions.

mtDNA in human longevity

Mitochondria are semi-autonomously functioning

organelles, harboring some life important cellular

process, such as the lipidic metabolism, the citric acid

cycle and the respiratory chain coupled to oxidative

phosphorylation (OXPHOS), that generates approxi-

mately 90% of cellular adenosine triphosphate (ATP)

[180]. Mitochondria are also provided with a small

genome, the mitochondrial DNA (mtDNA), that is

the only repository of genetic information outside the

nucleus. Human mtDNA is a 16,569 bp double-

stranded circular genome containing 37 genes encod-

ing 13 proteins (all of which are essential components

of the respiratory chain), 22 tRNAs and 2 rRNAs; it is

present in hundreds to thousands of copies per cell,

and transmitted as a non-recombinant unit only

through maternal inheritance [181,182]. The

mutation rate of mtDNA is about ten times higher

than nuclear genome since mtDNA is much more

exposed to ROS-induced damage than the nuclear

genome, because of the high vicinity to the oxidative

environment present in mitochondria [183]. More-

over, mtDNA has no histone-like protection, and less

efficient DNA repair systems, thus causing mtDNA to

accumulate various mutations in mitotic [184] and

post-mitotic tissues [185,186]. Nonetheless, it is

emerging that mtDNA is packaged into protein-

DNA complexes, called mitochondrial nucleoids (mt

nucleoids) that look like globular foci in human cells

[187]. Among the proteins discovered to interact with

mtDNA in mt nucleoids, some have known functions

related to mtDNA transaction, while others have not.

In mammals, among the proteins present in mtDNA

nucleoids there are factors involved in mtDNA

replication and transcription, such as TFAM, Twin-

kle, mtSSB, polymerase g, BRCA1, PRSS15 (LON),

and others involved in TCA cycle, such as aconitase

(for a review, see [188] and [189]). A possible role in

the protection of mtDNA from ROS damage can be

hypothesised for such proteins. The accumulation of

mutations in mtDNA, together with strong evidence

of a decline in the OXPHOS capacity of mitochondria

occurring with age in a variety of tissues such as

skeletal muscles, heart, brain, liver and other organs

[190,191] has given support to the “mitochondrial

theory of ageing” [192], an extension of the original

oxygen free radical theory of ageing proposed by

Harman [1]. It poses that accumulation of mutations

in mtDNA and consequent mitochondrial dysfunc-

tion are the major contributors to ageing and age-

related neurodegenerative diseases. In 2002, Lin and

his group found that mtDNA obtained from brain of

elderly subjects had a higher aggregate volume of

mutations than that obtained from brain of younger

subjects [193]. However, according to some authors,

specific mtDNA point mutations responsible for the

defects seen in ageing and age-related diseases have

yet to be identified [194].

Recently, somatic mutations in the control region

(CR) of the mtDNA have been associated with ageing

[195]. The A189G and T408A CR mutations

accumulate with age in skeletal muscle [196], while

a C150T mutation is more represented in white blood

cells and dermal fibroblasts of centenarians with

respect to young and old people in the Italian

population [197]. In any case, the correlation between

accumulation of somatic mutations, ageing and

longevity is still debated [198]. Recently, a murine

model has been set up in order to understand whether

the accumulation of mtDNA mutations is a cause or

rather an effect of ageing. In such a model, animals

expressing a proofreading-deficient version of mtDNA

polymerase g accumulate mtDNA mutations and

display features of accelerated ageing [199,200].

S. Salvioli et al.1312

Surprisingly, no increased oxidative stress occurs in

such mtDNA mutator mice [201]. On the basis of

these results, it can be assumed that the capability to

maintain mtDNA integrity and to avoid the accumu-

lation of mtDNA mutations is likely a feature of

longevity. However, this model does not really mimic

natural ageing, but it is rather to be considered as a

sort of genetic “disease”, since the mutator mice

accumulate mutations mainly in rapidly renewing

tissues rather than in post-mitotic ones, as it could be

expected, and the authors suggest that the resulting

phenotype could be due to the exhaustion of stem

cells, a phenomenon still not proven in physiological

ageing, where instead “old” microenvironment plays a

critical role for the function of cells (for a discussion

see Santoro et al. [198]).

Interestingly, recent findings by Sato et al. [202]

propose that mitochondrial complementation

phenomena do exist. They suggest that the mitochon-

dria can share mtDNA molecules and their products

as well, in order to avoid the expression of pathogenic

mutated forms of mtDNA. It is supposed that free

mixing of mitochondrial genetic components through-

out the mitochondrial network would protect mam-

malian mitochondria from direct expression of

respiratory defects caused by accumulated mtDNA

mutations. This innovative finding, if confirmed,

would challenge the mitochondrial theory of ageing

and lead to a re-consideration of the role of

mitochondria in ageing and longevity.

Beside somatic mutations, mtDNA is characterized

also by an inherited or inter-individual sequence

variability. Studies on mtDNA variations in human

populations have identified peculiar mutations that

were assumed to be neutral, thus avoiding elimination

by selection and becoming prevalent through genetic

drift. Such an assumption explains how mutations

which occurred thousands of years ago are nowadays

present at high frequency, and create groups of related

mtDNA haplotypes (called haplogroups), sharing a

specific set of stable polymorphic restriction sites

[203–205].

However, recent findings regarding the population-

and continent-specific distribution of such hap-

logroups, have suggested that such variants may not

be as neutral as previously supposed, but rather

influence and modulate mitochondrial metabolism, so

that factors such as climate and food or oxygen

availability can select them [205,206]. If this

hypothesis would come true, it is conceivable that

inherited mtDNA variability can impinge upon a

variety of phenotypes, including ageing, longevity and

age-related diseases. Accordingly, the analysis of

mtDNA haplogroups is currently providing new

insights into the association of mtDNA-inherited

variants in several neurodegenerative diseases such as

Parkinson’s and Alzheimer’s diseases [207–209].

As far as ageing is regarded, it has been observed

that in male centenarians from northern Italy mtDNA

haplogroup J is over-represented [210], suggesting a

protective role for this mtDNA variant against ageing.

However, when a large population from southern Italy

was studied, no association was found between

haplogroups J and longevity. On the whole, these

results once again support the idea that the effect of

mtDNA inherited variants on longevity is population-

specific and strongly depends on the interactions with

the environment.

This observation has been confirmed in two other

European studies [211,212] and in Japanese cente-

narians, where a sublineage of haplogroup D is more

frequent [213,214]. Furthermore, it has been found

that cytochrome b gene in mtDNA from centenarians

is characterised by inherited variants different from

those found in Parkinson’s patients [215]. Thus, the

available data support the hypothesis that mtDNA

lineages are qualitatively different from each other,

bearing mutations able to modify mitochondrial

functions, and consequentely affecting the probability

to reach longevity and avoid pathologies. To this

regard, it has been reported that mitochondria bearing

mtDNA belonging to haplogroups H and T displayed

a significant difference in the activity of complex I and

IV of respiratory chain in sperm cells [206]. We have

recently obtained data in vitro on a cybrid cell lines

model suggesting that mtDNA haplogroups can affect

the expression of cytokine and cytokine receptor

genes, thus suggesting that there is a cross-talk

between mitochondrial and nuclear genome, and

that mtDNA haplogroups modulate differently the

expression of nuclear genes [216].

Proteasome and immunoproteasome

Ageing is characterised by an accumulation of

oxidised proteins which are normally degraded by

the proteasome. Beside the increased oxidative stress,

this accumulation could be due to an impaired

function of the 20S proteasome in aged cells. To test

the importance of such phenomenon for human

ageing and longevity, we have analysed the expression

and the proteolytic activity of the proteasome in cells

from centenarians. Analysis of several proteasome

subunits RNA expression levels, determination of one

peptidase activity and identification of oxidised

proteins revealed that cells from centenarians have a

preserved proteasome function [217]. In addition, it

was found that cells from centenarians exhibit

characteristics similar to cells from younger rather

than the older control donors in all three assays. We

then moved to the study of possible involvement in

ageing and longevity of genetic variants of immuno-

proteasome subunits. Immunoproteasome is indeed a

crucial complex for the processing of antigens and

MHC-I presentation. We then studied polymorphisms

in LMP-2, LMP-7 and MECL-1 genes, whose

Genes, ageing and longevity in humans 1313

expression is needed for immunoproteasome assem-

bly. A non conservative nucleotide base pair change at

amino acid position 60 (in exon 3) in LMP2 gene,

resulting in two alleles, Arginine (R) or Histidine (H),

resulted to be associated with different autoimmune

diseases, but not with ageing [218,219]. However, it is

worthy to note that a modulation of proteasome

activity depending on the LMP2 R60H polymorphism

was observed in tissues derived from elderly people

(brain and blood) [219,220]. In particular, the

modulation of TNF-a induced-apoptosis in periph-

eral blood nuclear cells (likely mediated by IkB-a

degradation by proteasomes) was detectable only in

elderly subjects, being not present in samples from

young donors. Such results suggest that the effect of

the polymorphism is tissue-dependent and age-

dependent. In a recent study, we suggested that

LMP2 aminoacid 60 lies in a putative binding groove

for a regulatory molecule, able to enhance the 20S

proteasome activity [220]. On the whole, data

suggested how the activity/expression of such

hypothetical protein should be strictly regulated and

could vary on the basis of different factors, such as the

tissue analysed and the age of the subject. Further-

more, the putative regulatory site has been predicted

into the immunoproteasome, and no data about the

proteasome constitutive isoform are available. Hence,

we are tempted to speculate that the increase of

immunoproteasome content during ageing in specific

tissues such as human brain [221], could increase the

modulatory capability of the putative/predicted regu-

latory molecule, and in turn the importance of the

LMP2 codon 60 polymorphism, on the whole

proteasome activity. However, in cells like fibroblasts,

not directly involved in antigen-processing, it has been

recently reported that the levels of immunoprotea-

some subunits do not decrease but their induction by

IFNg is lost in senescent cells [222].

Conclusions

An impressive and coherent series of epidemiological

data in different populations (New England Amer-

icans, Mormons, Ashkenazi Jewish, Islandic, Okina-

wan Japanese, Italians, Irish, Dutch, among others)

indicate the presence of a strong familial component

of longevity. These studies demonstrate that parents,

siblings and offspring of long-lived subjects (but not

the spouses of the long-lived subjects who shared with

them most part of their adult life) have a significant

survival advantage, a higher probability to have been

or to became long-living persons and to have a lower

risk to undergo the most important age-related

diseases, such as cardio- and cerebro-vascular dis-

eases, diabetes and cancer, when compared to the

appropriate controls [223–229]. Thus longevity is

present in many generations of the same families in

spite of the great variations in life style and life

expectancy of the last century, and this strongly

suggests that this familiar trait is largely determined by

genetics. On the other side, many experimental

models have demonstrated that the modification of

even only one gene can deeply alter animal ageing or

longevity. In recent years, a great number of studies

have been performed in order to identify this genetic

component of human longevity, in particular by

seeking genes whose variants were correlated to

increased (or decreased) life-span. Different

approaches have been used, such as association

studies, case-control studies, epidemiological studies,

longitudinal studies on cohorts of old people, in the

attempt to find genes or gene variants that correlate

with longevity (or, at variance, with ageing or age-

associated diseases) in humans. Nevertheless, studies

on humans suffer by a variety of inescapable pitfalls

that render the results from these studies more

“confused” than those from strictly controlled animal

models in which variables such as life-style, environ-

mental conditions, gene pools, etc. are minimized.

Paradoxically this is also the strength of studies in

humans which do not suffer from the intrinsic

limitation consequent to the highly “artificial”

conditions in which animals used in biogerontological

studies to assess the determinants of longevity are

exposed life-long. The most clear example to this

regard is represented by studies on the ageing of the

immune system and the role played by immuno-

senescence in the onset of age-related pathologies.

Studies in humans and particularly on centenarians

clearly anticipated a variety of key observations such as

the progressive establishment of a peculiar chronic

inflammatory status we proposed to call inflamm-

aging, the accumulation of memory cells with age, the

exhaustion of virgin T cells and the role of viruses such

as cytomegalovirus (CMV), among other [230–238].

Such observations would be hampered or highly

biased by the “clean” conditions typical of well

controlled animal houses, quite distant from the real

“wild” conditions where every type of animal is

exposed life-long to a variety of pathogens. These were

indeed the conditions that modelled our genome and

that of other animals during evolution. We surmise

that similar bias may occur in the genetics of longevity,

when studies in humans and other animal models are

compared or extrapolated. Moreover, humans are

clearly different and more complex with respect to all

other animals either biologically or culturally. In

particular, all factors related to environment and

lifestyle are incomparably different from any labora-

tory environment and the effect of culture cannot be

underestimated. For these reasons, the results of these

studies are often “disturbing” in the sense that they are

difficult to compare each other, or even contrast.

Indeed, only in few cases the results have been

replicated in different populations (as in the case for

example of APOE4). On the contrary, as discussed

S. Salvioli et al.1314

all along this review, many associations between gene

variants and longevity have been found only in specific

populations. This should be not unexpected, since

ageing and longevity are complex traits resulting not

only and not exclusively from genetics, but rather from

the interactions between genetics, environment and

chance. In this perspective, it is conceivable that

longevity can be achieved by different combinations of

these three components, and thus, when gene pool

and environment change, the combinations leading to

longevity change as well [239].

In conclusion, the main lesson from these studies is

that is very difficult to identify “longevity genes” in

humans with “classical” association studies, because

wild type species (as humans are) are endowed with

such an intrinsic genetic complexity and environmen-

tal conditions can be so different from Country to

Country that render longevity-assurance combi-

nations, and thus “longevity genes”, almost exclu-

sively peculiar to any specific population. This should

not imply a diminished interest in regard to genes

whose effects on longevity are evident only in one or

few populations, but rather suggests that we urgently

need new approaches to move up one step on our

general understanding of the complex traits such as

human longevity. These could be, for example, studies

on very large population samples containing people of

different ethnic origin, so that internal comparisons

can be made avoiding methodological biases, or

studies on families (siblings or offspring of long-living

people, for example). Furthermore, high-throughput

analyses of many different polymorphisms must be

envisaged in order to obtain more informative data

about a candidate gene. Second, new computational

approaches have to be developed in order to interpret

the data. Third, in silico bioinformatics models

should be considered as a possible useful tool to

further implement benchtop data.

Finally, it has to be taken into account that longevity

is a post-reproductive phenomenon, so that longevity

as such has not been subjected to evolutionary

selection, and thus the genetics of longevity likely

does not conform to the classical genetic rules [239].

This aspect, together with other phenomena such as

antagonistic pleitropy, further complicates the study

of this peculiar aspect of human genetics. The

researches in this field are still in their infancy, and

the studies performed until now should be considered

little more than pioneering works that paved the way

to the discovery of this exciting aspect of science and

human life as well.

Acknowledgements

This work was supported by: EU Projects “T-CIA”

FP5 Contract n QLK6-CT-2002-02283 and

“GEHA—Genetics of Healthy Aging” FP6-503270

Grants; the PRRIITT program of the Emilia-Romagna

Region (and Fondi Strutturali Obiettivo 2); MIUR

(Italian Ministry of University) Fondo per gli

Investimenti della Ricerca di Base (FIRB) 2001,

protocol RBNE018AAP and #RBNE018R89; Italian

Ministry of Health Grant (Ricerche Finalizzate 2002

and 2003); FISM (Federazione Italiana Sclerosi

Multipla) Grant (Finalised Project “Immunoprotea-

some in Multiple Sclerosis: Genetics and Biological

Role in the Pathogenesis of the Disease”) to CF; Italian

Ministry of Health (Ricerca Finalizzata “Markers

genetici di sindrome coronaria acuta e valutazione

della L-arginina nella prevenzione di eventi ischemici”)

Grant to DM and CF; EU Project “ZINCAGE” 6th

FP, Contract n. FOOD-CT-2003-506850, and MIUR

(PRIN 2004) Grant to DM; University of Bologna

Ricerca Fondamentale Orientata (RFO 2005), and

Roberto and Cornelia Pallotti Legacy for Cancer

Research Grants to CF and SS.

References

[1] Harman D. Aging: A theory based on free radical and

radiation chemistry. J Gerontol 1956;11:298–300.

[2] Ridker PM. Inflammation, atherosclerosis, and cardiovascu-

lar risk: An epidemiologic view. Blood Coagul Fibrinolysis

1999;(Suppl 1):S9–S12.

[3] Chait A, Han CY, Oram JF, Heinecke JW. Thematic review

series: The immune system and atherogenesis. Lipoprotein-

associated inflammatory proteins: Markers or mediators of

cardiovascular disease? J Lipid Res 2005;46:389–403.

[4] Ross R. The pathogenesis of atherosclerosis—an update. N

Engl J Med 1986;314:488–500.

[5] Jaouad L, Milochevitch C, Khalil A. PON1 paraoxonase

activity is reduced during HDL oxidation and is an indicator

of HDL antioxidant capacity. Free Radic Res 2003; 37:

77–83.

[6] La Du BN, Aviram M, Billecke S, Navab M, Primo-Parmo S,

Sorenson RC, Standiford TJ. On the physiological role(s) of

the paraoxonases. Chem Biol Interact 1999;119–120:

379–388.

[7] Watson AD, Berliner JA, Hama SY, La Du BN, Faull KF,

Fogelman AM, Navab M. Protective effect of high density

lipoprotein associated paraoxonase. Inhibition of the biologi-

cal activity of minimally oxidized low density lipoprotein.

J Clin Invest 1995;96:2882–2891.

[8] Shih DM, Gu L, Xia YR, Navab M, Li WF, Hama S,

Castellani LW, Furlong CE, Costa LG, Fogelman AM, Lusis

AJ. Mice lacking serum paraoxonase are susceptible to

organophosphate toxicity and atherosclerosis. Nature

1998;394:284–287.

[9] Shih DM, Xia YR, Wang XP, Miller E, Castellani LW,

Subbanagounder G, Cheroutre H, Faull KF, Berliner JA,

Witztum JL, Lusis AJ. Combined serum paraoxonase

knockout/apolipoprotein E knockout mice exhibit increased

lipoprotein oxidation and atherosclerosis. J Biol Chem 2000;

275:17527–17535.

[10] Tward A, Xia YR, Wang XP, Shi YS, Park C, Castellani LW,

Lusis AJ, Shih DM. Decreased atherosclerotic lesion

formation in human serum paraoxonase transgenic mice.

Circulation 2002;106:484–490.

[11] Bonafe M, Marchegiani F, Cardelli M, Olivieri F, Cavallone

L, Giovagnetti S, Pieri C, Marra M, Antonicelli R, Troiano L,

Gueresi P, Passeri G, Berardelli M, Paolisso G, Barbieri M,

Tesei S, Lisa R, De Benedictis G, Franceschi C. Genetic

analysis of paraoxonase (PON1) locus reveals an increased

Genes, ageing and longevity in humans 1315

frequency of Arg192 allele in centenarians. Eur J Hum Genet

2002;10:292–296.

[12] Rea IM, McKeown PP, McMaster D, Young IS, Patterson C,

Savage MJ, Belton C, Marchegiani F, Olivieri F, Bonafe M,

Franceschi C. Paraoxonase polymorphisms PON1 192 and

55 and longevity in Italian centenarians and Irish nonagenar-

ians. A pooled analysis. Exp Gerontol 2004;39:629–635.

[13] Franceschi C, Olivieri F, Marchegiani F, Cardelli M,

Cavallone L, Capri M, Salvioli S, Valensin S, De Benedictis

G, Di Iorio A, Caruso C, Paolisso G, Monti D. Genes

involved in immune response/inflammation, IGF1/insulin

pathway and response to oxidative stress play a major role in

the genetics of human longevity: The lesson of centenarians.

Mech Ageing Dev 2005;126:351–361.

[14] Marchegiani F, Marra M, Spazzafumo L, James RW, Boemi

M, Olivieri F, Cardelli M, Cavallone L, Bonfigli AR,

Franceschi C. Paraoxonase activity and genotype predispose

to successful aging. J Gerontol A Biol Sci Med Sci

2006;61:541–546.

[15] Clendenning JB, Humbert R, Green ED, Wood C, Traver D,

Furlong CE. Structural organization of the human PON1

gene. Genomics 1996;35:586–589.

[16] Humbert R, Adler DA, Disteche CM, Hassett C, Omiecinski

CJ, Furlong CE. The molecular basis of the human serum

paraoxonase activity polymorphism. Nat Genet 1993;3:

73–76.

[17] Adkins S, Gan KN, Mody M, La Du BN. Molecular basis for

the polymorphic forms of human serum paraoxonase/aryles-

terase: Glutamine or arginine at position 191, for the

respective A or B allozymes. Am J Hum Genet 1993;52:

598–608.

[18] Garin MC, James RW, Dussoix P, Blanche H, Passa P,

Froguel P, Ruiz J. Paraoxonase polymorphism Met-Leu54 is

associated with modified serum concentrations of the

enzyme. A possible link between the paraoxonase gene and

increased risk of cardiovascular disease in diabetes. J Clin

Invest 1997;99:62–66.

[19] Marra M, Marchegiani F, Antonicelli R, Sirolla C,

Spazzafumo L, Olivieri F, Franceschi C, Testa R, Paolisso

G, James RW, Boemi M, Parati G. The PON1192 RR

genotype is associated with a higher prevalence of arterial

hypertension. J Hypertens 2006;24(7):1293–1298.

[20] Heijmans BT, Westendorp RG, Lagaay AM, Knook DL,

Kluft C, Slagboom PE. Common paraoxonase gene variants,

mortality risk and fatal cardiovascular events in elderly

subjects. Atherosclerosis 2000;149:91–97.

[21] Christiansen L, Bathum L, Frederiksen H, Christensen K.

Paraoxonase 1 polymorphisms and survival. Eur J Hum

Genet 2004;12:843–847.

[22] Tan Q, Christiansen L, Bathum L, Li S, Kruse TA,

Christensen K. Genetic association analysis of human

longevity in cohort studies of elderly subjects: An example

of the PON1 gene in the Danish 1905 birth cohort. Genetics

2006;172:1821–1828.

[23] Mackness M, Mackness B. Paraoxonase 1 and atherosclero-

sis: Is the gene or the protein more important? Free Radic

Biol Med 2004;37:1317–1323.

[24] Seres I, Paragh G, Deschene E, Fulop T, Jr., Khalil A. Study

of factors influencing the decreased HDL associated PON1

activity with aging. Exp Gerontol 2004;39:59–66.

[25] Khalil A, Wagner JR, Lacombe G, Dangoisse V, Fulop T, Jr.

Increased susceptibility of low-density lipoprotein (LDL) to

oxidation by gamma-radiolysis with age. FEBS Lett 1996;

392:45–48.

[26] Schachter F, Faure-Delanef L, Guenot F, Rouger H, Froguel

P, Lesueur-Ginot L, Cohen D. Genetic associations with

human longevity at the APOE and ACE loci. Nat Genet

1994;6:29–32.

[27] Carrieri G, Bonafe M, De Luca M, Rose G, Varcasia O,

Bruni A, Maletta R, Nacmias B, Sorbi S, Corsonello F,

Feraco E, Andreev KF, Yashin AI, Franceschi C, De

Benedictis G. Mitochondrial DNA haplogroups and

APOE4 allele are non-independent variables in sporadic

Alzheimer’s disease. Hum Genet 2001;108:194–198.

[28] Ordovas JM, Mooser V. Genes, lipids and aging: Is it all

accounted for by cardiovascular disease risk? Editorial review.

Curr Opin Lipidol 2005;16:121–126.

[29] De Benedictis G, Falcone E, Rose G, Ruffolo R, Spadafora P,

Baggio G, Bertolini S, Mari D, Mattace R, Monti D,

Morellini M, Sansoni P, Franceschi C. DNA multiallelic

systems reveal gene/longevity associations not detected by

diallelic systems. The APOB locus. Hum Genet 1997;99:

312–318.

[30] De Benedictis G, Carotenuto L, Carrieri G, De Luca M,

Falcone E, Rose G, Yashin AI, Bonafe M, Franceschi C. Age-

related changes of the 30APOB-VNTR genotype pool in

ageing cohorts. Ann Hum Genet 1998;62:115–122.

[31] Varcasia O, Garasto S, Rizza T, Andersen-Ranberg K, Jeune

B, Bathum L, Andreev K, Tan Q, Yashin AI, Bonafe M,

Franceschi C, De Benedictis G. Replication studies in

longevity: Puzzling findings in Danish centenarians at the

30APOB-VNTR locus. Ann Hum Genet 2001;65:371–376.

[32] Garasto S, Berardelli M, DeRango F, Mari V, Feraco E, De

Benedictis G. A study of the average effect of the 30APOB-

VNTR polymorphism on lipidemic parameters could explain

why the short alleles (,35 repeats) are rare in centenarians.

BMC Med Genet 2004;5:3.

[33] Garasto S, Rose G, Derango F, Berardelli M, Corsonello A,

Feraco E, Mari V, Maletta R, Bruni A, Franceschi C,

Carotenuto L, De Benedictis G. The study of APOA1,

APOC3 and APOA4 variability in healthy ageing people

reveals another paradox in the oldest old subjects. Ann Hum

Genet 2003;67:54–62.

[34] Barzilai N, Atzmon G, Schechter C, Schaefer EJ, Cupples

AL, Lipton R, Cheng S, Shuldiner AR. Unique lipoprotein

phenotype and genotype associated with exceptional long-

evity. JAMA 2003;290:2030–2040.

[35] Cellini E, Nacmias B, Olivieri F, Ortenzi L, Tedde A, Bagnoli

S, Petruzzi C, Franceschi C, Sorbi S. Cholesterol ester

transfer protein (CETP) I405V polymorphism and longevity

in Italian centenarians. Mech Ageing Dev 2005;126:

826–828.

[36] Blaschuk O, Burdzy K, Fritz I. Purification and character-

ization of a cell-aggregating factor (Clusterin), the major

glycoprotein in ram rete testis fluid. J Biol Chem 1983;

258:7714–7720.

[37] Jenne DE, Tschopp J. Molecular structure and functional

characterization of a human complement cytolysis found in

blood and seminal plasma: Identity to sulfated glycoprotein 2,

a constituent of rat testis fluid. Proc Natl Acad Sci

1989;86:7123–7127.

[38] Kirszbaum L, Sharpe JA, Murphy B, d’Apice AJF, Classon B,

Hudson P, Walker ID. Molecular cloning and characteriz-

ation of the novel, human complement-associated protein,

SP-40,40: A link between the complement and reproductive

systems. EMBO J 1989;8:711–718.

[39] de Silva HV, Harmony JAK, Stuart WD, Gil CM, Robbins J.

Apolipoprotein J: Structure and tissue distribution. Bio-

chemistry 1990;29:5380–5389.

[40] Tycko B, Feng L, Nguyen L, Francis A, Hays A, Chung WY,

Tang MX, Stern Y, Sahota A, Hendrie H, Mayeux R.

Polymorphisms in the human apolipoprotein-J/clusterin

gene: Ethnic variation and distribution in Alzheimer’s

disease. Hum Genet 1996;98:430–436.

[41] Trougakos IP, Petropoulou C, Franceschi C, Gonos ES.

Reduced expression levels of the senescence biomarker

S. Salvioli et al.1316

Clusterin/Apolipoprotein J in lymphocytes from healthy

centenarians. Ann N Y Acad Sci 2006;1067:294–300.

[42] Trougakos IP, Gonos ES. Clusterin/Apolipoprotein J in

human aging and cancer. Int J Biochem Cell Biol 2002;

34:1430–1448.

[43] Gonos ES, Burns JS, Mazars GR, Kobrna A, Riley TEW,

Barnet SC, Zafarana G, Ludwig R, Ikram Z, Powell AJ, Jat

PS. Rat embryo fibroblasts immortalised with SV40 large T

antigen undergo senescence upon its inactivation. Mol Cell

Biol 1996;16:5127–5138.

[44] Powell AJ, Darmon AJ, Gonos ES, Lam EWF, Peden KWC,

Jat PS. Different functions are required for initiation and

maintenance of immortalisation of rat embryo fibroblasts by

SV40 large T antigen. Oncogene 1999;18:7343–7350.

[45] Gonos ES, Derventzi A, Kveiborg M, Agiostratidou G,

Kassem M, Clark BFC, Jat PS, Rattan SIS. Cloning and

identification of genes that associate with mammalian

senescence. Exp Cell Res 1998;240:66–74.

[46] Dumont P, Burton M, Chen QM, Gonos ES, Frippiat C,

Mazarati JB, Eliaers F, Remacle J, Toussaint O. Induction of

replicative senescence biomarkers by sublethal successive

oxidative stresses in normal human fibroblasts. Free Radic

Biol Med 2000;28:361–373.

[47] Petropoulou C, Trougakos IP, Kolettas E, Toussaint O,

Gonos ES. Clusterin/apolipoprotein J is a novel biomarker of

cellular senescence, that does not affect the proliferative

capacity of human diploid fibroblasts. FEBS Lett 2001; 509:

287–297.

[48] Trougakos IP, Poulakou M, Stathatos M, Chalikia A,

Melidonis A, Gonos ES. Serum levels of the senescence

biomarker Clusterin/Apolipoprotein J increase significantly in

diabetes type II and during development of coronary heart

disease or at myocardial infarction. Exp Gerontol 2002; 37:

1175–1187.

[49] Trougakos IP, Lourda M, Agiostratidou G, Kletsas D, Gonos

ES. Differential effects of Clusterin/Apolipoprotein J on

cellular growth and survival. Free Radic Biol Med 2005; 38:

436–449.

[50] Trougakos IP, So A, Jansen B, Gleave ME, Gonos ES.

Silencing expression of the Clusterin/Apolipoprotein J gene in

human cancer cells using small interfering RNA induces

spontaneous apoptosis, reduced growth ability and cell

sensitization to genotoxic and oxidative stress. Cancer Res

2004;64:1834–1842.

[51] Puca AA, Daly MJ, Brewster SJ, Matise TC, Barrett J, Shea-

Drinkwater M, Kang S, Joyce E, Nicoli J, Benson E, Kunkel

LM, Perls T. A genome-wide scan for linkage to human

exceptional longevity identifies a locus on chromosome 4.

Proc Natl Acad Sci USA 2001;98:10505–10508.

[52] Geesaman BJ, Benson E, Brewster SJ, Kunkel LM, Blanche

H, Thomas G, Perls TT, Daly MJ, Puca AA. Haplotype-

based identification of a microsomal transfer protein marker

associated with the human lifespan. Proc Natl Acad Sci USA

2003;100:14115–141120.

[53] Beekman M, Blauw GJ, Houwing-Duistermaat JJ, Brandt

BW, Westendorp RG, Slagboom PE. Chromosome 4q25,

microsomal transfer protein gene, and human longevity:

Novel data and a meta-analysis of association studies.

J Gerontol A Biol Sci Med Sci 2006;61:355–362.

[54] Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura

T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP,

Giardino I, Brownlee M. Normalizing mitochondrial super-

oxide production blocks three pathways of hyperglycaemic

damage. Nature 2000;404:787–790.

[55] Schmidt AM, Hori O, Brett J, Yan SD, Wautier JL, Stern D.

Cellular receptors for advanced glycation end products.

Implications for induction of oxidant stress and cellular

dysfunction in the pathogenesis of vascular lesions. Arter-

ioscler Thromb 1994;14:1521–1528.

[56] Kirstein M, Brett J, Radoff S, Ogawa S, Stern D, Vlassara H.

Advanced protein glycosylation induces transendothelial

human monocyte chemotaxis and secretion of platelet-

derived growth factor: Role in vascular disease of diabetes and

aging. Proc Natl Acad Sci USA 1990;87:9010–9014.

[57] Vlassara H, Brownlee M, Manogue KR, Dinarello CA,

Pasagian A. Cachectin/TNF and IL-1 induced by glucose-

modified proteins: Role in normal tissue remodeling. Science

1988;240:1546–1548.

[58] Kirstein M, Aston C, Hintz R, Vlassara H. Receptor-specific

induction of insulin-like growth factor I in human monocytes

by advanced glycosylation end product-modified proteins. J

Clin Invest 1992;90:439–446.

[59] Ross R. Atherosclerosis—an inflammatory disease. N Engl J

Med 1999;340:115–126.

[60] Paolisso G, Ammendola S, Del Buono A, Gambardella A,

Riondino M, Tagliamonte MR, Rizzo MR, Carella C,

Varricchio M. Serum levels of insulin-like growth factor-I

(IGF-I) and IGF-binding protein-3 in healthy centenarians:

Relationship with plasma leptin and lipid concentrations,

insulin action, and cognitive function. J Clin Endocrinol

Metab 1997;82:2204–2209.

[61] Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E,

Giampieri C, Mugianesi E, Centurelli M, Franceschi C,

Paolisso G. Polymorphic variants of insulin-like growth factor

I (IGF-I) receptor and phosphoinositide 3-kinase genes affect

IGF-I plasma levels and human longevity: Cues for an

evolutionarily conserved mechanism of life span control. J

Clin Endocrinol Metab 2003;88:3299–3304.

[62] Paolisso G, Gambardella A, Ammendola S, D’Amore A,

Balbi V, Varricchio M, D’Onofrio F. Glucose tolerance and

insulin action in healthy centenarians. Am J Physiol 1996;

270:E890–E896.

[63] Paolisso G, Barbieri M, Rizzo MR, Carella C, Rotondi M,

Bonafe M, Franceschi C, Rose G, De Benedictis G. Low

insulin resistance and preserved b-cell function contribute to

human longevity but are not associated with TH-INS genes.

Exp Gerontol 2001;37:149–156.

[64] Spallarossa P, Brunelli C, Minuto F, Caruso D, Battistini M,

Caponnetto S, Cordera R. Insulin-like growth factor-1 and

angiographically documented coronary artery disease. Am J

Cardiol 1996;77:200–202.

[65] van den Beld AW, Bots ML, Janssen JA, Pols HA, Lamberts

SW, Grobbee DE. Endogenous hormones and carotid

atherosclerosis in elderly men. Am J Epidemiol 2003; 157:

25–31.

[66] Juul A, Scheike T, Davidsen M, Gyllenborg J, Jorgensen T.

Low serum insulin-like growth factor-1 is associated with

increased risk of ischemic heart disease: A population-based

case-control study. Circulation 2002;106:939–944.

[67] Laughlin GA, Barrett-Connor E, Criqui MH, Kritz-

Silverstein D. The prospective association of serum insulin-

like growth factor-1 (IGF-1) and IGF-binding protein-1

levels with all cause and cardiovascular disease mortality in

older adults: The Rancho Bernardo Study. J Clin Endocrinol

Metab 2004;89:114–120.

[68] Barbieri M, Ferrucci L, Ragno E, Corsi A, Bandinelli S,

Bonafe M, Olivieri F, Giovagnetti S, Franceschi C, Guralnik

JM, Paolisso G. Chronic inflammation and the effect of IGF-I

on muscle strength and power in older persons. Am J Physiol

Endocrinol Metab 2003;284:E481–E487.

[69] Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M,

Ottaviani E, De Benedictis G. Inflamm-aging. An evolution-

ary perspective on immunosenescence. Ann N Y Acad Sci

2000;908:244–254.

[70] Vanfleteren JR, Braeckman BP. Mechanisms of life span

determination in Caenorhabditis elegans. Neurobiol Aging

1999;20:487–502.

Genes, ageing and longevity in humans 1317

[71] Clancy DJ, Gems D, Harshman LG, Oldham S, Stocker H,

Hafen E, Leevers SJ, Partridge L. Extension of life-span by

loss of CHICO, a Drosophila insulin receptor substrate

protein. Science 2001;292:104–106.

[72] Gems D, Partridge L. Insulin/IGF signalling and ageing:

Seeing the bigger picture. Curr Opin Genet Dev 2001; 11:

287–292.

[73] Bartke A, Chandrashekar V, Dominici F, Turyn D, Kinney B,

Steger R, Kopchick JJ. Insulin-like growth factor 1 (IGF-1)

and aging: Controversies and new insights. Biogerontology

2003;4:1–8.

[74] Tatar M, Bartke A, Antebi A. The endocrine regulation of

aging by insulin-like signals. Science 2003;299:1346–1351.

[75] Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A,

Even PC, Cervera P, Le Bouc Y. IGF-1 receptor regulates

lifespan and resistance to oxidative stress in mice. Nature

2003;421:182–187.

[76] Kenyon C. A conserved regulatory system for aging. Cell

2001;105:165–168.

[77] Hsieh CC, DeFord JH, Flurkey K, Harrison DE, Papacon-

stantinou J. Effects of the Pit1 mutation on the insulin

signaling pathway: Implications on the longevity of the long-

lived Snell dwarf mouse. Mech Ageing Dev 2002; 123:

1245–1255.

[78] Dozmorov I, Bartke A, Miller RA. Array-based expression

analysis of mouse liver genes: Effect of age and of the

longevity mutant Prop1df. J Gerontol A Biol Sci Med Sci

2001;56:B72–B80.

[79] Steger RW, Bartke A, Cecim M. Premature ageing in

transgenic mice expressing different growth hormone genes.

J Reprod Fertil Suppl 1993;46:61–75.

[80] Sonntag WE, Lenham JE, Ingram RL. Effects of aging and

dietary restriction on tissue protein synthesis: Relationship to

plasma insulin-like growth factor-1. J Gerontol 1992; 47:

B159–B163.

[81] Sonntag WE, Lynch CD, Cefalu WT, Ingram RL, Bennett

SA, Thornton PL, Khan AS. Pleiotropic effects of growth

hormone and insulin-like growth factor (IGF)-1 on biological

aging: Inferences from moderate caloric-restricted animals.

J Gerontol A Biol Sci Med Sci 1999;54:B521–B538.

[82] Kojima T, Kamei H, Aizu T, Arai Y, Takayama M, Nakazawa

S, Ebihara Y, Inagaki H, Masui Y, Gondo Y, Sakaki Y, Hirose

N. Association analysis between longevity in the Japanese

population and polymorphic variants of genes involved in

insulin and insulin-like growth factor 1 signaling pathways.

Exp Gerontol 2004;39:1595–1598.

[83] van Heemst D, Beekman M, Mooijaart SP, Heijmans BT,

Brandt BW, Zwaan BJ, Slagboom PE, Westendorp RG.

Reduced insulin/IGF-1 signalling and human longevity.

Aging Cell 2005;4:79–85.

[84] Cardelli M, Marchegiani F, Cavallone L, Olivieri F,

Giovanetti S, Mugianesi E, Moresi R, Lisa R, Franceschi

C. A Polymorphism of the YTHDF2 gene (1p35) located in

an Alu-rich genomic domain is associated with human

longevity. J Gerontol A Biol Sci Med Sci 2006; in press.

[85] GenBank [Internet]. Bethesda (MD), National Center for

Biotechnology Information (US), National Library of

Medicine, National Institutes of Health. Available from:

http://www.ncbi.nlm.nih.gov/Genbank.

[86] Belcher JD, Mahaseth H, Welch TE, Otterbein LE, Hebbel

RP, Vercellotti GM. Heme oxygenase-1 is a modulator of

inflammation and vaso-occlusion in transgenic sickle mice.

J Clin Invest 2006;116:808–816.

[87] Haddad JJ. Science review: Redox and oxygen-sensitive

transcription factors in the regulation of oxidant-mediated

lung injury: Role for hypoxia-inducible factor-1a. Crit Care

2003;7:47–54.

[88] Arsura M, Panta GR, Bilyeu JD, Cavin LG, Sovak MA,

Oliver AA, Factor V, Heuchel R, Mercurio F, Thorgeirsson

SS, Sonenshein GE. Transient activation of NF-kappaB

through a TAK1/IKK kinase pathway by TGF-beta1 inhibits

AP-1/SMAD signaling and apoptosis: Implications in liver

tumor formation. Oncogene 2003;22:412–425.

[89] Lee HB, Yu MR, Yang Y, Jiang Z, Ha H. Reactive oxygen

species-regulated signaling pathways in diabetic nephropathy.

J Am Soc Nephrol 2003;14:S241–S245.

[90] Gomez-Cambronero LG, Sabater L, Pereda J, Cassinello N,

Camps B, Vina J, Sastre J. Role of cytokines and oxidative

stress in the pathophysiology of acute pancreatitis: Thera-

peutical implications. Curr Drug Targets Inflamm Allergy

2002;1:393–403.

[91] Taylor ER, Hurrell F, Shannon RJ, Lin TK, Hirst J, Murphy

MP. Reversible glutathionylation of complex I increases

mitochondrial superoxide formation. J Biol Chem 2003; 278:

19603–19610.

[92] Tieri P, Valensin S, Latora V, Castellani GC, Marchiori M,

Remondini D, Franceschi C. Quantifying the relevance of

different mediators in the human immune cell network.

Bioinformatics 2005;21:1639–1643.

[93] Cavallone L, Bonafe M, Olivieri F, Cardelli M, Marchegiani

F, Giovagnetti S, Di Stasio G, Giampieri C, Mugianesi E,

Stecconi R, Sciacca F, Grimaldi LM, De Benedictis G, Lio D,

Caruso C, Franceschi C. The role of IL-1 gene cluster in

longevity: A study in Italian population. Mech Ageing Dev

2003;124:533–538.

[94] Bonafe M, Olivieri F, Cavallone L, Giovagnetti S, Mayegiani

F, Cardelli M, Pieri C, Marra M, Antonicelli R, Lisa R, Rizzo

MR, Paolisso G, Monti D, Franceschi C. A gender-

dependent genetic predisposition to produce high levels of

IL-6 is detrimental for longevity. Eur J Immunol 2001; 31:

2357–2361.

[95] Wright N, de Lera TL, Garcia-Moruja C, Lillo R, Garcia-

Sanchez F, Caruz A, Teixido J. Transforming growth factor-

beta1 down-regulates expression of chemokine stromal

cell-derived factor-1: Functional consequences in cell

migration and adhesion. Blood 2003;102:1978–1984.

[96] My-Chan Dang P, Elbim C, Marie JC, Chiandotto M,

Gougerot-Pocidalo MA, El-Benna J. Anti-inflammatory

effect of interleukin-10 on human neutrophil respiratory

burst involves inhibition of GM-CSF-induced p47PHOX

phosphorylation through a decrease in ERK1/2 activity.

FASEB J 2006 May 23.

[97] Csiszar A, Ungvari Z, Koller A, Edwards JG, Kaley G. Aging-

induced proinflammatory shift in cytokine expression profile

in coronary arteries. FASEB J 2003;17:1183–1185.

[98] Paolisso G, Tagliamonte MR, Rizzo MR, Manzella D,

Gambardella A, Varricchio M. Oxidative stress and advan-

cing age: Results in healthy centenarians. J Am Geriatr Soc

1998;46:833–838.

[99] Barbieri M, Rizzo MR, Manzella D, Grella R, Ragno E,

Carbonella M, Abbatecola AM, Paolisso G. Glucose

regulation and oxidative stress in healthy centenarians.

Exp Gerontol 2003;38:137–143.

[100] Maccio A, Madeddu C, Massa D, Mudu MC, Lusso MR,

Gramignano G, Serpe R, Melis GB, Mantovani G.

Hemoglobin levels correlate with interleukin-6 levels in

patients with advanced untreated epithelial ovarian cancer:

Role of inflammation in cancer-related anemia. Blood 2005;

106:362–367.

[101] Summers WK. Alzheimer’s disease, oxidative injury, and

cytokines. J Alzheimers Dis 2004;6:651–657.

[102] Walston J, Xue Q, Semba RD, Ferrucci L, Cappola AR, Ricks

M, Guralnik J, Fried LP. Serum antioxidants, inflammation,

and total mortality in older women. Am J Epidemiol 2006;

163:18–26.

[103] Ferrucci L, Harris TB, Guralnik JM, Tracy RP, Corti MC,

Cohen HJ, Penninx B, Pahor M, Wallace R, Havlik RJ.

S. Salvioli et al.1318

Serum IL-6 level and the development of disability in older

persons. J Am Geriatr Soc 1999;47:639–646.

[104] Harris TB, Ferrucci L, Tracy RP, Corti MC, Wacholder S,

Ettinger WH, Jr., Heimovitz H, Cohen HJ, Wallace R.

Associations of elevated interleukin-6 and C-reactive protein

levels with mortality in the elderly. Am J Med 1999; 106:

506–512.

[105] Hurme M, Lehtimaki T, Jylha M, Karhunen PJ, Hervonen A.

Interleukin-6-174G/C polymorphism and longevity: A

follow-up study. Mech Ageing Dev 2005;126:417–418.

[106] Christiansen L, Bathum L, Andersen-Ranberg K, Jeune B,

Christensen K. Modest implication of interleukin-6 promoter

polymorphisms in longevity. Mech Ageing Dev 2004; 125:

391–395.

[107] Pes GM, Lio D, Carru C, Deiana L, Baggio G, Franceschi C,

Ferrucci L, Oliveri F, Scola L, Crivello A, Candore G,

Colonna-Romano G, Caruso C. Association between long-

evity and cytokine gene polymorphisms. A study in Sardinian

centenarians. Aging Clin Exp Res 2004;16:244–248.

[108] Capurso C, Solfrizzi V, D’Introno A, Colacicco AM,

Capurso SA, Semeraro C, Capurso A, Panza F. Interleukin

6-174 G/C promoter gene polymorphism in centenarians: No

evidence of association with human longevity or interaction

with apolipoprotein E alleles. Exp Gerontol 2004;39:

1109–1114.

[109] Carrieri G, Marzi E, Olivieri F, Marchegiani F, Cavallone L,

Cardelli M, Giovagnetti S, Stecconi R, Molendini C,

Trapassi C, De Benedictis G, Kletsas D, Franceschi C. The

G/C915 polymorphism of transforming growth factor beta1

is associated with human longevity: A study in Italian

centenarians. Aging Cell 2004;3:443–448.

[110] Okayama N, Hamanaka Y, Suehiro Y, Hasui Y, Nakamura J,

Hinoda Y. Association of interleukin-10 promoter single

nucleotide polymorphisms -819 T/C and -592 A/C with

aging. J Gerontol A Biol Sci Med Sci 2005;60:1525–1529.

[111] Lio D, Scola L, Crivello A, Colonna-Romano G, Candore G,

Bonafe M, Cavallone L, Franceschi C, Caruso C. Gender-

specific association between -1082 IL-10 promoter poly-

morphism and longevity. Genes Immun 2002;3:30–33.

[112] Lio D, Scola L, Crivello A, Colonna-Romano G, Candore G,

Bonafe M, Cavallone L, Marchegiani F, Olivieri F,

Franceschi C, Caruso C. Genetics, and longevity: Further

studies on the protective effects in men of IL-10 -1082

promoter SNP and its interaction with TNF-alpha -308

promoter SNP. J Med Genet 2003;40:296–299.

[113] Lio D, Candore G, Crivello A, Scola L, Colonna-Romano G,

Cavallone L, Hoffmann E, Caruso M, Licastro F, Caldarera

CM, Branzi A, Franceschi C, Caruso C. Opposite effects of

interleukin 10 common gene polymorphisms in cardiovas-

cular diseases and in successful ageing: Genetic background

of male centenarians is protective against coronary heart

disease. J Med Genet 2004;41:790–794.

[114] Scola L, Crivello A, Marino V, Gioia V, Serauto A, Candore

G, Colonna-Romano G, Caruso C, Lio D. IL-10 and TNF-

alpha polymorphisms in a sample of Sicilian patients affected

by tuberculosis: Implication for ageing and life span

expectancy. Mech Ageing Dev 2003;124:569–572.

[115] Candore G, Lio D, Colonna Romano G, Caruso C.

Pathogenesis of autoimmune diseases associated with 8.1

ancestral haplotype: Effect of multiple gene interactions.

Autoimmun Rev 2002;1:29–35.

[116] de Maat MP, Bladbjerg EM, Hjelmborg JB, Bathum L,

Jespersen J, Christensen K. Genetic influence on inflam-

mation variables in the elderly. Arterioscler Thromb Vasc Biol

2004;24:2168–2173.

[117] Giacconi R, Cipriano C, Albanese F, Boccoli G, Saba V,

Olivieri F, Franceschi C, Mocchegiani E. The -174G/C

polymorphism of IL-6 is useful to screen old subjects at risk

for atherosclerosis or to reach successful ageing. Exp Gerontol

2004;39:621–628.

[118] Antonicelli R, Olivieri F, Cavallone L, Spazzafumo L, Bonafe

M, Marchegiani F, Cardelli M, Galeazzi R, Giovagnetti S,

Perna GP, Franceschi C. Tumor necrosis factor-alpha gene

2308G . A polymorphism is associated with ST-elevation

myocardial infarction and with high plasma levels of

biochemical ischemia markers. Coron Artery Dis 2005; 16:

489–493.

[119] Antonicelli R, Olivieri F, Bonafe M, Cavallone L, Spazza-

fumo L, Marchegiani F, Cardelli M, Recanatini A,

Testarmata P, Boemi M, Parati G, Franceschi C. The

interleukin-6 -174 G . C promoter polymorphism is

associated with a higher risk of death after an acute coronary

syndrome in male elderly patients. Int J Cardiol 2005; 103:

266–271.

[120] Olivieri F, Antonicelli R, Cardelli M, Marchegiani F,

Cavallone L, Mocchegiani E, Franceschi C. Genetic

polymorphisms of inflammatory cytokines and myocardial

infarction in the elderly. Mech Ageing Dev 2006;127:

552–559.

[121] Testa R, Olivieri F, Bonfigli AR, Sirolla C, Boemi M,

Marchegiani F, Marra M, Cenerelli S, Antonicelli R, Dolci A,

Paolisso G, Franceschi C. Interleukin-6-174 G . C poly-

morphism affects the association between IL-6 plasma levels

and insulin resistance in type 2 diabetic patients. Diabetes

Res Clin Pract 2006;71:299–305.

[122] Belluco C, Olivieri F, Bonafe M, Giovagnetti S, Mammano

E, Scalerta R, Ambrosi A, Franceschi C, Nitti D, Lise M. -

174G . polymorphism of interleukin 6 gene promoter

affects interleukin 6 serum level in patients with colorectal

cancer. Clin Cancer Res 2003;9:2173–2176.

[123] Licastro F, Grimaldi LM, Bonafe M, Martina C, Olivieri F,

Cavallone L, Giovanietti S, Masliah E, Franceschi C.

Interleukin-6 gene alleles affect the risk of Alzheimer’s

disease and levels of the cytokine in blood and brain.

Neurobiol Aging 2003;24:921–926.

[124] Franceschi C, Valensin S, Lescai F, Olivieri F, Licastro F,

Grimaldi LM, Monti D, De Benedictis G, Bonafe M.

Neuroinflammation and the genetics of Alzheimer’s disease:

The search for a pro-inflammatory phenotype. Aging

(Milano) 2001;13:163–170.

[125] Seripa D, Matera MG, Dal Forno G, Gravina C, Masullo C,

Daniele A, Binetti G, Bonvicini C, Squitti R, Palermo MT,

Davis DG, Antuono P, Wekstein DR, Dobrina A, Gennarelli

M, Fazio VM. Genotypes and haplotypes in the IL-1 gene

cluster: Analysis of two genetically and diagnostically distinct

groups of Alzheimer patients. Neurobiol Aging 2005;26:

455–464.

[126] Barbieri M, Rizzo MR, Papa M, Acampora R, De Angelis L,

Olivieri F, Marchegiani F, Franceschi C, Paolisso G. Role of

interaction between variants in the PPARG and interleukin-6

genes on obesity related metabolic risk factors. Exp Gerontol

2005;40:599–604.

[127] Salvioli S, Capri M, Valensin S, Tieri P, Monti D, Ottaviani

E, Franceschi C. Inflamm-aging, cytokines and aging: State

of the art, new hypotheses on the role of mitochondria and

new perspectives from system biology. Curr Pharm Des

2006; in press.

[128] Short R, Williams DD, Bowden DM. Circulating antiox-

idants as determinants of the rate of biological aging in

pigtailed macaques (Macaca nemestrina). J Gerontol A Biol

Sci Med Sci 1997;52:B26–B38.

[129] Mezzetti A, Lapenna D, Romano F, Costantini F,

Pierdomenico SD, De Cesare D, Cuccurullo F, Riario-

Sforza G, Zuliani G, Fellin R. Systemic oxidative stress and

its relationship with age and illness. Associazione Medica

“Sabin”. J Am Geriatr Soc 1996;44:823–827.

Genes, ageing and longevity in humans 1319

[130] Landis GN, Tower J. Superoxide dismutase evolution and life

span regulation. Mech Ageing Dev 2005;126:365–379.

[131] Weisiger RA, Fridovich I. Mitochondrial superoxide simu-

tase. Site of synthesis and intramitochondrial localization. J

Biol Chem 1973;248:4793–4796.

[132] McCord JM, Fridovich I. Superoxide dismutase. An enzymic

function for erythrocuprein (hemocuprein). J Biol Chem

1969;244:6049–6055.

[133] Marklund SL. Human copper-containing superoxide dis-

mutase of high molecular weight. Proc Natl Acad Sci USA

1982;79:7634–7638.

[134] Spencer CC, Howell CE, Wright AR, Promislow DE. Testing

an ‘aging gene’ in long-lived drosophila strains: Increased

longevity depends on sex and genetic background. Aging Cell

2003;2:123–130.

[135] Sun J, Molitor J, Tower J. Effects of simultaneous over-

expression of Cu/ZnSOD and MnSOD on Drosophila

melanogaster life span. Mech Ageing Dev 2004;125:341–349.

[136] Duttaroy A, Parkes T, Emtage P, Kirby K, Boulianne GL,

Wang X, Hilliker AJ, Phillips JP. The manganese superoxide

dismutase gene of Drosophila: Structure, expression, and

evidence for regulation by MAP kinase. DNA Cell Biol 1997;

16:391–399.

[137] Phillips JP, Campbell SD, Michaud D, Charbonneau M,

Hilliker AJ. Null mutation of copper/zinc superoxide

dismutase in Drosophila confers hypersensitivity to paraquat

and reduced longevity. Proc Natl Acad Sci USA 1989; 86:

2761–2765.

[138] Hinerfeld D, Traini MD, Weinberger RP, Cochran B,

Doctrow SR, Harry J, Melov S. Endogenous mitochondrial

oxidative stress: Neurodegeneration, proteomic analysis,

specific respiratory chain defects, and efficacious antioxidant

therapy in superoxide dismutase 2 null mice. J Neurochem

2004;88:657–667.

[139] Andersen HR, Jeune B, Nybo H, Nielsen JB, Andersen-

Ranberg K, Grandjean P. Low activity of superoxide

dismutase and high activity of glutathione reductase in

erythrocytes from centenarians. Age Ageing 1998; 27:

643–648.

[140] Mecocci P, Polidori MC, Troiano L, Cherubini A, Cecchetti

R, Pini G, Straatman M, Monti D, Stahl W, Sies H,

Franceschi C, Senin U. Free Radic Biol Med 2000; 28:

1243–1248.

[141] Mutus B, Rabini RA, Franceschi C, Paolisso G, Rizzo MR,

Ragno E, Rappelli A, Braconi M, Mazzanti L. Cellular

resistance to homocysteine: A key for longevity. Athero-

sclerosis 2000;152:527–528.

[142] Rabini RA, Moretti N, Staffolani R, Salvolini E, Nanetti L,

Franceschi C, Mazzanti L. Reduced susceptibility to

peroxidation of erythrocyte plasma membranes from cen-

tenarians. Exp Gerontol 2002;37:657–663.

[143] Rabini RA, Vignini A, Martarelli D, Nanetti L, Salvolini E,

Rizzo MR, Ragno E, Paolisso G, Franceschi C, Mazzanti L.

Evidence for reduction of pro-atherosclerotic properties in

platelets from healthy centenarians. Exp Gerontol

2003;38:367–371.

[144] Mazzanti L, Rabini RA, Petruzzi E, Staffolani R, Salvolini E,

Vignini A, Braconi M, Franceschi C. Erythrocyte plasma

membranes obtained from centenarians show different

functional properties. J Am Geriatr Soc 2000;48:350–351.

[145] Hayes JD, Flanagan JU, Jowsey IR. Glutathione transferases.

Annu Rev Pharmacol Toxicol 2005;45:51–88.

[146] Taioli E, Mari D, Franceschi C, Bonafe M, Monti D,

Bertolini S, Marinelli D, Garte S. Polymorphisms of drug-

metabolizing enzymes in healthy nonagenarians and cen-

tenarians: Difference at GSTT1 locus. Biochem Biophys Res

Commun 2001;280:1389–1392.

[147] Geisler SA, Olshan AF, Cai J, Weissler M, Smith J, Bell D.

Glutathione S-transferase polymorphisms and survival from

head and neck cancer. Head Neck 2005;27:232–242.

[148] Christiansen L, Brasch-Andersen C, Bathum L, Kruse TA,

Christensen K. A longitudinal study of the effect of GSTT1

and GSTM1 gene copy number on survival. Mech Ageing

Dev 2006;127:597–599.

[149] Burkle A. Physiology and pathophysiology of poly(ADP-

ribosyl)ation. Bioessays 2001;23:795–806.

[150] Burkle A, Beneke S, Muiras ML. Poly(ADP-ribosyl)ation

and aging. Exp Gerontol 2004;39:1599–1601.

[151] Lindahl T, Satoh MS, Poirier GG, Klungland A. Post-

translational modification of poly(ADP-ribose) polymerase

induced by DNA strand breaks. Trends Biochem Sci

1995;20:405–411.

[152] D’Amours D, Desnoyers S, D’Silva I, Poirier GG.

Poly(ADP-ribosyl)ation reactions in the regulation of nuclear

functions. Biochem J 1999;342:249–268.

[153] de Murcia JM, Niedergang C, Trucco C, Ricoul M,

Dutrillaux B, Mark M, Oliver FJ, Masson M, Dierich A,

LeMeur M, Walztinger C, Chambon P, de Murcia G.

Requirement of poly(ADP-ribose) polymerase in recovery

from DNA damage in mice and in cells. Proc Natl Acad Sci

USA 1997;94:7303–7307.

[154] Grube K, Burkle A. Poly(ADP-ribose) polymerase activity in

mononuclear leukocytes of 13 mammalian species correlates

with species-specific life span. Proc Natl Acad Sci USA

1992;89:11759–11763.

[155] Beneke S, Alvarez-Gonzalez R, Burkle A. Comparative

characterisation of poly(ADP-ribose) polymerase-1 from two

mammalian species with different life span. Exp Gerontol

2000;35:989–1002.

[156] Muiras ML, Muller M, Schachter F, Burkle A. Increased

poly(ADP-ribose) polymerase activity in lymphoblastoid cell

lines from centenarians. J Mol Med 1998;76:346–354.

[157] Burkle A. PARP-1: A regulator of genomic stability linked

with mammalian longevity. Chembiochem 2001;2:725–728.

[158] Skaper SD. Poly(ADP-Ribose) polymerase-1 in acute

neuronal death and inflammation: A strategy for neuropro-

tection. Ann N Y Acad Sci 2003;993:217–228.

[159] Mabley JG, Soriano FG. Role of nitrosative stress and

poly(ADP-ribose) polymerase activation in diabetic vascular

dysfunction. Curr Vasc Pharmacol 2005;3:247–252.

[160] Mocchegiani E, Muzzioli M, Giacconi R, Cipriano C,

Gasparini N, Franceschi C, Gaetti R, Cavalieri E, Suzuki H.

Metallothioneins/PARP1/IL-6 interplay on natural killer cell

activity in elderly: Parallelism with nonagenarians and old

infected humans. Effect of zinc supply. Mech Ageing Dev

2003;124:459–468.

[161] De Benedictis G, Carotenuto L, Carrieri G, De Luca M,

Falcone E, Rose G, Cavalcanti S, Corsonello F, Feraco E,

Baggio G, Bertolini S, Mari D, Mattace R, Yashin AI, Bonafe

M, Franceschi C. Gene/longevity association studies at four

autosomal loci (REN, THO, PARP, SOD2). Eur J Hum

Genet 1998;6:534–541.

[162] Cottet F, Blanche H, Verasdonck P, Le Gall I, Schachter F,

Burkle A, Muiras ML. New polymorphisms in the human

poly(ADP-ribose) polymerase-1 coding sequence: Lack of

association with longevity or with increased cellular poly

(ADP-ribosyl)ation capacity. J Mol Med 2000;78:431–440.

[163] Burkle A, Diefenbach J, Brabeck C, Beneke S. Ageing ARP.

Pharmacol Res 2005;52:93–99.

[164] Tyner SD, Venkatachalam S, Choi J, Jones S, Ghebranious

N, Igelmann H, Lu X, Soron G, Cooper B, Brayton C, Hee

Park S, Thompson T, Karsenty G, Bradley A, Donehower

LA. p53 mutant mice that display early ageing-associated

phenotypes. Nature 2002;415:45–53.

[165] Donehower LA, Harvey M, Slagle BL, McArthur MJ,

Montgomery CA, Jr., Butel JS, Bradley A. Mice deficient for

S. Salvioli et al.1320

p53 are developmentally normal but susceptible to spon-

taneous tumours. Nature 1992;356:215–221.

[166] Malkin D, Li FP, Strong LC, Fraumeni JF, Jr., Nelson CE,

Kim DH, Kassel J, Gryka MA, Bischoff FZ, Tainsky MA,

Friend SH. Germ line p53 mutations in a familial syndrome

of breast cancer, sarcomas, and other neoplasms. Science

1990;250:1233–1238.

[167] Matlashewski GJ, Tuck S, Pim D, Lamb P, Schneider J,

Crawford LV. Primary structure polymorphism at amino acid

residue 72 of human p53. Mol Cell Biol 1987;7:961–963.

[168] Thomas M, Kalita A, Labrecque S, Pim D, Banks L,

Matlashewski G. Two polymorphic variants of wild-type p53

differ biochemically and biologically. Mol Cell Biol

1999;19:1092–1100.

[169] Bonafe M, Salvioli S, Barbi C, Mishto M, Trapassi C,

Gemelli C, Storci G, Olivieri F, Monti D, Franceschi C. p53

codon 72 genotype affects apoptosis by cytosine arabinoside

in blood leukocytes. Biochem Biophys Res Commun

2002;299:539–541.

[170] Dumont P, Leu JI, Della Pietra 3rd AC, George DL, Murphy

M. The codon 72 polymorphic variants of p53 have markedly

different apoptotic potential. Nat Genet 2003;33:357–365.

[171] Sullivan A, Syed N, Gasco M, Bergamaschi D, Trigiante G,

Attard M, Hiller L, Farrell PJ, Smith P, Lu X, Crook T.

Polymorphism in wild-type p53 modulates response to

chemotherapy in vitro and in vivo. Oncogene 2004;23:

3328–3337.

[172] Bonafe M, Salvioli S, Barbi C, Trapassi C, Tocco F, Storci G,

Invidia L, Vannini I, Rossi M, Marzi E, Mishto M, Capri M,

Olivieri F, Antonicelli R, Memo M, Uberti D, Nacmias B,

Sorbi S, Monti D, Franceschi C. The different apoptotic

potential of the p53 codon 72 alleles increases with age and

modulates in vivo ischaemia-induced cell death. Cell Death

Differ 2004;11:962–973.

[173] Pim D, Banks L. p53 polymorphic variants at codon 72 exert

different effects on cell cycle progression. Int J Cancer 2004;

108:196–199.

[174] Salvioli S, Bonafe M, Barbi C, Storci G, Trapassi C, Tocco F,

Gravina S, Rossi M, Tiberi L, Mondello C, Monti D,

Franceschi C. p53 codon 72 alleles influence the response

to anticancer drugs in cells from aged people by regulating

the cell cycle inhibitor p21WAF1. Cell Cycle 2005;4:

1264–1271.

[175] Weston A, Godbold JH. Polymorphisms of H-ras-1 and p53

in breast cancer and lung cancer: A meta-analysis. Environ

Health Perspect 1997;105:919–926.

[176] Bonafe M, Olivieri F, Mari D, Baggio G, Mattace R,

Berardelli M, Sansoni P, De Benedictis G, De Luca M,

Marchegiani F, Cavallone L, Cardelli M, Giovagnetti S,

Ferrucci L, Amadio L, Lisa R, Tucci MG, Troiano L, Pini G,

Gueresi P, Morellini M, Sorbi S, Passeri G, Barbi C, Valensin

S, Monti D, Deiana L, Pes GM, Carru C, Franceschi C. P53

codon 72 polymorphism and longevity: Additional data on

centenarians from continental Italy and Sardinia. Am J Hum

Genet 1999;65:1782–1785.

[177] Bonafe M, Olivieri F, Mari D, Baggio G, Mattace R, Sansoni

P, De Benedictis G, De Luca M, Bertolini S, Barbi C, Monti

D, Franceschi C. p53 variants predisposing to cancer are

present in healthy centenarians. Am J Hum Genet 1999; 64:

292–295.

[178] Bonafe M, Barbi C, Storci G, Salvioli S, Capri M, Olivieri F,

Valensin S, Monti D, Gonos ES, De Benedictis G,

Franceschi C. What studies on human longevity tell us

about the risk for cancer in the oldest old: Data and

hypotheses on the genetics and immunology of centenarians.

Exp Gerontol 2002;37:1263–1271.

[179] van Heemst D, Mooijaart SP, Beekman M, Schreuder J, de

Craen AJ, Brandt BW, Slagboom PE, Westendorp RG. Long

life study group. Variation in the human TP53 gene affects

old age survival and cancer mortality. Exp Gerontol 2005; 40:

11–15.

[180] Wallace DC. Mitochondrial DNA in aging and disease. Sci

Am 1997;277:40–47.

[181] Barthelemy C, Ogier de Baulny H, Diaz J, Cheval MA,

Frachon P, Romero N, Goutieres F, Fardeau M, Lombes A.

Late-onset mitochondrial DNA depletion: DNA copy

number, multiple deletions, and compensation. Ann Neurol

2001;49:607–617.

[182] Miller FJ, Rosenfeldt FL, Zhang C, Linnane AW, Nagley P.

Precise determination of mitochondrial DNA copy number

in human skeletal and cardiac muscle by a PCR-based assay:

Lack of change of copy number with age. Nucleic Acids Res

2003;31:e61.

[183] Shigenaga MK, Hagen TM, Ames BN. Oxidative damage

and mitochondrial decay in aging. Proc Natl Acad Sci USA

1994;91:10771–11078.

[184] Michikawa Y, Laderman K, Richter K, Attardi G. Role of

nuclear background and in vivo environment in variable

segregation behavior of the aging-dependent T414G

mutation at critical control site for human fibroblast

mtDNA replication. Somat Cell Mol Genet 1999;25:

333–342.

[185] Soong NW, Hinton DR, Cortopassi G, Arnheim N.

Mosaicism for a specific somatic mitochondrial DNA

mutation in adult human brain. Nat Genet 1992;2:318–323.

[186] Liu VW, Zhang C, Nagley P. Mutations in mitochondrial

DNA accumulate differentially in three different human

tissues during ageing. Nucleic Acids Res 1998;26:

1268–1275.

[187] Spelbrink JN, Li FY, Tiranti V, Nikali K, Yuan QP, Tariq M,

Wanrooij S, Garrido N, Comi G, Morandi L, Santoro L,

Toscano A, Fabrizi GM, Somer H, Croxen R, Beeson D,

Poulton J, Suomalainen A, Jacobs HT, Zeviani M, Larsson C.

Human mitochondrial DNA deletions associated with

mutations in the gene encoding Twinkle, a phage T7 gene

4-like protein localized in mitochondria. Nat Genet 2001; 28:

223–231.

[188] Chen XJ, Butow RA. The organization and inheritance of the

mitochondrial genome. Nat Rev Genet 2005;6:815–825.

[189] Shadel GS. Mitochondrial DNA, aconitase ‘wraps’ it up.

Trends Biochem Sci 2005;30:294–296.

[190] Muller-Hocker J. Cytochrome-c-oxidase deficient cardio-

myocytes in the human heart—an age-related phenomenon.

A histochemical ultracytochemical study. Am J Pathol 1989;

134:1167–1173.

[191] Muller-Hocker J, Schneiderbanger K, Stefani FH, Kaden-

bach B. Progressive loss of cytochrome c oxidase in the

human extraocular muscles in ageing—a cytochemical-

immunohistochemical study. Mutat Res 1992;275:115–124.

[192] Kowald A. The mitochondrial theory of aging. Biol Signals

Recept 2001;10(3–4):162–175.

[193] Lin MT, Simon DK, Ahn CH, Kim LM, Beal MF. High

aggregate burden of somatic mtDNA point mutations in

aging and Alzheimer’s disease brain. Hum Mol Genet

2002;11:133–145.

[194] Chinnery PF, Taylor GA, Howell N, Brown DT, Parsons TJ,

Turnbull DM. Point mutations of the mtDNA control region

in normal and neurodegenerative human brains. Am J Hum

Genet 2001;68:529–532.

[195] Coskun PE, Ruiz-Pesini E, Wallace DC. Control region

mtDNA variants: Longevity, climatic adaptation, and a

forensic conundrum. Proc Natl Acad Sci USA 2003; 100:

2174–2176.

[196] Wang Y, Michikawa Y, Mallidis C, Bai Y, Woodhouse L,

Yarasheski KE, Miller CA, Askanas V, Engel WK, Bhasin S,

Attardi G. Muscle-specific mutations accumulate with aging

in critical human mtDNA control sites for replication. Proc

Natl Acad Sci USA 2001;98:4022–4027.

Genes, ageing and longevity in humans 1321

[197] Zhang J, Asin-Cayuela J, Fish J, Michikawa Y, Bonafe M,

Olivieri F, Passarino G, De Benedictis G, Franceschi C,

Attardi G. Strikingly higher frequency in centenarians and

twins of mtDNA mutation causing remodeling of replication

origin in leukocytes. Proc Natl Acad Sci USA 2003; 100:

1116–1121.

[198] Santoro A, Salvioli S, Raule N, Capri M, Sevini F, Monti D,

Bellizzi D, Passarino G, Rose G, De Benedictis G, Franceschi

C. MtDNA involvement in human longevity. BBA—

Bioenergetics 2006; in press.

[199] Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN,

Rovio AT, Bruder CE, Bohlooly-Y M, Gidlof S, Oldfors A,

Wibom R, Tornell J, Jacobs HT, Larsson NG. Premature

ageing in mice expressing defective mitochondrial DNA

polymerase. Nature 2004;429:417–423.

[200] Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K,

Wohlgemuth SE, Hofer T, Seo AY, Sullivan R, Jobling WA,

Morrow JD, Van Remmen H, Sedivy JM, Yamasoba T,

Tanokura M, Weindruch R, Leeuwenburgh C, Prolla TA.

Mitochondrial DNA mutations, oxidative stress, and

apoptosis in mammalian aging. Science 2005;309:481–484.

[201] Trifunovic A, Hansson A, Wredenberg A, Rovio AT, Dufour

E, Khvorostov I, Spelbrink JN, Wibom R, Jacobs HT,

Larsson NG. Somatic mtDNA mutations cause aging

phenotypes without affecting reactive oxygen species pro-

duction. Proc Natl Acad Sci USA 2005;102:17993–17998.

[202] Sato A, Nakada K, Hayashi J. Mitochondrial dynamics and

aging: Mitochondrial interaction preventing individuals from

expression of respiratory deficiency caused by mutant

mtDNA. BBA 2006; in press.

[203] Torroni A, Wallace DC. Mitochondrial DNA variation in

human populations and implications for detection of

mitochondrial DNA mutations of pathological significance.

J Bioenerg Biomembr 1994;26:261–271.

[204] Wallace DC. William Allan Award Address. Mitochondrial

DNA variation in human evolution, degenerative disease, and

aging. Am J Hum Genet 1995;57:201–223.

[205] Wallace DC. Mitochondrial DNA sequence variation in

human evolution and disease. Proc Natl Acad Sci USA 1994;

91:8739–8746.

[206] Ruiz-Pesini E, Mishmar D, Brandon M, Procaccio V, Wallace

DC. Effects of purifyng and adaptive selection on regional

variation in human mtDNA. Science 2004;303:223–225.

[207] van der Walt JM, Nicodemus KK, Martin ER, Scott WK,

Nance MA, Watts RL, Hubble JP, Haines JL, Koller WC,

Lyons K, Pahwa R, Stern MB, Colcher A, Hiner BC,

Jankovic J, Ondo WG, Allen FH, Jr., Goetz CG, Small GW,

Mastaglia F, Stajich JM, McLaurin AC, Middleton LT, Scott

BL, Schmechel DE, Pericak-Vance MA, Vance JM.

Mitochondrial polymorphisms significantly reduce the risk

of Parkinson disease. Am J Hum Genet 2003;72:804–811.

[208] van Der Walt JM, Dementieva G, Martin ER, Scott WK,

Nicodemus KK, Kroner CC, Welsh-Bohmer KA, Saunders

AM, Roses AD, Small GW, Schmechel DE, Doraiswamy

PM, Gilbert JR, Haines JL, Vance JM, Pericak-Vance MA.

Analysis of European mitochondrial haplogroups with

Alzheimer disease risk. Neurosci Lett 2004;365:28–32.

[209] Ghezzi D, Marelli C, Achilli A, Goldwurm S, Pezzoli G,

Barone P, Pellecchia MT, Stanzione P, Brusa L, Bentivoglio

AR, Bonuccelli U, Petrozzi L, Abbruzzese G, Marchese R,

Cortelli P, Grimaldi D, Martinelli P, Ferrarese C, Garavaglia

B, Sangiorgi S, Carelli V, Torroni A, Albanese A, Zeviani M.

Mitochondrial DNA haplogroup K is associated with a lower

risk of Parkinson’s disease in Italians. Eur J Hum Genet

2005;13:748–752.

[210] De Benedictis G, Rose G, Carrieri G, De Luca M, Falcone E,

Passarino G, Bonafe M, Monti D, Baggio G, Bertolini S,

Mari D, Mattace R, Franceschi C. Mitochondrial DNA

inherited variants are associated with successful aging and

longevity in humans. FASEB J 1999;13:1532–1536.

[211] Ross OA, McCormack R, Curran MD, Duguid RA, Barnett

YA, Rea IM, Middleton D. Mitochondrial DNA polymorph-

ism: Its role in longevity of the Irish population. Exp Gerontol

2001;36:1161–1178.

[212] Niemi AK, Hervonen A, Hurme M, Karhunen PJ, Jylha M,

Majamaa K. Mitochondrial DNA polymorphisms associated

with longevity in a Finnish population. Hum Genet 2003;

112:29–33.

[213] Tanaka M, Gong JS, Zhang J, Yoneda M, Yagi K.

Mitochondrial genotype associated with longevity. Lancet

1998;351:185–186.

[214] Tanaka M, Gong J, Zhang J, Yamada Y, Borgeld HJ, Yagi K.

Mitochondrial genotype associated with longevity and its

inhibitory effect on mutagenesis. Mech Ageing Dev

2000;116:65–76.

[215] Tanaka M, Fuku N, Takeyasu T, Guo LJ, Hirose R,

Kurata M, Borgeld HJ, Yamada Y, Maruyama W, Arai Y,

Hirose N, Oshida Y, Sato Y, Hattori N, Mizuno Y, Iwata

S, Yagi K. Golden mean to longevity: Rareness of

mitochondrial cytochrome b variants in centenarians but

not in patients with Parkinson’s disease. J Neurosci Res

2002;70:347–355.

[216] Bellizzi D, Cavalcante P, Taverna D, Rose G, Passarino G,

Salvioli S, Franceschi C, De Benedictis G. Gene Expression

of cytokines and cytokine-receptors is modulated by common

variability of the mitochondrial DNA in cybrid cell lines.

Genes Cells 2006; in press.

[217] Chondrogianni N, Petropoulos I, Franceschi C, Friguet B,

Gonos ES. Fibroblast cultures from healthy centenarians

have an active proteasome. Exp Gerontol 2000;35:

721–728.

[218] Mishto M, Bonafe M, Salvioli S, Olivieri F, Franceschi C.

Age dependent impact of LMP polymorphisms on TNF-

alpha-induced apoptosis in human peripheral blood mono-

nuclear cells. Exp Gerontol 2002;37:301–308.

[219] Mishto M, Santoro A, Bellavista E, Bonafe M, Monti D,

Franceschi C. Immunoproteasomes and immunosenescence.

Ageing Res Rev 2003;2:419–432.

[220] Mishto M, Santoro A, Bellavista E, Sessions R, Textoris-

Taube K, Dal Piaz F, Carrard G, Forti K, Salvioli S, Friguet

B, Kloetzel PM, Rivett AJ, Franceschi C. A structural model

of 20S immunoproteasomes: Effect of LMP2 codon 60

polymorphism on expression, activity, intracellular localis-

ation and insight into the regulatory mechanisms. Biol Chem

2006;387:417–429.

[221] Mishto M, Bellavista E, Santoro A, Stolzing A, Ligorio C,

Nacmias B, Spazzafumo L, Chiappelli M, Licastro F, Sorbi

S, Pession A, Ohm T, Grune T, Franceschi C. Immunopro-

teasome and LMP2 polymorphism in aged and Alzheimer’s

disease brains. Neurobiol Aging 2006;27:54–66.

[222] Stratford FL, Chondrogianni N, Trougakos IP, Gonos ES,

Rivett AJ. Proteasome response to interferon-gamma is

altered in senescent human fibroblasts. FEBS Lett 2006; 580:

3989–3994.

[223] Atzmon G, Schechter C, Greiner W, Davidson D, Rennert G,

Barzilai N. Clinical phenotype of families with longevity. J

Am Geriatr Soc 2004;52:274–277.

[224] Terry DF, Wilcox MA, McCormick MA, Pennington JY,

Schoenhofen EA, Andersen SL, Perls TT. Lower all-cause,

cardiovascular, and cancer mortality in centenarians’ off-

spring. J Am Geriatr Soc 2004;52:2074–2076.

[225] Terry DF, Wilcox M, McCormick MA, Lawler E, Perls TT.

Cardiovascular advantages among the offspring of centenar-

ians. J Gerontol A Biol Sci Med Sci 2003;58:M425–M431.

[226] Karasik D, Hannan MT, Cupples LA, Felson DT, Kiel DP.

Genetic contribution to biological aging: The Framingham

study. J Gerontol A Biol Sci Med Sci 2004;59:218–226.

S. Salvioli et al.1322

[227] Ikeda A, Iso H, Toyoshima H, Kondo T, Mizoue T,

Koizumi A, Inaba Y, Tamakoshi A. JACC study

group. Parental longevity and mortality amongst Japanese

men and women: The JACC Study. J Intern Med 2006; 259:

285–295.

[228] Perls TT, Wilmoth J, Levenson R, Drinkwater M, Cohen M,

Bogan H, Joyce E, Brewster S, Kunkel L, Puca A. Life-long

sustained mortality advantage of siblings of centenarians.

Proc Natl Acad Sci USA 2002;99:8442–8847.

[229] Schoenmaker M, de Craen AJ, de Meijer PH, Beekman M,

Blauw GJ, Slagboom PE, Westendorp RG. Evidence of

genetic enrichment for exceptional survival using a family

approach: The Leiden longevity study. Eur J Hum Genet

2006;14:79–84.

[230] Cossarizza A, Ortolani C, Paganelli R, Barbieri D, Monti D,

Sansoni P, Fagiolo U, Castellani G, Bersani F, Londei M,

Franceschi C. CD45 isoforms expression on CD4 þ and

CD8 þ T cells throughout life, from newborns to centenar-

ians: Implications for T cell memory. Mech Ageing Dev

1996;86:173–195.

[231] Franceschi C, Bonafe M, Valensin S. Human immunosenes-

cence: The prevailing of innate immunity, the failing of

clonotypic immunity, and the filling of immunological space.

Vaccine 2000;18:1717–1720.

[232] Fagnoni FF, Vescovini R, Passeri G, Bologna G, Pedrazzoni

M, Lavagetto G, Casti A, Franceschi C, Passeri M, Sansoni

P. Shortage of circulating naive CD8(þ) T cells provides new

insights on immunodeficiency in aging. Blood 2000;95:

2860–2868.

[233] Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca

M, Ottaviani E, De Benedictis G. Inflamm-aging. An

evolutionary perspective on immunosenescence. Ann N Y

Acad Sci 2000;908:244–254.

[234] Franceschi C, Valensin S, Bonafe M, Paolisso G, Yashin AI,

Monti D, De Benedictis G. The network and the remodeling

theories of aging: Historical background and new perspec-

tives. Exp Gerontol 2000;35:879–896.

[235] Vescovini R, Telera A, Fagnoni FF, Biasini C, Medici MC,

Valcavi P, di Pede P, Lucchini G, Zanlari L, Passeri G, Zanni

F, Chezzi C, Franceschi C, Sansoni P. Different contribution

of EBVand CMV infections in very long-term carriers to age-

related alterations of CD8þ T cells. Exp Gerontol 2004; 39:

1233–1243.

[236] Poulain M, Pes GM, Grasland C, Carru C, Ferrucci L,

Baggio G, Franceschi C, Deiana L. Identification of a

geographic area characterized by extreme longevity in the

Sardinia island: The AKEA study. Exp Gerontol 2004; 39:

1423–1429.

[237] Ginaldi L, De Martinis M, Monti D, Franceschi C. Chronic

antigenic load and apoptosis in immunosenescence. Trends

Immunol 2005;26:79–84.

[238] De Martinis M, Franceschi C, Monti D, Ginaldi L.

Inflamm-ageing and lifelong antigenic load as major

determinants of ageing rate and longevity. FEBS Lett

2005;579:2035–2039.

[239] De Benedictis G, Franceschi C. The unusual genetics of

human longevity. Sci Aging Knowledge Environ 2006;

2006:pe20.

Genes, ageing and longevity in humans 1323