148 research outputs found

    Durée de vie, génétique et axe somatotrope

    Get PDF
    Longtemps descriptive, la recherche sur le vieillissement a profondément changé depuis la découverte de gènes régulant la durée de vie. Isolés en criblant le génome de simples nématodes, la plupart de ces gènes appartiennent à une voie de signalisation hautement conservée au cours de l’évolution. Leurs orthologues chez les vertébrés sont les familles des gènes de l’insuline, de l’insulin-like growth factor (IGF) et de leurs voies de signalisation. Très étudiés et connus pour leurs rôles dans la prolifération, la différenciation, la survie cellulaire et le métabolisme intermédiaire, on découvre maintenant leurs multiples fonctions dans le contrôle de la longévité et dans les réponses au stress oxydant, une des causes majeures du vieillissement cellulaire. La signalisation IGF chez les mammifères dépend d’un ensemble de signaux endocriniens que constitue l’axe somatotrope. En effet, plusieurs composantes de cet axe hormonal régulent efficacement la longévité, ce qui a été élégamment démontré par une série de modèles de souris génétiquement modifiées. Il est de plus en plus évident que le contrôle du vieillissement met en jeu des régulations hormonales dont l’ampleur des implications commence à peine à être découverte.Research on ageing made a big leap forward when genes regulating lifespan were discovered about a decade ago. First isolated by screening the genome of the nematode Caenorhabditis elegans, most of these genes belong to an essential signalling pathway that is highly conserved during animal evolution. Orthologous genes in vertebrate species are the families of genes coding for insulin, insulin-like growth factors (IGF) and related proteins. Intensively studied and well-known for their pivotal roles in proliferation, differentiation, survival and metabolism of most cells, we now discover their multiples functions with respect to the control of longevity and their ability to modulate the cell’s responses to oxidative stress, a major cause of cellular and organismal ageing. The activity of IGF signalling in mammals depends on a complex interplay of endocrine signals that together constitute the somatotropic axis. Accordingly, several components of this hormone axis, like growth hormone or growth hormone releasing hormone receptors, regulate efficiently animal longevity, which has been elegantly demonstrated by studies performed in genetically modified mouse models. From this and other work, it becomes increasingly clear that the control of ageing is a question of hormonal regulations. We here present several of these models and discuss the respective contributions of insulin and IGF signalling to the regulation of lifespan. We review data on the Klotho gene that acts on lifespan via surprising and not yet fully understood molecular mechanisms, connecting this new, hormone-like substance to IGF and insulin signalling. We further report recent evidence showing that human lifespan might be controlled in similar ways. Finally, we shed some light on clinical GH treatment in humans, from an endocrinologist’s point of view

    IGF-1 receptor regulates upward firing rate homeostasis via the mitochondrial calcium uniporter

    Get PDF
    Regulation of firing rate homeostasis constitutes a fundamental property of central neural circuits. While intracellular Ca2+ has long been hypothesized to be a feedback control signal, the molecular machinery enabling a network-wide homeostatic response remains largely unknown. We show that deletion of insulin-like growth factor-1 receptor (IGF-1R) limits firing rate homeostasis in response to inactivity, without altering the distribution of baseline firing rates. The deficient firing rate homeostatic response was due to disruption of both postsynaptic and intrinsic plasticity. At the cellular level, we detected a fraction of IGF-1Rs in mitochondria, colocalized with the mitochondrial calcium uniporter complex (MCUc). IGF-1R deletion suppressed transcription of the MCUc members and burst-evoked mitochondrial Ca2+ (mitoCa(2+)) by weakening mitochondria-to-cytosol Ca2+ coupling. Overexpression of either mitochondria-targeted IGF-1R or MCUc in IGF-1R-deficient neurons was sufficient to rescue the deficits in burst-to-mitoCa(2+) coupling and firing rate homeostasis. Our findings indicate that mitochondrial IGF-1R is a key regulator of the integrated homeostatic response by tuning the reliability of burst transfer by MCUc. Based on these results, we propose that MCUc acts as a homeostatic Ca2+ sensor. Faulty activation of MCUc may drive dysregulation of firing rate homeostasis in aging and in brain disorders associated with aberrant IGF-1R/MCUc signaling

    Components of the Hematopoietic Compartments in Tumor Stroma and Tumor-Bearing Mice

    Get PDF
    Solid tumors are composed of cancerous cells and non-cancerous stroma. A better understanding of the tumor stroma could lead to new therapeutic applications. However, the exact compositions and functions of the tumor stroma are still largely unknown. Here, using a Lewis lung carcinoma implantation mouse model, we examined the hematopoietic compartments in tumor stroma and tumor-bearing mice. Different lineages of differentiated hematopoietic cells existed in tumor stroma with the percentage of myeloid cells increasing and the percentage of lymphoid and erythroid cells decreasing over time. Using bone marrow reconstitution analysis, we showed that the tumor stroma also contained functional hematopoietic stem cells. All hematopoietic cells in the tumor stroma originated from bone marrow. In the bone marrow and peripheral blood of tumor-bearing mice, myeloid populations increased and lymphoid and erythroid populations decreased and numbers of hematopoietic stem cells markedly increased with time. To investigate the function of hematopoietic cells in tumor stroma, we co-implanted various types of hematopoietic cells with cancer cells. We found that total hematopoietic cells in the tumor stroma promoted tumor development. Furthermore, the growth of the primary implanted Lewis lung carcinomas and their metastasis were significantly decreased in mice reconstituted with IGF type I receptor-deficient hematopoietic stem cells, indicating that IGF signaling in the hematopoietic tumor stroma supports tumor outgrowth. These results reveal that hematopoietic cells in the tumor stroma regulate tumor development and that tumor progression significantly alters the host hematopoietic compartment

    Delayed and Accelerated Aging Share Common Longevity Assurance Mechanisms

    Get PDF
    Mutant dwarf and calorie-restricted mice benefit from healthy aging and unusually long lifespan. In contrast, mouse models for DNA repair-deficient progeroid syndromes age and die prematurely. To identify mechanisms that regulate mammalian longevity, we quantified the parallels between the genome-wide liver expression profiles of mice with those two extremes of lifespan. Contrary to expectation, we find significant, genome-wide expression associations between the progeroid and long-lived mice. Subsequent analysis of significantly over-represented biological processes revealed suppression of the endocrine and energy pathways with increased stress responses in both delayed and premature aging. To test the relevance of these processes in natural aging, we compared the transcriptomes of liver, lung, kidney, and spleen over the entire murine adult lifespan and subsequently confirmed these findings on an independent aging cohort. The majority of genes showed similar expression changes in all four organs, indicating a systemic transcriptional response with aging. This systemic response included the same biological processes that are triggered in progeroid and long-lived mice. However, on a genome-wide scale, transcriptomes of naturally aged mice showed a strong association to progeroid but not to long-lived mice. Thus, endocrine and metabolic changes are indicative of “survival” responses to genotoxic stress or starvation, whereas genome-wide associations in gene expression with natural aging are indicative of biological age, which may thus delineate pro- and anti-aging effects of treatments aimed at health-span extension

    Long-Term IGF-I Exposure Decreases Autophagy and Cell Viability

    Get PDF
    A reduction in IGF-I signaling has been found to increase lifespan in multiple organisms despite the fact that IGF-I is a trophic factor for many cell types and has been found to have protective effects against multiple forms of damage in acute settings. The increase in longevity seen in response to reduced IGF-I signaling suggests that there may be differences between the acute and chronic impact of IGF-I signaling. We have examined the possibility that long-term stimulation with IGF-I may have a negative impact at the cellular level using quiescent human fibroblasts. We find that fibroblast cells exposed to IGF-I for 14 days have reduced long-term viability as judged by colony forming assays, which is accompanied by an accumulation of senescent cells. In addition we observe an accumulation of cells with depolarized mitochondria and a reduction in autophagy in the long-term IGF-I treated cultures. An examination of mice with reduced IGF-I levels reveals evidence of enhanced autophagy and fibroblast cells derived from these mice have a larger mitochondrial mass relative to controls indicating that changes in mitochondrial turnover occurs in animals with reduced IGF-I. The results indicate that chronic IGF-I stimulation leads to mitochondrial dysfunction and reduced cell viability

    Does Reduced IGF-1R Signaling in Igf1r+/− Mice Alter Aging?

    Get PDF
    Mutations in insulin/IGF-1 signaling pathway have been shown to lead to increased longevity in various invertebrate models. Therefore, the effect of the haplo- insufficiency of the IGF-1 receptor (Igf1r+/−) on longevity/aging was evaluated in C57Bl/6 mice using rigorous criteria where lifespan and end-of-life pathology were measured under optimal husbandry conditions using large sample sizes. Igf1r+/− mice exhibited reductions in IGF-1 receptor levels and the activation of Akt by IGF-1, with no compensatory increases in serum IGF-1 or tissue IGF-1 mRNA levels, indicating that the Igf1r+/− mice show reduced IGF-1 signaling. Aged male, but not female Igf1r+/− mice were glucose intolerant, and both genders developed insulin resistance as they aged. Female, but not male Igf1r+/− mice survived longer than wild type mice after lethal paraquat and diquat exposure, and female Igf1r+/− mice also exhibited less diquat-induced liver damage. However, no significant difference between the lifespans of the male Igf1r+/− and wild type mice was observed; and the mean lifespan of the Igf1r+/− females was increased only slightly (less than 5%) compared to wild type mice. A comprehensive pathological analysis showed no significant difference in end-of-life pathological lesions between the Igf1r+/− and wild type mice. These data show that the Igf1r+/− mouse is not a model of increased longevity and delayed aging as predicted by invertebrate models with mutations in the insulin/IGF-1 signaling pathway

    The disruption of proteostasis in neurodegenerative diseases

    Get PDF
    Cells count on surveillance systems to monitor and protect the cellular proteome which, besides being highly heterogeneous, is constantly being challenged by intrinsic and environmental factors. In this context, the proteostasis network (PN) is essential to achieve a stable and functional proteome. Disruption of the PN is associated with aging and can lead to and/or potentiate the occurrence of many neurodegenerative diseases (ND). This not only emphasizes the importance of the PN in health span and aging but also how its modulation can be a potential target for intervention and treatment of human diseases.info:eu-repo/semantics/publishedVersio

    [Lifespan and IGF receptors in the brain.]

    No full text
    International audienceUsing a mouse model relevant for humans, lifespan can be prolonged by reducing IGF-I signaling selectively in the central nervous system. This effect occurred through changes in specific neuroendocrine pathways. Investigating the pathophysiological mechanism, we found that IGF receptors in the brain steered the development of the somatotropic axis, which in turn altered the individual growth trajectory and lifespan. Our work is experimental proof that chronically low IGF-I and low growth hormone (GH) levels favor long lifespan and postpone age-related mortality. Our results, together with other recent reports, challenge the notion that GH can slow down or prevent human aging. This is important because growth hormone is sometimes poposed to elderly people as a substitutive treatment in order to compensate the negative effects of aging. double dagger

    Les récepteurs centraux de l’IGF-1 contrôlent la longévité chez la souris

    No full text
    L’IGF-I, hormone apparentée à l’insuline, est un acteur essentiel de la croissance et du métabolisme des mammifères. Chez la souris, diminuer l’IGF-I (insulin growth factor) ralentit le vieillissement et prolonge la durée de vie tout en augmentant la résistance au stress oxydant. Nos résultats récents indiquent que le cerveau joue un rôle important dans ce processus. Pour les obtenir, nous avons invalidé sélectivement dans le cerveau de souris le gène du récepteur de l’IGF-1. Chez les hétérozygotes, on y constate alors une diminution de l’activité d’IGF-I de 50 % avec pour conséquence, d’abord une baisse significative d’IGF-I dans l’ensemble de l’organisme, puis une nette augmentation de la durée de vie des animaux. Ces résultats suggèrent que la longévité d’un individu dépend du degré de stimulation de la croissance dès après la naissance. Ils suggèrent aussi que l’hormone de croissance (GH), qui augmente fortement l’expression d’IGF-I dans de nombreux tissus, aurait plutôt un effet négatif sur la durée de vie, alors que la GH est parfois utilisée chez des personnes âgées dans l’espoir justement de compenser les effets du vieillissement
    corecore