Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Trends Biochem Sci ; 45(7): 578-592, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32531228

RESUMEN

Aging is a major risk factor for numerous human pathologies, including cardiovascular, metabolic, musculoskeletal, and neurodegenerative conditions and various malignancies. While our understanding of aging is far from complete, recent advances suggest that targeting fundamental aging processes can delay, prevent, or alleviate age-related disorders. Cellular senescence is physiologically beneficial in several contexts, but it has causal roles in multiple chronic diseases. New studies have illustrated the promising feasibility and safety to selectively ablate senescent cells from tissues, a therapeutic modality that holds potential for treating multiple chronic pathologies and extending human healthspan. Here, we review molecular links between cellular senescence and age-associated complications and highlight novel therapeutic avenues that may be exploited to target senescent cells in future geriatric medicine.


Asunto(s)
Senescencia Celular , Humanos , Neoplasias/patología , Enfermedades Neurodegenerativas/patología , Fenotipo
2.
Am J Transplant ; 24(3): 391-405, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37913871

RESUMEN

In clinical organ transplantation, donor and recipient ages may differ substantially. Old donor organs accumulate senescent cells that have the capacity to induce senescence in naïve cells. We hypothesized that the engraftment of old organs may induce senescence in younger recipients, promoting age-related pathologies. When performing isogeneic cardiac transplants between age-mismatched C57BL/6 old donor (18 months) mice and young and middle-aged C57BL/6 (3- or 12- month-old) recipients , we observed augmented frequencies of senescent cells in draining lymph nodes, adipose tissue, livers, and hindlimb muscles 30 days after transplantation. These observations went along with compromised physical performance and impaired spatial learning and memory abilities. Systemic levels of the senescence-associated secretory phenotype factors, including mitochondrial DNA (mt-DNA), were elevated in recipients. Of mechanistic relevance, injections of mt-DNA phenocopied effects of age-mismatched organ transplantation on accelerating aging. Single treatment of old donor animals with senolytics prior to transplantation attenuated mt-DNA release and improved physical capacities in young recipients. Collectively, we show that transplanting older organs induces senescence in transplant recipients, resulting in compromised physical and cognitive capacities. Depleting senescent cells with senolytics, in turn, represents a promising approach to improve outcomes of older organs.


Asunto(s)
Senescencia Celular , Trasplante de Órganos , Animales , Ratones , Senoterapéuticos , Ratones Endogámicos C57BL , Trasplante de Órganos/efectos adversos , ADN/farmacología , Envejecimiento/fisiología
3.
Gerontology ; 70(1): 7-14, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37879300

RESUMEN

BACKGROUND: As the largest organ in the human body, the skin is continuously exposed to intrinsic and extrinsic stimuli that impact its functionality and morphology with aging. Skin aging entails dysregulation of skin cells and loss, fragmentation, or fragility of extracellular matrix fibers that are manifested macroscopically by wrinkling, laxity, and pigmentary abnormalities. Age-related skin changes are the focus of many surgical and nonsurgical treatments aimed at improving overall skin appearance and health. SUMMARY: As a hallmark of aging, cellular senescence, an essentially irreversible cell cycle arrest with apoptosis resistance and a secretory phenotype, manifests across skin layers by affecting epidermal and dermal cells. Knowledge of skin-specific senescent cells, such as melanocytes (epidermal aging) and fibroblasts (dermal aging), will promote our understanding of age-related skin changes and how to optimize patient outcomes in esthetic procedures. KEY MESSAGES: This review provides an overview of skin aging in the context of cellular senescence and discusses senolytic intervention strategies to selectively target skin senescent cells that contribute to premature skin aging.


Asunto(s)
Senoterapéuticos , Envejecimiento de la Piel , Humanos , Envejecimiento/fisiología , Senescencia Celular/fisiología , Melanocitos , Piel
4.
EMBO J ; 38(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30737259

RESUMEN

Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age-related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post-mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age-related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent-like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length-independent telomere damage in cardiomyocytes activates the classical senescence-inducing pathways, p21CIP and p16INK4a, and results in a non-canonical senescence-associated secretory phenotype, which is pro-fibrotic and pro-hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age-related myocardial dysfunction and in the wider setting to ageing in post-mitotic tissues.


Asunto(s)
Cardiomegalia/patología , Senescencia Celular , Daño del ADN , Fibrosis/patología , Mitosis , Miocitos Cardíacos/patología , Acortamiento del Telómero , Envejecimiento , Animales , Cardiomegalia/etiología , Femenino , Fibrosis/etiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monoaminooxidasa/fisiología , Miocitos Cardíacos/metabolismo , Fenotipo , ARN/fisiología , Ratas Sprague-Dawley , Telomerasa/fisiología
5.
Semin Immunol ; 40: 101275, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-31088710

RESUMEN

Senescent cells (SCs) arise from normal cells in multiple organs due to inflammatory, metabolic, DNA damage, or tissue damage signals. SCs are non-proliferating but metabolically active cells that can secrete a range of pro-inflammatory and proteolytic factors as part of the senescence-associated secretory phenotype (SASP). Senescent cell anti-apoptotic pathways (SCAPs) protect SCs from their own pro-apoptotic SASP. SCs can chemo-attract immune cells and are usually cleared by these immune cells. During aging and in multiple chronic diseases, SCs can accumulate in dysfunctional tissues. SCs can impede innate and adaptive immune responses. Whether immune system loss of capacity to clear SCs promotes immune system dysfunction, or conversely whether immune dysfunction permits SC accumulation, are important issues that are not yet fully resolved. SCs may be able to assume distinct states that interact differentially with immune cells, thereby promoting or inhibiting SC clearance, establishing a chronically pro-senescent and pro-inflammatory environment, leading to modulation of the SASP by the immune cells recruited and activated by the SASP. Therapies that enhance immune cell-mediated clearance of SCs could provide a lever for reducing SC burden. Such therapies could include vaccines, small molecule immunomodulators, or other approaches. Senolytics, drugs that selectively eliminate SCs by transiently disabling their SCAPs, may prove to alleviate immune dysfunction in older individuals and thereby accelerate immune-mediated clearance of SCs. The more that can be understood about the interplay between SCs and the immune system, the faster new interventions may be developed to delay, prevent, or treat age-related dysfunction and the multiple senescence-associated chronic diseases and disorders.


Asunto(s)
Apoptosis , Senescencia Celular , Sistema Inmunológico/fisiología , Envejecimiento , Enfermedad Crónica/terapia , Humanos , Fenotipo
6.
Handb Exp Pharmacol ; 274: 165-180, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34697668

RESUMEN

Obesity is a major risk factor for the development of comorbidities such as type 2 diabetes, neurodegenerative disorders, osteoarthritis, cancer, cardiovascular and renal diseases. The onset of obesity is linked to an increase of senescent cells within adipose tissue and other organs. Cellular senescence is a stress response that has been shown to be causally linked to aging and development of various age-related diseases such as obesity. The senescence-associated-secretory phenotype of senescent cells creates a chronic inflammatory milieu that leads to local and systemic dysfunction. The elimination of senescent cells using pharmacological approaches (i.e., senolytics) has been shown to delay, prevent, or alleviate obesity-related organ dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2 , Senoterapéuticos , Senescencia Celular/fisiología , Humanos , Obesidad/tratamiento farmacológico
7.
J Am Soc Nephrol ; 32(8): 1987-2004, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34135081

RESUMEN

BACKGROUND: Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS: Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS: Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS: Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.


Asunto(s)
Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Isquemia/fisiopatología , Riñón/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Quinasas p21 Activadas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Enfermedad Crónica , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Dasatinib/farmacología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal , Expresión Génica , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Humanos , Isquemia/etiología , Riñón/irrigación sanguínea , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteopontina/genética , Inhibidores de Proteínas Quinasas/farmacología , Obstrucción de la Arteria Renal/complicaciones , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Regulación hacia Arriba , Quinasas p21 Activadas/genética
8.
J Physiol ; 599(11): 2869-2886, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33745126

RESUMEN

KEY POINTS: Critical illness myopathy (CIM) is a frequently observed negative consequence of modern critical care. Chronic Janus kinase (JAK)/signal transducer and activator of transcription activation impairs muscle size and function and is prominent following mechanical ventilation. We identify pSTAT-3 activation in tibialis anterior of CIM patients, before examining the potential benefits of JAK1/2 inhibition in an experimental model of CIM, where muscle mass and function are impaired. CIM activates complement cascade and increased monocyte infiltration in the soleus muscle, which was ameliorated by JAK1/2 inhibition, leading to reduced muscle degeneration and improved muscle force. Here, we demonstrate that JAK1/2 inhibition augments CIM muscle function through regulation of the complement cascade. ABSTRACT: Critical illness myopathy (CIM) is frequently observed in response to modern critical care with negative consequences for patient quality of life, morbidity, mortality and healthcare costs. Janus kinase (JAK)/signal transducer and activator of transcription (STAT) activation is observed in limb muscles following controlled mechanical ventilation. Chronic JAK/STAT activation promotes loss of muscle mass and function. Thus, we hypothesized that JAK1/2 inhibition would improve muscle outcomes for CIM. Following 12 days of intensive care unit conditions, pSTAT-3 levels increased in tibialis anterior muscle of CIM patients (P = 0.0489). The potential of JAK1/2 inhibition was assessed in an experimental model of CIM, where soleus muscle size and force are impaired. JAK1/2 inhibition restores soleus force (P < 0.0001). CIM activated muscle complement cascade, which was ameliorated by JAK1/2 inhibition (P < 0.05, respectively). Soleus macrophage number corresponded with complement activity, leading to reduced muscle degeneration and augmented muscle function (P < 0.05). Thus, JAK/STAT inhibition improves soleus function by modulating the complement cascade and muscle monocyte infiltration. Collectively, we demonstrate that JAK/STAT inhibition augments muscle function in CIM.


Asunto(s)
Quinasas Janus , Enfermedades Musculares , Animales , Complemento C3 , Enfermedad Crítica , Humanos , Músculo Esquelético , Calidad de Vida , Ratas , Transductores
9.
J Cell Physiol ; 236(2): 1332-1344, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32657444

RESUMEN

Cell stress may give rise to insuperable growth arrest, which is defined as cellular senescence. Stenotic kidney (STK) ischemia and injury induced by renal artery stenosis (RAS) may be associated with cellular senescence. Mesenchymal stem cells (MSCs) decrease some forms of STK injury, but their ability to reverse senescence in RAS remains unknown. We hypothesized that RAS evokes STK senescence, which would be ameliorated by MSCs. Mice were studied after 4 weeks of RAS, RAS treated with adipose tissue-derived MSCs 2 weeks earlier, or sham. STK senescence-associated ß-galactosidase (SA-ß-Gal) activity was measured. Protein and gene expression was used to assess senescence and the senescence-associated secretory phenotype (SASP), and staining for renal fibrosis, inflammation, and capillary density. In addition, senescence was assessed as p16+ and p21+ urinary exosomes in patients with renovascular hypertension (RVH) without or 3 months after autologous adipose tissue-derived MSC delivery, and in healthy volunteers (HV). In RAS mice, STK SA-ß-Gal activity increased, and senescence and SASP marker expression was markedly elevated. MSCs improved renal function, fibrosis, inflammation, and capillary density, and attenuated SA-ß-Gal activity, but most senescence and SASP levels remained unchanged. Congruently, in human RVH, p21+ urinary exosomes were elevated compared to HV, and only slightly improved by MSC, whereas p16+ exosomes remained unchanged. Therefore, RAS triggers renal senescence in both mice and human subjects. MSCs decrease renal injury, but only partly mitigate renal senescence. These observations support exploration of targeted senolytic therapy in RAS.


Asunto(s)
Senescencia Celular/genética , Trasplante de Células Madre Mesenquimatosas , Obstrucción de la Arteria Renal/terapia , beta-Galactosidasa/genética , Tejido Adiposo/citología , Animales , Modelos Animales de Enfermedad , Exosomas/genética , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/terapia , Riñón/metabolismo , Riñón/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Obstrucción de la Arteria Renal/genética , Obstrucción de la Arteria Renal/patología
10.
Am J Physiol Renal Physiol ; 318(5): F1167-F1176, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32223312

RESUMEN

Cellular senescence, a permanent arrest of cell proliferation, is characterized by a senescence-associated secretory phenotype (SASP), which reinforces senescence and exerts noxious effects on adjacent cells. Recent studies have suggested that transplanting small numbers of senescent cells suffices to provoke tissue inflammation. We hypothesized that senescent cells can directly augment renal injury. Primary scattered tubular-like cells (STCs) acquired from pig kidneys were irradiated by 10 Gy of cesium radiation, and 3 wk later cells were characterized for levels of senescence and SASP markers. Control or senescent STCs were then prelabeled and injected (5 × 105 cells) into the aorta of C57BL/6J mice. Four weeks later, renal oxygenation was studied in vivo using 16.4-T magnetic resonance imaging and function by plasma creatinine level. Renal markers of SASP, fibrosis, and microvascular density were evaluated ex vivo. Per flow cytometry, irradiation induced senescence in 80-99% of STCs, which showed increased gene expression of senescence and SASP markers, senescence-associated ß-galactosidase staining, and cytokine levels (especially IL-6) secreted in conditioned medium. Four weeks after injection, cells were detected engrafted in the mouse kidneys with no evidence for rejection. Plasma creatinine and renal tissue hypoxia increased in senescent compared with control cells. Senescent kidneys were more fibrotic, with fewer CD31+ endothelial cells, and showed upregulation of IL-6 gene expression. Therefore, exogenously delivered senescent renal STCs directly injure healthy mouse kidneys. Additional studies are needed to determine the role of endogenous cellular senescence in the pathogenesis of kidney injury and evaluate the utility of senolytic therapy.


Asunto(s)
Proliferación Celular , Senescencia Celular , Túbulos Renales/trasplante , Riñón/cirugía , Animales , Proliferación Celular/efectos de la radiación , Células Cultivadas , Senescencia Celular/efectos de la radiación , Femenino , Fibrosis , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Túbulos Renales/efectos de la radiación , Masculino , Ratones Endogámicos C57BL , Fenotipo , Sus scrofa , Trasplante Heterólogo
11.
Am J Physiol Renal Physiol ; 315(5): F1493-F1499, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30019935

RESUMEN

There is no therapy that promotes maturation and functionality of a dialysis arteriovenous fistula (AVF). The search for such therapies largely relies on evaluation of vascular responses and putative therapies in experimental AVFs. We studied an AVF in mice with chronic kidney disease (CKD). We demonstrate numerous stressors in the vein of the AVF-CKD group, including pathological shear, mitogenic, inflammatory, and hypoxia-reoxygenation stress. Because stress promotes premature senescence, we examined whether senescence is induced in the vein of the AVF-CKD model. We demonstrate a senescence phenotype in the AVF-CKD model, as indicated by increased expression of p16Ink4a, p21Cip1, and p53 and expected changes for certain senescence-associated microRNAs. RNA-sequencing analysis demonstrated differential expression of ~10,000 genes, including upregulation of proinflammatory and proliferative genes, in the vein of the AVF-CKD group. The vein in the AVF-CKD group exhibited telomere erosion and increased senescence-associated ß-galactosidase activity and staining. Senescence was induced in the artery of the AVF-CKD group and in the vein of the AVF without CKD. Finally, given the rapidly rising clinical interest in senolytics, we provide proof of concept of senolytics as a therapeutic approach by demonstrating that senolytics decrease p16Ink4a expression in the AVF-CKD model. This study introduces a novel concept underlying the basis for maturational and functional failure in human dialysis AVFs and identifies a new target for senolytic therapy.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Senescencia Celular , Complicaciones Posoperatorias/patología , Diálisis Renal , Insuficiencia Renal Crónica/terapia , Cola (estructura animal)/irrigación sanguínea , Remodelación Vascular , Venas/cirugía , Animales , Derivación Arteriovenosa Quirúrgica/efectos adversos , Velocidad del Flujo Sanguíneo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Dasatinib/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/fisiopatología , Quercetina/farmacología , Flujo Sanguíneo Regional , Estrés Mecánico , Grado de Desobstrucción Vascular , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética , Venas/metabolismo , Venas/patología , Venas/fisiopatología
12.
Proc Natl Acad Sci U S A ; 112(46): E6301-10, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578790

RESUMEN

Chronic, low grade, sterile inflammation frequently accompanies aging and age-related diseases. Cellular senescence is associated with the production of proinflammatory chemokines, cytokines, and extracellular matrix (ECM) remodeling proteases, which comprise the senescence-associated secretory phenotype (SASP). We found a higher burden of senescent cells in adipose tissue with aging. Senescent human primary preadipocytes as well as human umbilical vein endothelial cells (HUVECs) developed a SASP that could be suppressed by targeting the JAK pathway using RNAi or JAK inhibitors. Conditioned medium (CM) from senescent human preadipocytes induced macrophage migration in vitro and inflammation in healthy adipose tissue and preadipocytes. When the senescent cells from which CM was derived had been treated with JAK inhibitors, the resulting CM was much less proinflammatory. The administration of JAK inhibitor to aged mice for 10 wk alleviated both adipose tissue and systemic inflammation and enhanced physical function. Our findings are consistent with a possible contribution of senescent cells and the SASP to age-related inflammation and frailty. We speculate that SASP inhibition by JAK inhibitors may contribute to alleviating frailty. Targeting the JAK pathway holds promise for treating age-related dysfunction.


Asunto(s)
Adipocitos/enzimología , Senescencia Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Quinasas Janus/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/enzimología , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Senescencia Celular/genética , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Macrófagos/citología , Macrófagos/enzimología , Ratones , ARN Interferente Pequeño/genética , Transducción de Señal/genética
13.
Gerontology ; 62(2): 163-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26372907

RESUMEN

BACKGROUND: Growth hormone (GH)-resistant/deficient mice experience improved glucose homeostasis and substantially increased lifespan. Recent evidence suggests that long-lived GH-resistant/deficient mice are protected from white adipose tissue (WAT) dysfunction, including WAT cellular senescence, impaired adipogenesis and loss of subcutaneous WAT in old age. This preservation of WAT function has been suggested to be a potential mechanism for the extended lifespan of these mice. OBJECTIVE: The objective of this study was to examine WAT senescence, WAT distribution and glucose homeostasis in dwarf GH receptor antagonist (GHA) transgenic mice, a unique mouse strain having decreased GH action but normal longevity. METHODS: 18-month-old female GHA mice and wild-type (WT) littermate controls were used. Prior to dissection, body composition, fasting blood glucose as well as glucose and insulin tolerance tests were performed. WAT distribution was determined by weighing four distinct WAT depots at the time of dissection. Cellular senescence in four WAT depots was assessed using senescence-associated ß-galactosidase staining to quantify the senescent cell burden, and real-time qPCR to quantify gene expression of senescence markers p16 and IL-6. RESULTS: GHA mice had a 22% reduction in total body weight, a 33% reduction in lean mass and a 10% increase in body fat percentage compared to WT controls. GHA mice had normal fasting blood glucose and improved insulin sensitivity; however, they exhibited impaired glucose tolerance. Moreover, GHA mice displayed enhanced lipid storage in the inguinal subcutaneous WAT depot (p < 0.05) and a 1.7-fold increase in extra-/intraperitoneal WAT ratio compared to controls (p < 0.05). Measurements of WAT cellular senescence showed no difference between GHA mice and WT controls. CONCLUSIONS: Similar to other mice with decreased GH action, female GHA mice display reduced age-related lipid redistribution and improved insulin sensitivity, but no change in cellular senescence. The similar abundance of WAT senescent cells in GHA and control mice suggests that any protection against generation of senescent cells afforded by decreased GH action, low insulin-like growth factor 1 and/or improved insulin sensitivity in the GHA mice may be offset by their severe adiposity, since obesity is known to increase senescence.


Asunto(s)
Adipocitos Blancos , Glucemia/metabolismo , Senescencia Celular/genética , Receptores de Somatotropina/genética , Grasa Subcutánea , Adipogénesis/genética , Animales , Femenino , Intolerancia a la Glucosa/metabolismo , Homeostasis/genética , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/metabolismo , Síndrome de Laron , Ratones , Ratones Transgénicos , beta-Galactosidasa/metabolismo
14.
Cancer ; 121(10): 1540-7, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25529481

RESUMEN

Young adult childhood cancer survivors are at an increased risk of frailty, a physiologic phenotype typically found among older adults. This phenotype is associated with new-onset chronic health conditions and mortality among both older adults and childhood cancer survivors. Mounting evidence suggests that poor fitness, muscular weakness, and cognitive decline are common among adults treated for childhood malignancies, and that risk factors for these outcomes are not limited to those treated with cranial radiation. Although the pathobiology of this phenotype is not known, early cellular senescence, sterile inflammation, and mitochondrial dysfunction in response to initial cancer or treatment-related insults are hypothesized to play a role. To the authors' knowledge, interventions to prevent or remediate frailty among childhood cancer survivors have not been tested to date. Pharmaceutical, nutraceutical, and lifestyle interventions have demonstrated some promise.


Asunto(s)
Disfunción Cognitiva/etiología , Debilidad Muscular/etiología , Neoplasias , Sobrevivientes/estadística & datos numéricos , Adulto , Senescencia Celular , Niño , Enfermedad Crónica , Disfunción Cognitiva/epidemiología , Disfunción Cognitiva/prevención & control , Irradiación Craneana/efectos adversos , Suplementos Dietéticos/estadística & datos numéricos , Tolerancia al Ejercicio , Fuerza de la Mano , Humanos , Incidencia , Inflamación , Mitocondrias , Debilidad Muscular/epidemiología , Debilidad Muscular/prevención & control , Factores de Riesgo , Conducta de Reducción del Riesgo
15.
Curr Opin Clin Nutr Metab Care ; 17(4): 324-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24848532

RESUMEN

PURPOSE OF REVIEW: Possible mechanisms in cellular senescence and the senescence-associated secretory phenotype (SASP) that drive and promote chronic inflammation in multiple age-related chronic diseases are considered. RECENT FINDINGS: A series of studies about the SASP indicate that senescent cells may be involved in the development of chronic inflammatory diseases associated with aging. SUMMARY: Aging is a complex biological process accompanied by a state of chronic, low-grade, 'sterile' inflammation, which is a major contributor to the development of many age-related chronic disorders including atherosclerosis, osteoarthritis, Alzheimer's disease, type 2 diabetes, cancers, and others. It appears that cellular senescence plays a role in causing inflammation through the SASP. A better understanding of the contribution of senescent cells to the pathologies of chronic inflammatory disorders could have potentially profound diagnostic and therapeutic implications.


Asunto(s)
Envejecimiento , Senescencia Celular , Enfermedad de Alzheimer/fisiopatología , Aterosclerosis/fisiopatología , Enfermedad Crónica , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Hipertensión/fisiopatología , Inflamación/fisiopatología , Neoplasias/fisiopatología , Osteoartritis/fisiopatología , Fenotipo
16.
Proc Natl Acad Sci U S A ; 108(1): 143-8, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21173238

RESUMEN

Brown fat is specialized for energy expenditure and has therefore been proposed to function as a defense against obesity. Despite recent advances in delineating the transcriptional regulation of brown adipocyte differentiation, cellular lineage specification and developmental cues specifying brown-fat cell fate remain poorly understood. In this study, we identify and isolate a subpopulation of adipogenic progenitors (Sca-1(+)/CD45(-)/Mac1(-); referred to as Sca-1(+) progenitor cells, ScaPCs) residing in murine brown fat, white fat, and skeletal muscle. ScaPCs derived from different tissues possess unique molecular expression signatures and adipogenic capacities. Importantly, although the ScaPCs from interscapular brown adipose tissue (BAT) are constitutively committed brown-fat progenitors, Sca-1(+) cells from skeletal muscle and subcutaneous white fat are highly inducible to differentiate into brown-like adipocytes upon stimulation with bone morphogenetic protein 7 (BMP7). Consistent with these findings, human preadipocytes isolated from subcutaneous white fat also exhibit the greatest inducible capacity to become brown adipocytes compared with cells isolated from mesenteric or omental white fat. When muscle-resident ScaPCs are re-engrafted into skeletal muscle of syngeneic mice, BMP7-treated ScaPCs efficiently develop into adipose tissue with brown fat-specific characteristics. Importantly, ScaPCs from obesity-resistant mice exhibit markedly higher thermogenic capacity compared with cells isolated from obesity-prone mice. These data establish the molecular characteristics of tissue-resident adipose progenitors and demonstrate a dynamic interplay between these progenitors and inductive signals that act in concert to specify brown adipocyte development.


Asunto(s)
Adipocitos Marrones/fisiología , Tejido Adiposo Blanco/citología , Antígenos Ly/metabolismo , Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo , Músculo Esquelético/citología , Células Madre/fisiología , Adipocitos Marrones/citología , Animales , Western Blotting , Proteína Morfogenética Ósea 7/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Termogénesis/fisiología
17.
JAMA ; 321(9): 901-902, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30835305
18.
Ageing Res Rev ; 99: 102400, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945306

RESUMEN

It has been four years since long COVID-the protracted consequences that survivors of COVID-19 face-was first described. Yet, this entity continues to devastate the quality of life of an increasing number of COVID-19 survivors without any approved therapy and a paucity of clinical trials addressing its biological root causes. Notably, many of the symptoms of long COVID are typically seen with advancing age. Leveraging this similarity, we posit that Geroscience-which aims to target the biological drivers of aging to prevent age-associated conditions as a group-could offer promising therapeutic avenues for long COVID. Bearing this in mind, this review presents a translational framework for studying long COVID as a state of effectively accelerated biological aging, identifying research gaps and offering recommendations for future preclinical and clinical studies.


Asunto(s)
Envejecimiento , COVID-19 , Humanos , SARS-CoV-2 , Gerociencia , Síndrome Post Agudo de COVID-19
19.
Endocr Rev ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38500373

RESUMEN

Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type II diabetes mellitus (T2DM). Drugs that selectively induce senescent cell removal, "senolytics", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics", appear to delay the onset or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. Over thirty clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.

20.
Acta Physiol (Oxf) ; 240(5): e14128, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38551103

RESUMEN

AIM: Mechanical ventilation (MV) results in diminished diaphragm size and strength, termed ventilator-induced diaphragm dysfunction (VIDD). VID increases dependence, prolongs weaning, and increases discharge mortality rates. The Janus kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) pathway is implicated in VIDD, upregulated following MV. JAK/STAT inhibition alleviates chronic muscle wasting conditions. This study aimed to explore the therapeutic potential of Ruxolitinib, an FDA approved JAK1/2 inhibitor (JI) for the treatment of VIDD. METHODS: Rats were subjected to 5 days controlled MV (CMV) with and without daily Ruxolitinib gavage. Muscle fiber size and function were assessed. RNAseq, mitochondrial morphology, respirometry, and mass spectrometry were determined. RESULTS: CMV significantly reduced diaphragm size and specific force by 45% (p < 0.01), associated with a two-fold P-STAT3 upregulation (p < 0.001). CMV disrupted mitochondrial content and reduced the oxygen consumption rate (p < 0.01). Expression of the motor protein myosin was unaffected, however CMV alters myosin function via post-translational modifications (PTMs). Daily administration of JI increased animal survival (40% vs. 87%; p < 0.05), restricted P-STAT3 (p < 0.001), and preserved diaphragm size and specific force. JI was associated with preserved mitochondrial content and respiratory function (p < 0.01), and the reversal or augmentation of myosin deamidation PTMs of the rod and head region. CONCLUSION: JI preserved diaphragm function, leading to increased survival in an experimental model of VIDD. Functional enhancement was associated with maintenance of mitochondrial content and respiration and the reversal of ventilator-induced PTMs of myosin. These results demonstrate the potential of repurposing Ruxolitinib for treatment of VIDD.


Asunto(s)
Diafragma , Nitrilos , Pirazoles , Pirimidinas , Respiración Artificial , Animales , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Diafragma/fisiopatología , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Nitrilos/farmacología , Ratas , Respiración Artificial/efectos adversos , Masculino , Pirazoles/farmacología , Pirazoles/uso terapéutico , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA