Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(4)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33586672

RESUMO

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.


Assuntos
Movimento Celular/imunologia , Intestinos , Osteoporose Pós-Menopausa , Ovariectomia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Quimiocina CCL20/genética , Quimiocina CCL20/imunologia , Feminino , Humanos , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Knockout , Osteoporose Pós-Menopausa/imunologia , Osteoporose Pós-Menopausa/microbiologia , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Fator de Necrose Tumoral alfa/genética
2.
Nat Commun ; 11(1): 468, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980603

RESUMO

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.


Assuntos
Microbioma Gastrointestinal/imunologia , Osteoporose/etiologia , Hormônio Paratireóideo/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th17/imunologia , Animais , Transplante de Microbiota Fecal , Feminino , Vida Livre de Germes , Bacilos Gram-Positivos Formadores de Endosporo/imunologia , Hiperparatireoidismo Primário/complicações , Hiperparatireoidismo Primário/imunologia , Hiperparatireoidismo Primário/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoporose/imunologia , Osteoporose/microbiologia , Fator de Necrose Tumoral alfa/imunologia
3.
J Bone Miner Res ; 34(2): 349-360, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30399207

RESUMO

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.


Assuntos
Reabsorção Óssea/metabolismo , Osteócitos/metabolismo , Hormônio Paratireóideo/metabolismo , Ligante RANK/biossíntese , Receptores de Interleucina-17/metabolismo , Transdução de Sinais , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Hiperparatireoidismo Primário/genética , Hiperparatireoidismo Primário/metabolismo , Hiperparatireoidismo Primário/patologia , Interleucina-17/genética , Interleucina-17/metabolismo , Camundongos , Camundongos Knockout , Osteócitos/patologia , Hormônio Paratireóideo/genética , Ligante RANK/genética , Receptores de Interleucina-17/genética
4.
EMBO Rep ; 19(1): 156-171, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158349

RESUMO

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.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Hormônios e Agentes Reguladores de Cálcio/uso terapêutico , Osteoporose Pós-Menopausa/tratamento farmacológico , Linfócitos T Reguladores/efeitos dos fármacos , Teriparatida/uso terapêutico , Idoso , Animais , Biomarcadores/metabolismo , Cálcio/uso terapêutico , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Sialoproteína de Ligação à Integrina/genética , Sialoproteína de Ligação à Integrina/metabolismo , Contagem de Linfócitos , Camundongos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoporose Pós-Menopausa/genética , Osteoporose Pós-Menopausa/metabolismo , Osteoporose Pós-Menopausa/patologia , Ovariectomia , Fator de Transcrição Sp7/genética , Fator de Transcrição Sp7/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Resultado do Tratamento , Vitamina D/análogos & derivados , Vitamina D/uso terapêutico
5.
J Clin Invest ; 126(6): 2049-63, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27111232

RESUMO

A eubiotic microbiota influences many physiological processes in the metazoan host, including development and intestinal homeostasis. Here, we have shown that the intestinal microbiota modulates inflammatory responses caused by sex steroid deficiency, leading to trabecular bone loss. In murine models, sex steroid deficiency increased gut permeability, expanded Th17 cells, and upregulated the osteoclastogenic cytokines TNFα (TNF), RANKL, and IL-17 in the small intestine and the BM. In germ-free (GF) mice, sex steroid deficiency failed to increase osteoclastogenic cytokine production, stimulate bone resorption, and cause trabecular bone loss, demonstrating that the gut microbiota is central in sex steroid deficiency-induced trabecular bone loss. Furthermore, we demonstrated that twice-weekly treatment of sex steroid-deficient mice with the probiotics Lactobacillus rhamnosus GG (LGG) or the commercially available probiotic supplement VSL#3 reduces gut permeability, dampens intestinal and BM inflammation, and completely protects against bone loss. In contrast, supplementation with a nonprobiotic strain of E. coli or a mutant LGG was not protective. Together, these data highlight the role that the gut luminal microbiota and increased gut permeability play in triggering inflammatory pathways that are critical for inducing bone loss in sex steroid-deficient mice. Our data further suggest that probiotics that decrease gut permeability have potential as a therapeutic strategy for postmenopausal osteoporosis.


Assuntos
Microbioma Gastrointestinal/fisiologia , Hormônios Esteroides Gonadais/deficiência , Osteoporose Pós-Menopausa/etiologia , Osteoporose Pós-Menopausa/prevenção & controle , Probióticos/farmacologia , Animais , Remodelação Óssea , Fenômenos Fisiológicos do Sistema Digestório , Modelos Animais de Doenças , Feminino , Vida Livre de Germes , Humanos , Interleucina-17/metabolismo , Lacticaseibacillus rhamnosus , Camundongos , Camundongos Endogâmicos C57BL , Osteoporose Pós-Menopausa/patologia , Permeabilidade , Ligante RANK/metabolismo , Células Th17/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
J Bone Miner Res ; 31(5): 949-63, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26614970

RESUMO

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.


Assuntos
Estrogênios/deficiência , Sulfeto de Hidrogênio/metabolismo , Osteogênese , Osteoporose Pós-Menopausa/metabolismo , Via de Sinalização Wnt , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Feminino , Humanos , Camundongos , Osteoporose Pós-Menopausa/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Proteínas Wnt/metabolismo
7.
Cell Metab ; 22(5): 799-810, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26456334

RESUMO

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.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Hiperparatireoidismo Primário/metabolismo , Interleucina-17/metabolismo , Animais , Doenças Ósseas Metabólicas/tratamento farmacológico , Doenças Ósseas Metabólicas/etiologia , Doenças Ósseas Metabólicas/patologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Humanos , Hiperparatireoidismo Primário/complicações , Hiperparatireoidismo Primário/tratamento farmacológico , Hiperparatireoidismo Primário/patologia , Interleucina-17/biossíntese , Camundongos , Receptores Tipo I de Fatores de Necrose Tumoral/biossíntese , Transdução de Sinais , Linfócitos T/metabolismo , Linfócitos T/patologia , Fator de Necrose Tumoral alfa/biossíntese
8.
J Bone Miner Res ; 30(4): 695-705, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25359628

RESUMO

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.


Assuntos
Anabolizantes/farmacologia , Osso e Ossos/efeitos dos fármacos , Ligante de CD40/imunologia , Hormônio Paratireóideo/farmacologia , Linfócitos T/imunologia , Anabolizantes/administração & dosagem , Animais , Ligante de CD40/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hormônio Paratireóideo/administração & dosagem
9.
Blood ; 122(14): 2346-57, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23954891

RESUMO

Estrogen deficiency expands hemopoietic stem and progenitor cells (HSPCs) and mature blood lineages, but the involved mechanism and the affected HSPC populations are mostly unknown. Here we show that ovariectomy (ovx) expands short-term HSPCs (ST-HSPCs) and improves blood cell engraftment and host survival after bone marrow (BM) transplantation through a dual role of the T-cell costimulatory molecule CD40 ligand (CD40L). This surface receptor is required for ovx to stimulate T-cell production of Wnt10b, a Wnt ligand that activates Wnt signaling in HSPCs and stromal cells (SCs). Moreover, CD40L is required for ovx to increase SC production of the hemopoietic cytokines interleukin (IL)-6, IL-7, and granulocyte macrophage-colony-stimulating factor. Attesting to the relevance of CD40L and Wnt10b, ovx fails to expand ST-HSPCs in CD40L-null mice and in animals lacking global or T-cell expression of Wnt10b. In summary, T cells expressed CD40L, and the resulting increased production of Wnt10b and hemopoietic cytokines by T cells and SCs, respectively, plays a pivotal role in the mechanism by which ovx regulates hemopoiesis. The data suggest that antiestrogens may represent pharmacological targets to improve ST-HSPC function through activation of the microenvironment.


Assuntos
Ligante de CD40/biossíntese , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Ovariectomia , Linfócitos T/metabolismo , Proteínas Wnt/biossíntese , Animais , Transplante de Medula Óssea/imunologia , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Proteínas Wnt/imunologia
10.
Blood ; 120(22): 4352-62, 2012 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22955916

RESUMO

Intermittent parathyroid hormone (iPTH) treatment expands hemopoietic stem and progenitor cells (HSPCs), but the involved mechanisms and the affected HSPC populations are mostly unknown. Here we show that T cells are required for iPTH to expand short-term HSPCs (ST-HSPCs) and improve blood cell engraftment and host survival after BM transplantation. Silencing of PTH/PTH-related protein receptor (PPR) in T cells abrogates the effects of iPTH, thus demonstrating a requirement for direct PPR signaling in T cells. Mechanistically, iPTH expands ST-HSPCs by activating Wnt signaling in HSPCs and stromal cells (SCs) through T-cell production of the Wnt ligand Wnt10b. Attesting to the relevance of Wnt10b, iPTH fails to expand ST-HSPCs in mice with Wnt10b(-/-) T cells. Moreover, iPTH fails to promote engraftment and survival after BM transplantation in Wnt10b null mice. In summary, direct PPR signaling in T cells and the resulting production of Wnt10b play a pivotal role in the mechanism by which iPTH expands ST-HSPCs. The data suggest that T cells may provide pharmacologic targets for HSPC expansion.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Linfócitos T/fisiologia , Animais , Transplante de Medula Óssea , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/efeitos dos fármacos , Fatores de Tempo , Proteínas Wnt/genética
11.
Proc Natl Acad Sci U S A ; 109(12): E725-33, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393015

RESUMO

Intermittent parathyroid hormone (iPTH) treatment stimulates T-cell production of the osteogenic Wnt ligand Wnt10b, a factor required for iPTH to activate Wnt signaling in osteoblasts and stimulate bone formation. However, it is unknown whether iPTH induces Wnt10b production and bone anabolism through direct activation of the parathyroid hormone (PTH)/PTH-related protein receptor (PPR) in T cells. Here, we show that conditional silencing of PPR in T cells blunts the capacity of iPTH to induce T-cell production of Wnt10b; activate Wnt signaling in osteoblasts; expand the osteoblastic pool; and increase bone turnover, bone mineral density, and trabecular bone volume. These findings demonstrate that direct PPR signaling in T cells plays an important role in PTH-induced bone anabolism by promoting T-cell production of Wnt10b and suggest that T cells may provide pharmacological targets for bone anabolism.


Assuntos
Osso e Ossos/metabolismo , Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Animais , Densidade Óssea , Feminino , Inativação Gênica , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Microtomografia por Raio-X/métodos
12.
Proc Natl Acad Sci U S A ; 108(2): 768-73, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187391

RESUMO

The bone loss induced by ovariectomy (ovx) has been linked to increased production of osteoclastogenic cytokines by bone marrow cells, including T cells and stromal cells (SCs). It is presently unknown whether regulatory interactions between these lineages contribute to the effects of ovx in bone, however. Here, we show that the T-cell costimulatory molecule CD40 ligand (CD40L) is required for ovx to expand SCs; promote osteoblast proliferation and differentiation; regulate the SC production of the osteoclastogenic factors macrophage colony-stimulating factor, receptor activator of nuclear factor-κB ligand, and osteoprotegerin; and up-regulate osteoclast formation. CD40L is also required for ovx to activate T cells and stimulate their production of TNF. Accordingly, ovx fails to promote bone loss and increase bone resorption in mice depleted of T cells or lacking CD40L. Therefore, cross-talk between T cells and SCs mediated by CD40L plays a pivotal role in the disregulation of osteoblastogenesis and osteoclastogenesis induced by ovx.


Assuntos
Ligante de CD40/metabolismo , Osteoblastos/citologia , Osteoclastos/citologia , Linfócitos T/citologia , Animais , Técnicas de Cocultura , Estrogênios/metabolismo , Humanos , Ligantes , Camundongos , NF-kappa B/metabolismo , Osteoporose/metabolismo , Osteoprotegerina/metabolismo , Ovariectomia/métodos , Fator de Necrose Tumoral alfa/metabolismo
13.
PLoS One ; 5(8): e12290, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20808842

RESUMO

BACKGROUND: Hyperparathyroidism in humans and continuous parathyroid hormone (cPTH) treatment in mice cause bone loss by regulating the production of RANKL and OPG by stromal cells (SCs) and osteoblasts (OBs). Recently, it has been reported that T cells are required for cPTH to induce bone loss as the binding of the T cell costimulatory molecule CD40L to SC receptor CD40 augments SC sensitivity to cPTH. However it is unknown whether direct PTH stimulation of T cells is required for cPTH to induce bone loss, and whether T cells contribute to the bone catabolic activity of PTH with mechanisms other than induction of CD40 signaling in SCs. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that silencing of PTH receptor 1 (PPR) in T cells blocks the bone loss and the osteoclastic expansion induced by cPTH, thus demonstrating that PPR signaling in T cells is central for PTH-induced reduction of bone mass. Mechanistic studies revealed that PTH activation of the T cell PPR stimulates T cell production of the osteoclastogenic cytokine tumor necrosis factor alpha (TNF). Attesting to the relevance of this effect, disruption of T cell TNF production prevents PTH-induced bone loss. We also show that a novel mechanism by which TNF mediates PTH induced osteoclast formation is upregulation of CD40 expression in SCs, which increases their RANKL/OPG production ratio. CONCLUSIONS/SIGNIFICANCE: These findings demonstrate that PPR signaling in T cells plays an essential role in PTH induced bone loss by promoting T cell production of TNF. A previously unknown effect of TNF is to increase SC expression of CD40, which in turn increases SC osteoclastogenic activity by upregulating their RANKL/OPG production ratio. PPR-dependent stimulation of TNF production by T cells and the resulting TNF regulation of CD40 signaling in SCs are potential new therapeutic targets for the bone loss of hyperparathyroidism.


Assuntos
Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/metabolismo , Inativação Gênica , Hormônio Paratireóideo/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo/deficiência , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Linfócitos T/metabolismo , Animais , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Feminino , Humanos , Masculino , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Linfócitos T/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese
14.
Dev Cell ; 16(1): 35-46, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19154717

RESUMO

Developing organs require iron for a myriad of functions, but embryos deleted of the major adult transport proteins, transferrin or its receptor transferrin receptor1 (TfR1(-/-)), still initiate organogenesis, suggesting that non-transferrin pathways are important. To examine these pathways, we developed chimeras composed of fluorescence-tagged TfR1(-/-) cells and untagged wild-type cells. In the kidney, TfR1(-/-) cells populated capsule and stroma, mesenchyme and nephron, but were underrepresented in ureteric bud tips. Consistently, TfR1 provided transferrin to the ureteric bud, but not to the capsule or the stroma. Instead of transferrin, we found that the capsule internalized ferritin. Since the capsule expressed a novel receptor called Scara5, we tested its role in ferritin uptake and found that Scara5 bound serum ferritin and then stimulated its endocytosis from the cell surface with consequent iron delivery. These data implicate cell type-specific mechanisms of iron traffic in organogenesis, which alternatively utilize transferrin or non-transferrin iron delivery pathways.


Assuntos
Ferritinas/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Ferro/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores da Transferrina/metabolismo , Receptores Depuradores Classe A/metabolismo , Transferrina/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Quimera/fisiologia , Endocitose/fisiologia , Proteínas de Ligação ao Ferro/genética , Rim/embriologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/fisiologia , Receptores de Superfície Celular/genética , Receptores da Transferrina/genética , Receptores Depuradores Classe A/genética
15.
Cell Metab ; 8(2): 132-45, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18680714

RESUMO

Parathyroid hormone (PTH) promotes bone catabolism by targeting bone marrow (BM) stromal cells (SCs) and their osteoblastic progeny. Here we show that a continuous infusion of PTH that mimics hyperparathyroidism fails to induce osteoclast formation, bone resorption, and cortical bone loss in mice lacking T cells. T cells provide proliferative and survival cues to SCs and sensitize SCs to PTH through CD40 ligand (CD40L), a surface molecule of activated T cells that induces CD40 signaling in SCs. As a result, deletion of T cells or T cell-expressed CD40L blunts the bone catabolic activity of PTH by decreasing bone marrow SC number, the receptor activator of nuclear factor-kappaB ligand (RANKL)/OSTEOPROTEGERN (OPG) ratio, and osteoclastogenic activity. Therefore, T cells play an essential permissive role in hyperparathyroidism as they influence SC proliferation, life span, and function through CD40L. T cell-SC crosstalk pathways may thus provide pharmacological targets for PTH-induced bone disease.


Assuntos
Osso e Ossos/metabolismo , Ligante de CD40/metabolismo , Hiperparatireoidismo/metabolismo , Osteoporose/metabolismo , Hormônio Paratireóideo/metabolismo , Linfócitos T/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Osso e Ossos/imunologia , Osso e Ossos/fisiopatologia , Ligante de CD40/imunologia , Proliferação de Células/efeitos dos fármacos , Feminino , Hiperparatireoidismo/induzido quimicamente , Hiperparatireoidismo/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteoclastos/metabolismo , Osteoporose/imunologia , Osteoporose/fisiopatologia , Hormônio Paratireóideo/imunologia , Hormônio Paratireóideo/farmacologia , Ligante RANK/imunologia , Ligante RANK/metabolismo , Transdução de Sinais/fisiologia , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Células Estromais/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
16.
J Am Soc Nephrol ; 18(2): 407-13, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17229907

RESUMO

Neutrophil gelatinase-associated lipocalin (NGAL) is expressed and secreted by immune cells, hepatocytes, and renal tubular cells in various pathologic states. NGAL exerts bacteriostatic effects, which are explained by its ability to capture and deplete siderophores, small iron-binding molecules that are synthesized by certain bacteria as a means of iron acquisition. Consistently, NGAL deficiency in genetically modified mice leads to an increased growth of bacteria. However, growing evidence suggests effects of the protein beyond fighting microorganisms. NGAL acts as a growth and differentiation factor in multiple cell types, including developing and mature renal epithelia, and some of this activity is enhanced in the presence of siderophore:iron complexes. This has led to the hypothesis that eukaryotes might synthesize siderophore-like molecules that bind NGAL. Accordingly, NGAL-mediated iron shuttling between the extracellular and intracellular spaces may explain some of the biologic activities of the protein. Interest in NGAL has been sparked by the observation that NGAL is massively upregulated after renal tubular injury and may participate in limiting kidney damage. This review summarizes the current knowledge about the dual effects of NGAL as a siderophore:iron-binding protein and as a growth factor and examines the role of these effects in renal injury.


Assuntos
Proteínas de Fase Aguda/fisiologia , Proteínas de Transporte/sangue , Ferro/metabolismo , Rim/lesões , Neutrófilos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas de Fase Aguda/biossíntese , Animais , Diferenciação Celular/fisiologia , Humanos , Rim/citologia , Rim/embriologia , Nefropatias/fisiopatologia , Túbulos Renais/fisiologia , Lipocalina-2 , Lipocalinas , Camundongos , Modelos Animais , Proteínas Proto-Oncogênicas/biossíntese , Transdução de Sinais
17.
Curr Opin Nephrol Hypertens ; 15(4): 442-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775460

RESUMO

PURPOSE OF REVIEW: Neutrophil gelatinase-associated lipocalin (NGAL) is a member of the lipocalin superfamily of carrier proteins. NGAL is the first known mammalian protein which specifically binds organic molecules called siderophores, which are high-affinity iron chelators. Here, we review the expression, siderophore-dependent biological activities and clinical significance of NGAL in epithelial development and in kidney disease. RECENT FINDINGS: NGAL expression is rapidly induced in the nephron in response to renal epithelial injury. This has led to the establishment of NGAL assays that detect renal damage in the human. Additionally, only when complexed with siderophore and iron as a trimer, NGAL induces mesenchymal-epithelial transition (or nephron formation) in embryonic kidney in vitro and protects adult kidney from ischemia-reperfusion injury in vivo. While the structure of the NGAL: siderophore: iron complex has thus far only been solved for bacterially synthesized siderophores, new evidence suggests the presence of mammalian siderophore-like molecules. SUMMARY: NGAL is rapidly and massively induced in renal epithelial injury and NGAL: siderophore: iron complexes may comprise a physiological renoprotective mechanism. The data have implications for the diagnosis and treatment of acute renal injury.


Assuntos
Proteínas de Fase Aguda/metabolismo , Ferro/metabolismo , Néfrons/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Traumatismo por Reperfusão/metabolismo , Sideróforos/metabolismo , Doença Aguda , Proteínas de Fase Aguda/genética , Animais , Transporte Biológico/genética , Epitélio/metabolismo , Epitélio/patologia , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Lipocalina-2 , Lipocalinas , Camundongos , Néfrons/lesões , Néfrons/patologia , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas/genética , Traumatismo por Reperfusão/genética , Sideróforos/genética
18.
J Am Soc Nephrol ; 16(7): 1993-2002, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15917337

RESUMO

Mammalian nephrogenesis depends on the interaction between the ureteric bud and the metanephric mesenchyme. As the ureteric bud undergoes branching and segmentation, the stalks differentiate into the collecting system of the mature kidney, while the tip cells interact with the adjacent cells of the metanephric mesenchyme, inducing their conversion into nephrons. This induction is mediated by secreted factors. For identifying novel mediators, the tips of the ureteric tree were isolated and microarray analyses were performed using manually refined, multistep gene ontology annotations. For identifying conserved factors, two databases were developed, one from mouse E12.5 and one from rat E13.5 ureteric buds. The overlap of mouse and rat data sets yielded 20 different transcripts that were enriched in the ureteric bud compared with metanephric mesenchyme and predicted to code for secreted proteins. Real-time reverse transcriptase-PCR and in situ hybridization confirmed these identifications. One of the genes that was highly specific to the ureteric bud tip was cytokine-like factor 1 (CLF-1). Recombinant CLF-1 in complex with its physiologic ligand, cardiotrophin-like cytokine (CLC), triggered phosphorylation of signal transducer and activator of transcription 3 in mesenchyme, a pathway characteristic of mesenchymal-to-epithelial conversion. Indeed, when applied to isolated rat metanephric mesenchyme, CLF-1/CLC (3 nM) induced mature nephron structures expressing glomerular and tubular markers. These results underline the power of this first comprehensive gene expression analysis of the ureteric bud tip to identify bioactive molecules.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Rim/embriologia , Receptores de Citocinas/fisiologia , Transativadores/fisiologia , Animais , Diferenciação Celular/genética , Citocinas/fisiologia , Bases de Dados Genéticas , Células Epiteliais/fisiologia , Perfilação da Expressão Gênica , Mesoderma/fisiologia , Camundongos , Organogênese/genética , Ratos , Receptores de Citocinas/genética , Fator de Transcrição STAT3 , Transdução de Sinais
19.
J Clin Invest ; 115(3): 610-21, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15711640

RESUMO

Neutrophil gelatinase-associated lipocalin (Ngal), also known as siderocalin, forms a complex with iron-binding siderophores (Ngal:siderophore:Fe). This complex converts renal progenitors into epithelial tubules. In this study, we tested the hypothesis that Ngal:siderophore:Fe protects adult kidney epithelial cells or accelerates their recovery from damage. Using a mouse model of severe renal failure, ischemia-reperfusion injury, we show that a single dose of Ngal (10 microg), introduced during the initial phase of the disease, dramatically protects the kidney and mitigates azotemia. Ngal activity depends on delivery of the protein and its siderophore to the proximal tubule. Iron must also be delivered, since blockade of the siderophore with gallium inhibits the rescue from ischemia. The Ngal:siderophore:Fe complex upregulates heme oxygenase-1, a protective enzyme, preserves proximal tubule N-cadherin, and inhibits cell death. Because mouse urine contains an Ngal-dependent siderophore-like activity, endogenous Ngal might also play a protective role. Indeed, Ngal is highly accumulated in the human kidney cortical tubules and in the blood and urine after nephrotoxic and ischemic injury. We reveal what we believe to be a novel pathway of iron traffic that is activated in human and mouse renal diseases, and it provides a unique method for their treatment.


Assuntos
Proteínas de Fase Aguda , Endocitose , Ferro/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas Oncogênicas , Traumatismo por Reperfusão , Sideróforos/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/uso terapêutico , Proteínas de Fase Aguda/urina , Animais , Creatinina/sangue , Células Epiteliais/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Humanos , Rim/citologia , Necrose do Córtex Renal/tratamento farmacológico , Necrose do Córtex Renal/metabolismo , Necrose do Córtex Renal/patologia , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Lipocalina-2 , Lipocalinas , Substâncias Macromoleculares , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/uso terapêutico , Proteínas Oncogênicas/urina , Proteínas Proto-Oncogênicas , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
20.
Am J Physiol Cell Physiol ; 287(6): C1547-59, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15282194

RESUMO

Intracellular iron regulates gene expression by inhibiting the interaction of iron regulatory proteins (IRPs) with RNA motifs called iron-responsive elements (IREs). To assay this interaction in living cells we have developed two fluorescent IRE-based reporters that rapidly, reversibly, and specifically respond to changes in cellular iron status as well as signaling that modifies IRP activity. The reporters were also sufficiently sensitive to distinguish apo- from holotransferrin in the medium, to detect the effect of modifiers of the transferrin pathway such as HFE, and to detect the donation or chelation of iron by siderophores bound to the lipocalin neutrophil gelatinase-associated lipocalin (Ngal). In addition, alternative configurations of the IRE motif either enhanced or repressed fluorescence, permitting a ratio analysis of the iron-dependent response. These characteristics make it possible to visualize iron-IRP-IRE interactions in vivo.


Assuntos
Genes Reporter/fisiologia , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/metabolismo , Ferro/farmacologia , Transdução de Sinais/fisiologia , Proteínas de Fase Aguda/metabolismo , Proteínas de Bactérias/genética , Linhagem Celular Transformada , Citoplasma/metabolismo , Citometria de Fluxo , Corantes Fluorescentes/metabolismo , Expressão Gênica , Humanos , Ferro/metabolismo , Proteína 1 Reguladora do Ferro/genética , Proteína 2 Reguladora do Ferro/genética , Lipocalina-2 , Lipocalinas , Proteínas Luminescentes/genética , Proteínas Oncogênicas/metabolismo , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA