Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Immun Ageing ; 19(1): 8, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35105357

RESUMEN

BACKGROUND: Rapamycin (Rapa), acarbose (ACA), and 17α-estradiol (17aE2, males only) have health benefits that increase lifespan of mice. Little is known about how these three agents alter the network of pathways downstream of insulin/IGF1 signals as well as inflammatory/stress responses. RESULTS: ACA, Rapa, and 17aE2 (in males, but not in females) oppose age-related increases in the MEK1- ERK1/2-MNK1/2 cascade, and thus reduce phosphorylation of eIF4E, a key component of cap-dependent translation. In parallel, these treatments (in both sexes) reduce age-related increases in the MEK3-p38MAPK-MK2 pathway, to decrease levels of the acute phase response proteins involved in inflammation. CONCLUSION: Each of three drugs converges on the regulation of both the ERK1/2 signaling pathway and the p38-MAPK pathway. The changes induced by treatments in ERK1/2 signaling are seen in both sexes, but the 17aE2 effects are male-specific, consistent with the effects on lifespan. However, the inhibition of age-dependent p38MAPK pathways and acute phase responses is triggered in both sexes by all three drugs, suggesting new approaches to prevention or reversal of age-related inflammatory changes in a clinical setting independent of lifespan effects.

2.
Br J Haematol ; 188(5): 745-756, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31498883

RESUMEN

Haploidentical stem cell transplantation (haploSCT) is becoming a major transplant modality for lymphoma. To assess the effects of donor characteristics, stem cell source and conditioning on outcomes, we identified 474 adults with Hodgkin (HL; 240), peripheral T-cell (PTCL; 88), diffuse large B-cell (77), mantle cell (40) or follicular lymphoma (FL; 29), who received haploSCT with post-transplant cyclophosphamide. Median follow-up of alive patients was 32 months. On multivariate analysis, acute graft-versus-host disease (GVHD) grade 2-4 was lower with offspring donors or bone marrow cells, whereas extensive chronic GVHD was higher in partial response at haploSCT or when using sisters, haploidentical donors beyond first degree, or female donors in male patients. Progression-free survival (PFS) was better for FL, HL and PTCL, whereas overall survival (OS) was better for HL and PTCL. Complete remission at haploSCT improved PFS and OS whereas these were negatively affected by cytomegalovirus donor positive/recipient positive status. No other donor characteristics (age, gender, human leucocyte antigen mismatch, ABO incompatibility) affected PFS or OS except use of haploidentical donors beyond first degree, which negatively affected OS. PFS and OS are mostly influenced by disease status and lymphoma subtype, supporting the use of any first degree haploidentical family member as a donor.


Asunto(s)
Ciclofosfamida/administración & dosificación , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Linfoma , Acondicionamiento Pretrasplante , Enfermedad Aguda , Adolescente , Adulto , Factores de Edad , Anciano , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Linfoma/mortalidad , Linfoma/terapia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores Sexuales , Tasa de Supervivencia , Trasplante Haploidéntico
3.
J Immunol ; 191(9): 4648-55, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078700

RESUMEN

CD4 T cell function declines significantly during aging. Although the mammalian target of rapamycin (TOR) has been implicated in aging, the roles of the TOR complexes (TORC1, TORC2) in the functional declines of CD4 T cells remain unknown. In this study, we demonstrate that aging increases TORC2 signaling in murine CD4 T cells, a change blocked by long-term exposure to rapamycin, suggesting that functional defects may be the result of enhanced TORC2 function. Using overexpression of Rheb to activate TORC1 and Rictor plus Sin1 to augment TORC2 in naive CD4 T cells from young mice, we demonstrated that increased TORC2, but not TORC1, signaling results in aging-associated biochemical changes. Furthermore, elevated TORC2 signaling in naive CD4 T cells from young mice leads to in vivo functional declines. The data presented in this article suggest a novel model in which aging increases TORC2 signaling and leads to CD4 T cell defects in old mice.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Envejecimiento , Animales , Linfocitos T CD4-Positivos/inmunología , Proteínas Portadoras/metabolismo , Células Cultivadas , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/biosíntesis , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/genética , Neuropéptidos/biosíntesis , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
4.
J Immunol ; 189(12): 5582-9, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23136198

RESUMEN

Previous in vitro studies showed that CD4 T cells from old mice have defects in TCR signaling, immune synapse formation, activation, and proliferation. We reported that removing a specific set of surface glycoproteins by ex vivo treatment with O-sialoglycoprotein endopeptidase (OSGE) can reverse many aspects of the age-related decline in CD4 T cell function. However, the specific mechanism by which this process occurs remains unclear, and it is unknown whether this enzymatic treatment can also restore important aspects of adaptive immunity in vivo. By using an in vivo model of the immune response based on adoptive transfer of CD4 T cells from pigeon cytochrome C-specific transgenic H-2(k/k) TCR-Vα(11)Vß(3) CD4(+) mice to syngeneic hosts, we demonstrate that aging diminishes CD28 costimulatory signals in CD4 T cells. These age-associated defects include changes in phosphorylation of AKT and expression of glucose transporter type I, inducible T cell costimulatory molecule, and CD40L, suggesting that the lack of CD28 costimulation contributes to age-dependent loss of CD4 function. All of these deficits can be reversed by ex vivo OSGE treatment. Blocking B7-CD28 interactions on T cells prevents OSGE-mediated restoration of T cell function, suggesting that changes in surface glycosylation, including CD28, may be responsible for the age-related costimulation decline. Finally, we show that the age-related decline in CD4 cognate helper function for IgG production and long-term humoral immunity can also be restored by OSGE treatment of CD4 T cells prior to adoptive transfer.


Asunto(s)
Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/inmunología , Senescencia Celular/inmunología , Inmunoglobulina G/biosíntesis , Metaloendopeptidasas/farmacología , Regulación hacia Arriba/inmunología , Traslado Adoptivo , Envejecimiento/efectos de los fármacos , Envejecimiento/inmunología , Animales , Especificidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Proliferación Celular , Senescencia Celular/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regulación hacia Arriba/efectos de los fármacos
5.
bioRxiv ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36798157

RESUMEN

In defiance of the paradigm that calories from all sources are equivalent, we and others have shown that dietary protein is a dominant regulator of healthy aging. The restriction of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan when initiated in young or adult mice. However, many interventions are less efficacious or even deleterious when initiated in aged animals. Here, we investigate the physiological, metabolic, and molecular consequences of consuming a diet with a 67% reduction of all amino acids (Low AA), or of isoleucine alone (Low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. We find that both diet regimens effectively reduce adiposity and improve glucose tolerance, which were benefits that were not mediated by reduced calorie intake. Both diets improve specific aspects of frailty, slow multiple molecular indicators of aging rate, and rejuvenate the aging heart and liver at the molecular level. These results demonstrate that Low AA and Low Ile diets can drive youthful physiological and molecular signatures, and support the possibility that these dietary interventions could help to promote healthy aging in older adults.

6.
Life Sci ; 328: 121904, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37406767

RESUMEN

Long-lived mouse models and treatments that extend lifespan, such as Rapamycin, acarbose and 17α- -estradiol, lead to reduction in mTORC1 activity, declines in cap-dependent translation and increases in cap-independent translation. In addition, these treatments reduce the MEK-ERK-MNK (ERK1-2) signaling cascade, leading to reduction in eIF4E phosphorylation, which also regulates mRNA translation. Here, we report that Canagliflozin, a drug that extends lifespan only in male mice reduces mTORC1 and ERK1-2 signaling in male mice only. The data suggest reduction in mTORC1 and ERK pathways are common mechanisms shared by both genetic and pharmacological models of slowed aging in mice. Our data also reveal a significant sexual dimorphism in the ERK1-2 signaling pathway which might help to explain why some drugs can extend lifespan in males but have no effects in female mice.


Asunto(s)
Canagliflozina , Longevidad , Masculino , Femenino , Ratones , Animales , Canagliflozina/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosforilación
7.
Commun Biol ; 6(1): 768, 2023 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-37481675

RESUMEN

Aging manifests as progressive deteriorations in homeostasis, requiring systems-level perspectives to investigate the gradual molecular dysregulation of underlying biological processes. Here, we report systemic changes in the molecular regulation of biological processes under multiple lifespan-extending interventions. Differential Rank Conservation (DIRAC) analyses of mouse liver proteomics and transcriptomics data show that mechanistically distinct lifespan-extending interventions (acarbose, 17α-estradiol, rapamycin, and calorie restriction) generally tighten the regulation of biological modules. These tightening patterns are similar across the interventions, particularly in processes such as fatty acid oxidation, immune response, and stress response. Differences in DIRAC patterns between proteins and transcripts highlight specific modules which may be tightened via augmented cap-independent translation. Moreover, the systemic shifts in fatty acid metabolism are supported through integrated analysis of liver transcriptomics data with a mouse genome-scale metabolic model. Our findings highlight the power of systems-level approaches for identifying and characterizing the biological processes involved in aging and longevity.


Asunto(s)
Metabolismo de los Lípidos , Longevidad , Animales , Ratones , Envejecimiento , Modelos Animales de Enfermedad , Hígado , Ácidos Grasos
8.
J Exp Med ; 203(2): 281-7, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16446380

RESUMEN

T cell-specific adapter protein (TSAd) is a SRC-homology-2 (SH2) domain-containing intracellular signaling molecule that is required for T cell antigen receptor (TCR)-induced cytokine synthesis in T cells. How TSAd functions in TCR signal transduction is not clear. Previous work has suggested a nuclear role for this adapter. However, other evidence suggests that TSAd also functions in the cytoplasm. Using T cells from TSAd-deficient mice, we now show that the major role of TSAd in the cytoplasm is in activation of the LCK protein tyrosine kinase at the outset of TCR signal transduction. Consequently, TSAd regulates several downstream signaling events, including intracellular calcium mobilization and activation of the Ras-extracellular signal-regulated kinase signaling pathway. TSAd regulates LCK activity directly through physical interaction with LCK SH3 and SH2 domains. These studies reveal TSAd as a positive regulator of proximal TCR signal transduction and provide important new information on the mechanism of TCR-induced LCK activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Activación Enzimática/genética , Activación Enzimática/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
9.
Aging Cell ; 21(9): e13685, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35930768

RESUMEN

Glycosylphosphatidylinositol-specific phospholipase D1 (GPLD1) hydrolyzes inositol phosphate linkages in proteins anchored to the cell membrane. Mice overexpressing GPLD1 show enhanced neurogenesis and cognition. Snell dwarf (DW) and growth hormone receptor knockout (GKO) mice show delays in age-dependent cognitive decline. We hypothesized that augmented GPLD1 might contribute to retained cognitive function in these mice. We report that DW and GKO show higher GPLD1 levels in the liver and plasma. These mice also have elevated levels of hippocampal brain-derived neurotrophic factor (BDNF) and of doublecortin (DCX), suggesting a mechanism for maintenance of cognitive function at older ages. GPLD1 was not increased in the hippocampus of DW or GKO mice, suggesting that plasma GPLD1 increases elevated these brain proteins. Alteration of the liver and plasma GPLD1 was unaltered in mice with liver-specific GHR deletion, suggesting that the GH effect was not intrinsic to the liver. GPLD1 was also induced by caloric restriction and by each of four drugs that extend lifespan. The proteome of DW and GKO mice is molded by selective translation of mRNAs, involving cap-independent translation (CIT) of mRNAs marked by N6 methyladenosine. Because GPLD1 protein increases were independent of the mRNA level, we tested the idea that GPLD1 might be regulated by CIT. 4EGI-1, which enhances CIT, increased GPLD1 protein without changes in GPLD1 mRNA in cultured fibroblasts and mice. Furthermore, transgenic overexpression of YTHDF1, which promotes CIT by reading m6A signals, also led to increased GPLD1 protein, showing that elevation of GPLD1 reflects selective mRNA translation.


Asunto(s)
Encéfalo , Fosfolipasa D , Animales , Encéfalo/metabolismo , Hígado/metabolismo , Longevidad/genética , Ratones , Fosfolipasa D/genética , ARN Mensajero/metabolismo , Receptores de Somatotropina/metabolismo
10.
Aging Cell ; 20(5): e13345, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33742521

RESUMEN

We hypothesized that rapamycin (Rapa), acarbose (ACA), which both increase mouse lifespan, and 17α-estradiol, which increases lifespan in males (17aE2) all share common intracellular signaling pathways with long-lived Snell dwarf, PAPPA-KO, and Ghr-/- mice. The long-lived mutant mice exhibit reduction in mTORC1 activity, declines in cap-dependent mRNA translation, and increases in cap-independent translation (CIT). Here, we report that Rapa and ACA prevent age-related declines in CIT target proteins in both sexes, while 17aE2 has the same effect only in males, suggesting increases in CIT. mTORC1 activity showed the reciprocal pattern, with age-related increases blocked by Rapa, ACA, and 17aE2 (in males only). METTL3, required for addition of 6-methyl-adenosine to mRNA and thus a trigger for CIT, also showed an age-dependent increase blunted by Rapa, ACA, and 17aE2 (in males). Diminution of mTORC1 activity and increases in CIT-dependent proteins may represent a shared pathway for both long-lived-mutant mice and drug-induced lifespan extension in mice.


Asunto(s)
Acarbosa/farmacología , Estradiol/farmacología , Longevidad/efectos de los fármacos , Longevidad/genética , Biosíntesis de Proteínas , Sirolimus/farmacología , Animales , Femenino , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metiltransferasas/metabolismo , Ratones , Caperuzas de ARN , Transducción de Señal/efectos de los fármacos
11.
Cell Immunol ; 259(1): 100-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19577230

RESUMEN

Activation of lck-fyn kinases during T cell receptor signaling leads to Vav phosphorylation, activation of downstream targets including Rac1, and a transient decline in ezrin and moesin phosphorylation. We have shown that age increases Rac1 activity and lowers ezrin and moesin phosphorylation in resting mouse CD4 cells, changes that could be the results of alterations in lck-Vav signaling. Analysis of Vav in CD4 cells from old mice shows increases in the phosphorylation of two key regulatory residues, Tyr160 and Tyr174, suggesting enhancement of Vav GTPase activity. In addition, analysis of lck status also shows age-related increases in phosphorylation of two key residues, Tyr394 and Tyr505, which have opposite effects on lck function. These changes in lck-Vav signals in resting CD4 cells may contribute in turn to age-related increases in Rac1 activity and declines in phosphorylation of cytoskeletal proteins including Ezrin and Moesin.


Asunto(s)
Envejecimiento/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Animales , Complejo CD3/metabolismo , Proteínas de Unión al ADN/metabolismo , Masculino , Ratones , Neuropéptidos/metabolismo , Fosforilación/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
12.
J Mol Endocrinol ; 63(2): 123-138, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31357177

RESUMEN

It has been hypothesized that transcriptional changes associated with lower mTORC1 activity in mice with reduced levels of growth hormone and insulin-like growth factor 1 are responsible for the longer healthy lifespan of these mutant mice. Cell lines and tissues from these mice show alterations in the levels of many proteins that cannot be explained by corresponding changes in mRNAs. Such post-transcriptional modulation may be the result of preferential mRNA translation by the cap-independent translation of mRNA bearing the N6-methyl-adenosine (m6A) modification. The long-lived endocrine mutants - Snell dwarf, growth hormone receptor deletion and pregnancy-associated plasma protein-A knockout - all show increases in the N6-adenosine-methyltransferases (METTL3/14) that catalyze 6-methylation of adenosine (m6A) in the 5' UTR region of select mRNAs. In addition, these mice have elevated levels of YTH domain-containing protein 1 (YTHDF1), which recognizes m6A and promotes translation by a cap-independent mechanism. Consistently, multiple proteins that can be translated by the cap-independent mechanism are found to increase in these mice, including DNA repair and mitochondrial stress response proteins, without changes in corresponding mRNA levels. Lastly, a drug that augments cap-independent translation by inhibition of cap-dependent pathways (4EGI-1) was found to elevate levels of the same set of proteins and able to render cells resistant to several forms of in vitro stress. Augmented translation by cap-independent pathways facilitated by m6A modifications may contribute to the stress resistance and increased healthy longevity of mice with diminished GH and IGF-1 signals.


Asunto(s)
Sistema Endocrino/metabolismo , Longevidad , Mutación/genética , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , Regulación hacia Arriba , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Fibroblastos/metabolismo , Humanos , Hidrazonas/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Noqueados , Ratones Mutantes , Proteolisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/metabolismo , Tiazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos
14.
Aging Cell ; 16(1): 52-60, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27618784

RESUMEN

Studies of the mTOR pathway have prompted speculation that diminished mTOR complex-1 (mTORC1) function may be involved in controlling the aging process. Our previous studies have shown diminished mTORC1 activity in tissues of three long-lived mutant mice: Snell dwarf mice, growth hormone receptor gene disrupted mice (GHRKO), and in this article, mice deficient in the pregnancy-associated protein-A (PAPPA-KO). The ways in which lower mTOR signals slow aging and age-related diseases are, however, not well characterized. Here, we show that Snell, GHKRO, and PAPPA-KO mice express high levels of two proteins involved in DNA repair, O-6-methylguanine-DNA methyltransferase (MGMT) and N-myc downstream-regulated gene 1 (NDRG1). Furthermore, we report that lowering mTOR enhances MGMT and NDRG1 protein expression via post-transcriptional mechanisms. We show that the CCR4-NOT complex, a post-transcriptional regulator of gene expression, is downstream of the mTORC1 pathway and may be responsible for the upregulation of MGMT and NDRG1 in all three varieties of long-lived mice. Our data thus suggest a novel link between DNA repair and mTOR signaling via post-transcriptional regulation involving specific alteration in the CCR4-NOT complex, whose modulation could control multiple aspects of the aging process.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Enanismo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Longevidad , Proteína Plasmática A Asociada al Embarazo/deficiencia , Receptores de Somatotropina/deficiencia , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Noqueados , Modelos Biológicos , Proteína Plasmática A Asociada al Embarazo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR4/metabolismo , Receptores de Somatotropina/metabolismo , Sirolimus/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
15.
Aging Cell ; 16(6): 1256-1266, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28834262

RESUMEN

Interventions that extend lifespan in mice can show substantial sexual dimorphism. Here, we show that male-specific lifespan extension with two pharmacological treatments, acarbose (ACA) and 17-α estradiol (17aE2), is associated, in males only, with increased insulin sensitivity and improved glucose tolerance. Females, which show either smaller (ACA) or no lifespan extension (17aE2), do not derive these metabolic benefits from drug treatment. We find that these male-specific metabolic improvements are associated with enhanced hepatic mTORC2 signaling, increased Akt activity, and phosphorylation of FOXO1a - changes that might promote metabolic health and survival in males. By manipulating sex hormone levels through gonadectomy, we show that sex-specific changes in these metabolic pathways are modulated, in opposite directions, by both male and female gonadal hormones: Castrated males show fewer metabolic responses to drug treatment than intact males, and only those that are also observed in intact females, while ovariectomized females show some responses similar to those seen in intact males. Our results demonstrate that sex-specific metabolic benefits occur concordantly with sexual dimorphism in lifespan extension. These sex-specific effects can be influenced by the presence of both male and female gonadal hormones, suggesting that gonadally derived hormones from both sexes may contribute to sexual dimorphism in responses to interventions that extend mouse lifespan.


Asunto(s)
Acarbosa/metabolismo , Aminoácidos/metabolismo , Cromo/metabolismo , Estradiol/metabolismo , Hormonas Gonadales/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ácidos Nicotínicos/metabolismo , Animales , Femenino , Humanos , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Caracteres Sexuales , Transducción de Señal
16.
Endocrinology ; 156(2): 565-75, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25456069

RESUMEN

The involvement of mammalian target of rapamycin (mTOR) in lifespan control in invertebrates, calorie-restricted rodents, and extension of mouse lifespan by rapamycin have prompted speculation that diminished mTOR function may contribute to mammalian longevity in several settings. We show here that mTOR complex-1 (mTORC1) activity is indeed lower in liver, muscle, heart, and kidney tissue of Snell dwarf and global GH receptor (GHR) gene-disrupted mice (GHR-/-), consistent with previous studies. Surprisingly, activity of mTORC2 is higher in fasted Snell and GHR-/- than in littermate controls in all 4 tissues tested. Resupply of food enhanced mTORC1 activity in both controls and long-lived mutant mice but diminished mTORC2 activity only in the long-lived mice. Mice in which GHR has been disrupted only in the liver do not show extended lifespan and also fail to show the decline in mTORC1 and increase in mTORC2 seen in mice with global loss of GHR. The data suggest that the antiaging effects in the Snell dwarf and GHR-/- mice are accompanied by both a decline in mTORC1 in multiple organs and an increase in fasting levels of mTORC2. Neither the lifespan nor mTOR effects appear to be mediated by direct GH effects on liver or by the decline in plasma IGF-I, a shared trait in both global and liver-specific GHR-/- mice. Our data suggest that a more complex pattern of hormonal effects and intertissue interactions may be responsible for regulating both lifespan and mTORC2 function in these mouse models of delayed aging.


Asunto(s)
Enanismo Hipofisario/metabolismo , Ayuno/fisiología , Complejos Multiproteicos/metabolismo , Receptores de Somatotropina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Hígado/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos
17.
Ageing Res Rev ; 10(1): 26-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19941976

RESUMEN

It has been postulated that the cytoskeleton controls many aspects of T cell function, including activation, proliferation and apoptosis. Recent advances in our understanding of F-actin polymerization and the Ezrin-Radixin-Moesin (ERM) family of cytoskeleton signal proteins have provided new insights into immunological synapse formation during T cell activation. During aging there is a significant decline of T cell function largely attributable to declines in activation of CD4 T cells and defects in the formation of the immunological synapse. Here we discuss recent progress in the understanding of how aging alters F-actin and ERM proteins in mouse CD4 T cells, and the implications of these changes for the T cell activation process.


Asunto(s)
Envejecimiento/fisiología , Linfocitos T CD4-Positivos/fisiología , Citoesqueleto/fisiología , Transducción de Señal/fisiología , Animales , Proteínas del Citoesqueleto/fisiología , Humanos
18.
Immunobiology ; 216(1-2): 66-71, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20400202

RESUMEN

Declines in immune function have been associated with declines in the function of naïve CD4 T cells. In vitro studies of naïve CD4 T cells in TCR-specific transgenic AND mice have shown age-related defects in immunosynapse formation, activation, proliferation and cytokine production. Previous work has also documented age-related alteration in the glycosylation of surface proteins involved in TCR signaling, and shown that enzymatic treatments to remove specific surface glycoproteins can restore in vitro function in CD4 cells from aged mice. Here an adoptive transfer system shows that a large percentage of naïve CD4 T cells from old mice fail to express CD69 and expand in antigen-primed mice, but these declines in CD69 and expansion can be restored by ex vivo pretreatment of the T cells with the bacterial enzyme O-sialoglycoprotein endopeptidase (OSGE). OSGE treatment also repairs the age-dependent loss of CD69 expression after in vivo activation.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Lectinas Tipo C/metabolismo , Metaloendopeptidasas/farmacología , Envejecimiento/inmunología , Animales , Antígenos/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Glicosilación/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética
19.
J Immunol ; 179(10): 6403-9, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982027

RESUMEN

Cytoskeletal proteins of the ezrin-radixin-moesin (ERM) family contribute to T cell activation in response to Ag, and also to T cell polarization in response to connective tissue matrix proteins and chemokine gradients. Previous work has shown that T cells from aged mice are defective in their ability to develop molecular linkages between surface macromolecules and the underlying cytoskeletal framework, both for proteins that move to the synapse and those that are excluded from the site of T cell-APC interaction. T cells from aged mice also show defective cytoskeletal rearrangements and lamellipodia formation when placed in contact with slides coated with Abs to the TCR/CD3 complex. In this study, we show that old CD4 T cells differ from young CD4 T cells in several aspects of ERM biochemistry, including ERM phosphorylation and ERM associations with CD44, CD43, and EBP50. In addition, CD4 T cells from aged mice show defects in the Rho GTPase activities known to control ERM function.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Proteínas del Citoesqueleto/inmunología , Citoesqueleto/inmunología , Proteínas de Microfilamentos/inmunología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Comunicación Celular/inmunología , Polaridad Celular/inmunología , Quimiocinas/inmunología , Quimiocinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/patología , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Masculino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/inmunología , Seudópodos/inmunología , Seudópodos/metabolismo , Seudópodos/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas de Unión al GTP rho/inmunología , Proteínas de Unión al GTP rho/metabolismo
20.
Immunology ; 119(2): 178-86, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16805789

RESUMEN

Previous work has shown that the function of mouse CD4+ T cells can be augmented by an enzyme, O-sialoglycoprotein endopeptidase (OSGE), which cleaves surface CD43, suggesting the idea that the high levels of glycosylated CD43 found on T cells from aged mice may contribute to immune senescence. New results now show that OSGE improves T-cell function even in mice lacking CD43, showing that other glycoproteins must contribute to the OSGE effect on function. Evaluation of other enzymes found two whose ability to stimulate CD4 activation was higher in aged than in young T cells. One of these, PNGase F, is a glycosidase specific for N-linked glycans, and the other, ST-Siase(2,3) from Salmonella typhimurium, is specific for alpha2,3-linked terminal sialic acid residues. Parallel lectin-binding experiments showed that removal of alpha2,3-linked sialic acid residues vulnerable to PNGase F and ST-Siase(2,3) was also greater in old than in young T cells. The preferential ability of PNGase F and ST-Siase(2,3) to improve the function of T cells from aged mice may involve cleavage of glycoproteins containing alpha2,3-linked sialic acid residues on N-linked or O-linked glycans or both.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Leucosialina/inmunología , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidasas/inmunología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Calcio/metabolismo , Células Cultivadas , Citocinas/biosíntesis , Glicosilación , Lectinas/metabolismo , Lectinas Tipo C , Leucosialina/genética , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Neuraminidasa/inmunología , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/inmunología , Receptores de Interleucina-2/metabolismo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA