Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(10)2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37079375

RESUMEN

The intake of dietary phosphate far exceeds recommended levels; however, the long-term health consequences remain relatively unknown. Here, the chronic physiological response to sustained elevated and reduced dietary phosphate consumption was investigated in mice. Although serum phosphate levels were brought into homeostatic balance, the prolonged intake of a high-phosphate diet dramatically and negatively impacted bone volume; generated a sustained increase in the phosphate responsive circulating factors FGF23, PTH, osteopontin and osteocalcin; and produced a chronic low-grade inflammatory state in the BM, marked by increased numbers of T cells expressing IL-17a, RANKL, and TNF-α. In contrast, a low-phosphate diet preserved trabecular bone while increasing cortical bone volume over time, and it reduced inflammatory T cell populations. Cell-based studies identified a direct response of T cells to elevated extracellular phosphate. Neutralizing antibodies against proosteoclastic cytokines RANKL, TNF-α, and IL-17a blunted the high-phosphate diet-induced bone loss identifying bone resorption as a regulatory mechanism. Collectively, this study illuminates that habitual consumption of a high-phosphate diet in mice induces chronic inflammation in bone, even in the absence of elevated serum phosphate. Furthermore, the study supports the concept that a reduced phosphate diet may be a simple yet effective strategy to reduce inflammation and improve bone health during aging.


Asunto(s)
Resorción Ósea , Fósforo Dietético , Ratones , Animales , Interleucina-17 , Factor de Necrosis Tumoral alfa , Linfocitos T , Citocinas , Inflamación , Fosfatos
2.
Brain ; 146(5): 2107-2119, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36345573

RESUMEN

Synaptic dysfunction is one of the earliest pathological processes that contribute to the development of many neurological disorders, including Alzheimer's disease and frontotemporal lobar degeneration. However, the synaptic function of many disease-causative genes and their contribution to the pathogenesis of the related diseases remain unclear. In this study, we investigated the synaptic role of fused in sarcoma, an RNA-binding protein linked to frontotemporal lobar degeneration and amyotrophic lateral sclerosis, and its potential pathological role in frontotemporal lobar degeneration using pyramidal neuron-specific conditional knockout mice (FuscKO). We found that FUS regulates the expression of many genes associated with synaptic function in a hippocampal subregion-specific manner, concomitant with the frontotemporal lobar degeneration-linked behavioural disinhibition. Electrophysiological study and molecular pathway analyses further reveal that fused in sarcoma differentially regulates synaptic and neuronal properties in the ventral hippocampus and medial prefrontal cortex, respectively. Moreover, fused in sarcoma selectively modulates the ventral hippocampus-prefrontal cortex projection, which is known to mediate the anxiety-like behaviour. Our findings unveil the brain region- and synapse-specific role of fused in sarcoma, whose impairment might lead to the emotional symptoms associated with frontotemporal lobar degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Sarcoma , Animales , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/patología , Demencia Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Proteína FUS de Unión a ARN/genética , Sarcoma/metabolismo , Sarcoma/patología
3.
JBMR Plus ; 6(7): e10636, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35866149

RESUMEN

Cyclic adenosine monophosphate (cAMP)-dependent phosphodiesterase (PDE) inhibitors such as pentoxifylline (PTX) suppress cAMP degradation and promote cAMP-dependent signal transduction. PDE inhibitors increase bone formation and bone mass in preclinical models and are used clinically to treat psoriatic arthritis by targeting inflammatory mediators including activated T cells. T cell activation requires two signals: antigen-dependent CD3-activation, which stimulates cAMP production; and CD28 co-stimulation, which downregulates cAMP-signaling, through PDE activation. PDE-inhibitors consequently suppress T cell activation by disrupting CD28 co-stimulation. Interestingly, we have reported that when CD8+ T cells are activated in the absence of CD28 co-stimulation, they secrete Wnt-10b, a bone anabolic Wnt ligand that promotes bone formation. In the present study, we investigated whether the bone anabolic activity of the PDE-inhibitor PTX, has an immunocentric basis, involving Wnt-10b production by CD8+ T cells. When wild-type (WT) mice were administered PTX, biochemical markers of both bone resorption and formation were significantly increased, with net bone gain in the axial skeleton, as quantified by micro-computed tomography (µCT). By contrast, PTX increased only bone resorption in T cell knockout (KO) mice, causing net bone loss. Reconstituting T cell-deficient mice with WT, but not Wnt-10b knockout (KO) CD8+ T cells, rescued bone formation and prevented bone loss. To study the role of cAMP signaling in Wnt-10b expression, reverse-transcription polymerase chain reaction (RT-PCR) and luciferase-reporter assays were performed using primary T cells. PDE inhibitors intensified Wnt-10b promoter activity and messenger RNA (mRNA) accumulation in CD3 and CD28 activated CD8+ T cells. In contrast, inhibiting the cAMP pathway mediators protein kinase A (PKA) and cAMP response element-binding protein (CREB), suppressed Wnt-10b expression by T cells activated in the absence of CD28 co-stimulation. In conclusion, the data demonstrate a key role for Wnt-10b production by CD8+ T cells in the bone anabolic response to PDE-inhibitors and reveal competing T cell-independent pro-resorptive properties of PTX, which dominate under T cell-deficient conditions. Selective targeting of CD8+ T cells by PDE inhibitors may be a beneficial approach for promoting bone regeneration in osteoporotic conditions. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

4.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33586672

RESUMEN

Estrogen deficiency causes a gut microbiome-dependent expansion of BM Th17 cells and TNF-α-producing T cells. The resulting increased BM levels of IL-17a (IL-17) and TNF stimulate RANKL expression and activity, causing bone loss. However, the origin of BM Th17 cells and TNF+ T cells is unknown. Here, we show that ovariectomy (ovx) expanded intestinal Th17 cells and TNF+ T cells, increased their S1P receptor 1-mediated (S1PR1-mediated) egress from the intestine, and enhanced their subsequent influx into the BM through CXCR3- and CCL20-mediated mechanisms. Demonstrating the functional relevance of T cell trafficking, blockade of Th17 cell and TNF+ T cell egress from the gut or their influx into the BM prevented ovx-induced bone loss. Therefore, intestinal T cells are a proximal target of sex steroid deficiency relevant for bone loss. Blockade of intestinal T cell migration may represent a therapeutic strategy for the treatment of postmenopausal bone loss.


Asunto(s)
Movimiento Celular/inmunología , Intestinos , Osteoporosis Posmenopáusica , Ovariectomía , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Femenino , Humanos , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Noqueados , Osteoporosis Posmenopáusica/inmunología , Osteoporosis Posmenopáusica/microbiología , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Factor de Necrosis Tumoral alfa/genética
5.
Elife ; 102021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33432923

RESUMEN

Genetic factors account for the majority of the variance of human bone mass, but the contribution of non-genetic factors remains largely unknown. By utilizing maternal/offspring transmission, cohabitation, or fecal material transplantation (FMT) studies, we investigated the influence of the gut microbiome on skeletal maturation. We show that the gut microbiome is a communicable regulator of bone structure and turnover in mice. In addition, we found that the acquisition of a specific bacterial strain, segmented filamentous bacteria (SFB), a gut microbe that induces intestinal Th17 cell expansion, was sufficient to negatively impact skeletal maturation. These findings have significant translational implications, as the identification of methods or timing of microbiome transfer may lead to the development of bacteriotherapeutic interventions to optimize skeletal maturation in humans. Moreover, the transfer of SFB-like microbes capable of triggering the expansion of human Th17 cells during therapeutic FMT procedures could lead to significant bone loss in fecal material recipients.


Asunto(s)
Microbioma Gastrointestinal , Esqueleto/crecimiento & desarrollo , Animales , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Ratones
6.
J Clin Invest ; 130(4): 1767-1781, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917685

RESUMEN

Parathyroid hormone (PTH) is a critical regulator of skeletal development that promotes both bone formation and bone resorption. Using microbiota depletion by wide-spectrum antibiotics and germ-free (GF) female mice, we showed that the microbiota was required for PTH to stimulate bone formation and increase bone mass. Microbiota depletion lowered butyrate levels, a metabolite responsible for gut-bone communication, while reestablishment of physiologic levels of butyrate restored PTH-induced anabolism. The permissive activity of butyrate was mediated by GPR43 signaling in dendritic cells and by GPR43-independent signaling in T cells. Butyrate was required for PTH to increase the number of bone marrow (BM) regulatory T cells (Tregs). Tregs stimulated production of the osteogenic Wnt ligand Wnt10b by BM CD8+ T cells, which activated Wnt-dependent bone formation. Together, these data highlight the role that butyrate produced by gut luminal microbiota plays in triggering regulatory pathways, which are critical for the anabolic action of PTH in bone.


Asunto(s)
Butiratos/metabolismo , Microbioma Gastrointestinal , Osteogénesis , Hormona Paratiroidea/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Ratones , Ratones Noqueados , Hormona Paratiroidea/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfocitos T Reguladores/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
7.
Nat Commun ; 11(1): 468, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980603

RESUMEN

Bone loss is a frequent but not universal complication of hyperparathyroidism. Using antibiotic-treated or germ-free mice, we show that parathyroid hormone (PTH) only caused bone loss in mice whose microbiota was enriched by the Th17 cell-inducing taxa segmented filamentous bacteria (SFB). SFB+ microbiota enabled PTH to expand intestinal TNF+ T and Th17 cells and increase their S1P-receptor-1 mediated egress from the intestine and recruitment to the bone marrow (BM) that causes bone loss. CXCR3-mediated TNF+ T cell homing to the BM upregulated the Th17 chemoattractant CCL20, which recruited Th17 cells to the BM. This study reveals mechanisms for microbiota-mediated gut-bone crosstalk in mice models of hyperparathyroidism that may help predict its clinical course. Targeting the gut microbiota or T cell migration may represent therapeutic strategies for hyperparathyroidism.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Osteoporosis/etiología , Hormona Paratiroidea/inmunología , Subgrupos de Linfocitos T/inmunología , Células Th17/inmunología , Animales , Trasplante de Microbiota Fecal , Femenino , Vida Libre de Gérmenes , Bacilos Grampositivos Formadores de Endosporas/inmunología , Hiperparatiroidismo Primario/complicaciones , Hiperparatiroidismo Primario/inmunología , Hiperparatiroidismo Primario/microbiología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoporosis/inmunología , Osteoporosis/microbiología , Factor de Necrosis Tumoral alfa/inmunología
8.
J Bone Miner Res ; 34(2): 349-360, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30399207

RESUMEN

Primary hyperparathyroidism (PHPT) is a condition where elevated PTH levels lead to bone loss, in part through increased production of the osteoclastogenic factor IL-17A, by bone marrow (BM) T-helper 17 (Th17) cells, a subset of helper CD4+ T cells. In animals, PHPT is modeled by continuous PTH treatment (cPTH). In mice, an additional critical action of cPTH is the capacity to increase the production of RANKL by osteocytes. However, a definitive link between IL-17A and osteocytic expression of RANKL has not been made. Here we show that cPTH fails to induce cortical and trabecular bone loss and causes less intense bone resorption in conditional knock-out (IL-17RAΔOCY ) male and female mice lacking the expression of IL-17A receptor (IL-17RA) in dentin matrix protein 1 (DMP1)-8kb-Cre-expressing cells, which include osteocytes and some osteoblasts. Therefore, direct IL-17RA signaling in osteoblasts/osteocytes is required for cPTH to exert its bone catabolic effects. In addition, in vivo, silencing of IL-17RA signaling in in DMP1-8kb-expressing cells blunts the capacity of cPTH to stimulate osteocytic RANKL production, indicating that cPTH augments osteocytic RANKL expression indirectly, via an IL-17A/IL-17RA-mediated mechanism. Thus, osteocytic production of RANKL and T cell production of IL-17A are both critical for the bone catabolic activity of cPTH. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Resorción Ósea/metabolismo , Osteocitos/metabolismo , Hormona Paratiroidea/metabolismo , Ligando RANK/biosíntesis , Receptores de Interleucina-17/metabolismo , Transducción de Señal , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Hiperparatiroidismo Primario/genética , Hiperparatiroidismo Primario/metabolismo , Hiperparatiroidismo Primario/patología , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Ratones Noqueados , Osteocitos/patología , Hormona Paratiroidea/genética , Ligando RANK/genética , Receptores de Interleucina-17/genética
9.
Immunity ; 49(6): 1116-1131.e7, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30446387

RESUMEN

Nutritional supplementation with probiotics can prevent pathologic bone loss. Here we examined the impact of supplementation with Lactobacillus rhamnosus GG (LGG) on bone homeostasis in eugonadic young mice. Micro-computed tomography revealed that LGG increased trabecular bone volume in mice, which was due to increased bone formation. Butyrate produced in the gut following LGG ingestion, or butyrate fed directly to germ-free mice, induced the expansion of intestinal and bone marrow (BM) regulatory T (Treg) cells. Interaction of BM CD8+ T cells with Treg cells resulted in increased secretion of Wnt10b, a bone anabolic Wnt ligand. Mechanistically, Treg cells promoted the assembly of a NFAT1-SMAD3 transcription complex in CD8+ cells, which drove expression of Wnt10b. Reducing Treg cell numbers, or reconstitution of TCRß-/- mice with CD8+ T cells from Wnt10b-/- mice, prevented butyrate-induced bone formation and bone mass acquisition. Thus, butyrate concentrations regulate bone anabolism via Treg cell-mediated regulation of CD8+ T cell Wnt10b production.


Asunto(s)
Butiratos/farmacología , Osteogénesis/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Proteínas Wnt/metabolismo , Animales , Butiratos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular , Proliferación Celular/efectos de los fármacos , Femenino , Lacticaseibacillus rhamnosus/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Probióticos/administración & dosificación , Probióticos/metabolismo , Linfocitos T Reguladores/citología , Proteínas Wnt/genética
10.
EMBO Rep ; 19(1): 156-171, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29158349

RESUMEN

Teriparatide is a bone anabolic treatment for osteoporosis, modeled in animals by intermittent PTH (iPTH) administration, but the cellular and molecular mechanisms of action of iPTH are largely unknown. Here, we show that Teriparatide and iPTH cause a ~two-threefold increase in the number of regulatory T cells (Tregs) in humans and mice. Attesting in vivo relevance, blockade of the Treg increase in mice prevents the increase in bone formation and trabecular bone volume and structure induced by iPTH Therefore, increasing the number of Tregs is a pivotal mechanism by which iPTH exerts its bone anabolic activity. Increasing Tregs pharmacologically may represent a novel bone anabolic therapy, while iPTH-induced Treg increase may find applications in inflammatory conditions and transplant medicine.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Hormonas y Agentes Reguladores de Calcio/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Teriparatido/uso terapéutico , Anciano , Animales , Biomarcadores/metabolismo , Calcio/uso terapéutico , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Sialoproteína de Unión a Integrina/genética , Sialoproteína de Unión a Integrina/metabolismo , Recuento de Linfocitos , Ratones , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoporosis Posmenopáusica/genética , Osteoporosis Posmenopáusica/metabolismo , Osteoporosis Posmenopáusica/patología , Ovariectomía , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Resultado del Tratamiento , Vitamina D/análogos & derivados , Vitamina D/uso terapéutico
11.
Cell Rep ; 14(5): 1218-1231, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26832412

RESUMEN

In an immune response, CD4(+) T cells expand into effector T cells and then contract to survive as long-lived memory cells. To identify age-associated defects in memory cell formation, we profiled activated CD4(+) T cells and found an increased induction of the ATPase CD39 with age. CD39(+) CD4(+) T cells resembled effector T cells with signs of metabolic stress and high susceptibility to undergo apoptosis. Pharmacological inhibition of ATPase activity dampened effector cell differentiation and improved survival, suggesting that CD39 activity influences T cell fate. Individuals carrying a low-expressing CD39 variant responded better to vaccination with an increase in vaccine-specific memory T cells. Increased inducibility of CD39 after activation may contribute to the impaired vaccine response with age.


Asunto(s)
Envejecimiento/inmunología , Antígenos CD/metabolismo , Apirasa/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos/inmunología , Adenosina Trifosfatasas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Proliferación Celular , Supervivencia Celular , Células Clonales , Humanos , Persona de Mediana Edad , Estrés Fisiológico , Adulto Joven
12.
J Bone Miner Res ; 31(5): 949-63, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26614970

RESUMEN

Hydrogen sulfide (H2 S) is a gasotransmitter known to regulate bone formation and bone mass in unperturbed mice. However, it is presently unknown whether H2 S plays a role in pathologic bone loss. Here we show that ovariectomy (ovx), a model of postmenopausal bone loss, decreases serum H2 S levels and the bone marrow (BM) levels of two key H2 S-generating enzymes, cystathione ß-synthase (CBS) and cystathione γ-lyase (CSE). Treatment with the H2 S-donor GYY4137 (GYY) normalizes serum H2 S in ovx mice, increases bone formation, and completely prevents the loss of trabecular bone induced by ovx. Mechanistic studies revealed that GYY increases murine osteoblastogenesis by activating Wnt signaling through increased production of the Wnt ligands Wnt16, Wnt2b, Wnt6, and Wnt10b in the BM. Moreover, in vitro treatment with 17ß-estradiol upregulates the expression of CBS and CSE in human BM stromal cells (hSCs), whereas an H2 S-releasing drug induces osteogenic differentiation of hSCs. In summary, regulation of H2 S levels is a novel mechanism by which estrogen stimulates osteoblastogenesis and bone formation in mice and human cells. Blunted production of H2 S contributes to ovx-induced bone loss in mice by limiting the compensatory increase in bone formation elicited by ovx. Restoration of H2 S levels is a potential novel therapeutic approach for postmenopausal osteoporosis. © 2015 American Society for Bone and Mineral Research.


Asunto(s)
Estrógenos/deficiencia , Sulfuro de Hidrógeno/metabolismo , Osteogénesis , Osteoporosis Posmenopáusica/metabolismo , Vía de Señalización Wnt , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Femenino , Humanos , Ratones , Osteoporosis Posmenopáusica/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Proteínas Wnt/metabolismo
13.
Cell Metab ; 22(5): 799-810, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26456334

RESUMEN

Primary hyperparathyroidism (PHPT) is a common cause of bone loss that is modeled by continuous PTH (cPTH) infusion. Here we show that the inflammatory cytokine IL-17A is upregulated by PHPT in humans and cPTH in mice. In humans, IL-17A is normalized by parathyroidectomy. In mice, treatment with anti-IL-17A antibody and silencing of IL-17A receptor IL-17RA prevent cPTH-induced osteocytic and osteoblastic RANKL production and bone loss. Mechanistically, cPTH stimulates conventional T cell production of TNFα (TNF), which increases the differentiation of IL-17A-producing Th17 cells via TNF receptor 1 (TNFR1) signaling in CD4(+) cells. Moreover, cPTH enhances the sensitivity of naive CD4(+) cells to TNF via GαS/cAMP/Ca(2+) signaling. Accordingly, conditional deletion of GαS in CD4(+) cells and treatment with the calcium channel blocker diltiazem prevents Th17 cell expansion and blocks cPTH-induced bone loss. Neutralization of IL-17A and calcium channel blockers may thus represent novel therapeutic strategies for hyperparathyroidism.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Hiperparatiroidismo Primario/metabolismo , Interleucina-17/metabolismo , Animales , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/etiología , Enfermedades Óseas Metabólicas/patología , Bloqueadores de los Canales de Calcio/uso terapéutico , Humanos , Hiperparatiroidismo Primario/complicaciones , Hiperparatiroidismo Primario/tratamiento farmacológico , Hiperparatiroidismo Primario/patología , Interleucina-17/biosíntesis , Ratones , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/biosíntesis
15.
J Bone Miner Res ; 30(4): 695-705, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25359628

RESUMEN

T cells are known to potentiate the bone anabolic activity of intermittent parathyroid hormone (iPTH) treatment. One of the involved mechanisms is increased T cell secretion of Wnt10b, a potent osteogenic Wnt ligand that activates Wnt signaling in stromal cells (SCs). However, additional mechanisms might play a role, including direct interactions between surface receptors expressed by T cells and SCs. Here we show that iPTH failed to promote SC proliferation and differentiation into osteoblasts (OBs) and activate Wnt signaling in SCs of mice with a global or T cell-specific deletion of the T cell costimulatory molecule CD40 ligand (CD40L). Attesting to the relevance of T cell-expressed CD40L, iPTH induced a blunted increase in bone formation and failed to increase trabecular bone volume in CD40L(-/-) mice and mice with a T cell-specific deletion of CD40L. CD40L null mice exhibited a blunted increase in T cell production of Wnt10b and abrogated CD40 signaling in SCs in response to iPTH treatment. Therefore, expression of the T cell surface receptor CD40L enables iPTH to exert its bone anabolic activity by activating CD40 signaling in SCs and maximally stimulating T cell production of Wnt10b.


Asunto(s)
Anabolizantes/farmacología , Huesos/efectos de los fármacos , Ligando de CD40/inmunología , Hormona Paratiroidea/farmacología , Linfocitos T/inmunología , Anabolizantes/administración & dosificación , Animales , Ligando de CD40/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hormona Paratiroidea/administración & dosificación
16.
Nat Med ; 18(10): 1518-24, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23023500

RESUMEN

The ability of the human immune system to respond to vaccination declines with age. We identified an age-associated defect in T cell receptor (TCR)-induced extracellular signal-regulated kinase (ERK) phosphorylation in naive CD4(+) T cells, whereas other signals, such as ζ chain-associated protein kinase 70 (ZAP70) and phospholipase C-γ1 phosphorylation, were not impaired. The defective ERK signaling was caused by the dual specific phosphatase 6 (DUSP6), whose protein expression increased with age due to a decline in repression by miR-181a. Reconstitution of miR-181a lowered DUSP6 expression in naive CD4(+) T cells in elderly individuals. DUSP6 repression using miR-181a or specific siRNA and DUSP6 inhibition by the allosteric inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one improved CD4(+) T cell responses, as seen by increased expression of activation markers, improved proliferation and supported preferential T helper type 1 cell differentiation. DUSP6 is a potential intervention target for restoring T cell responses in the elderly, which may augment the effectiveness of vaccination.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , MicroARNs/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Ciclohexilaminas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Indenos/farmacología , Activación de Linfocitos , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Fosforilación , Receptores de Antígenos de Linfocitos T/genética , Proteína Tirosina Quinasa ZAP-70/metabolismo
17.
Semin Immunol ; 24(5): 365-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22560928

RESUMEN

With increasing age, the ability of the immune system to protect against new antigenic challenges or to control chronic infections erodes. Decline in thymic function and cumulating antigenic experiences of acute and chronic infections threaten T cell homeostasis, but insufficiently explain the failing immune competence and the increased susceptibility for autoimmunity. Alterations in signaling pathways in the aging T cells account for many of the age-related defects. Signaling threshold calibrations seen with aging frequently built on mechanisms that are operational in T cell development and T cell differentiation or are adaptations to the changing environment in the aging host. Age-related changes in transcription of receptors and signaling molecules shift the balance towards inhibitory pathways, most dominantly seen in CD8 T cells and to a lesser degree in CD4 T cells. Prominent examples are the expression of negative regulatory receptors of the CD28 and the TNF receptor superfamilies as well the expression of various cytoplasmic and nuclear dual-specific phosphatases.


Asunto(s)
Diferenciación Celular , Senescencia Celular , Transducción de Señal , Linfocitos T/inmunología , Envejecimiento , Animales , Humanos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/metabolismo
18.
Proc Natl Acad Sci U S A ; 109(25): E1629-37, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22615393

RESUMEN

Autoantibodies to common autoantigens and neoantigens, such as IgG Fc and citrullinated peptides, are immunological hallmarks of rheumatoid arthritis (RA). We examined whether a failure in maintaining tolerance is mediated by defects in T-cell receptor activation threshold settings. RA T cells responded to stimulation with significantly higher ERK phosphorylation (P < 0.001). Gene expression arrays of ERK pathway members suggested a higher expression of KRAS and BRAF, which was confirmed by quantitative PCR (P = 0.003), Western blot, and flow cytometry (P < 0.01). Partial silencing of KRAS and BRAF lowered activation-induced phosphorylated ERK levels (P < 0.01). In individual cells, levels of these signaling molecules correlated with ERK phosphorylation, attesting that their concentrations are functionally important. In confocal studies, B-RAF/K-RAS clustering was increased in RA T cells 2 min after T-cell receptor stimulation (P < 0.001). Overexpression of B-RAF and K-RAS in normal CD4 T cells amplified polyclonal T-cell proliferation and facilitated responses to citrullinated peptides. We propose that increased expression of B-RAF and K-RAS lowers T-cell activation thresholds in RA T cells, enabling responses to autoantigens.


Asunto(s)
Artritis Reumatoide/inmunología , GTP Fosfohidrolasas/metabolismo , Genes ras , Tolerancia Inmunológica , Proteínas Proto-Oncogénicas B-raf/metabolismo , Linfocitos T/inmunología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Subgrupos de Linfocitos T
19.
Proc Natl Acad Sci U S A ; 109(15): E879-88, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22434910

RESUMEN

T cell-dependent B-cell responses decline with age, suggesting defective CD4 T-cell function. CD4 memory T cells from individuals older than 65 y displayed increased and sustained transcription of the dual-specific phosphatase 4 (DUSP4) that shortened expression of CD40-ligand (CD40L) and inducible T-cell costimulator (ICOS) (both P < 0.001) and decreased production of IL-4, IL-17A, and IL-21 (all P < 0.001) after in vitro activation. In vivo after influenza vaccination, activated CD4 T cells from elderly individuals had increased DUSP4 transcription (P = 0.002), which inversely correlated with the expression of CD40L (r = 0.65, P = 0.002), ICOS (r = 0.57, P = 0.008), and IL-4 (r = 0.66, P = 0.001). In CD4 KO mice reconstituted with DUSP4 OT-II T cells, DUSP4 had a negative effect on the expansion of antigen-specific B cells (P = 0.003) and the production of ova-specific antibodies (P = 0.03) after immunization. Silencing of DUSP4 in memory CD4 T cells improved CD40L (P < 0.001), IL-4 (P = 0.007), and IL-21 (P = 0.04) expression significantly more in the elderly than young adults. Consequently, the ability of CD4 memory T cells to support B-cell differentiation that was impaired in the elderly (P = 0.004) was restored. Our data suggest that increased DUSP4 expression in activated T cells in the elderly in part accounts for defective adaptive immune responses.


Asunto(s)
Envejecimiento/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Animales , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Silenciador del Gen , Humanos , Inmunidad Humoral/inmunología , Memoria Inmunológica/genética , Gripe Humana/inmunología , Activación de Linfocitos/inmunología , Ratones , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación , Adulto Joven
20.
J Exp Med ; 208(4): 775-85, 2011 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-21422171

RESUMEN

Zinc is a trace element that is essential for innate and adaptive immune responses. In addition to being a structural element of many proteins, zinc also functions as a neurotransmitter and an intracellular messenger. Temporal or spatial changes in bioavailable zinc may influence the activity of several enzymes, including kinases and phosphatases. We provide evidence that zinc functions as an ionic signaling molecule after T cell activation. Cytoplasmic zinc concentrations increased within 1 min after T cell receptor (TCR) triggering, in particular in the subsynaptic compartment. The increase depended on the extracellular zinc concentrations and was inhibited by silencing zinc transporter Zip6. Increased zinc influx reduced the recruitment of SHP-1 to the TCR activation complex, augmented ZAP70 phosphorylation and sustained calcium influx. By calibrating TCR activation thresholds, increased extracellular zinc bioavailability facilitated the induction of T cell proliferative responses to suboptimal stimuli.


Asunto(s)
Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Zinc/metabolismo , Adulto , Proteína Tirosina Quinasa CSK , Proteínas de Transporte de Catión/fisiología , Citoplasma/metabolismo , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteína Tirosina Quinasa ZAP-70/fisiología , Familia-src Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...