Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33586672

ABSTRACT

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.


Subject(s)
Cell Movement/immunology , Intestines , Osteoporosis, Postmenopausal , Ovariectomy , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Female , Humans , Intestines/immunology , Intestines/microbiology , Mice , Mice, Knockout , Osteoporosis, Postmenopausal/immunology , Osteoporosis, Postmenopausal/microbiology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Tumor Necrosis Factor-alpha/genetics
2.
Elife ; 102021 01 12.
Article in English | MEDLINE | ID: mdl-33432923

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Skeleton/growth & development , Animals , Fecal Microbiota Transplantation , Feces/microbiology , Female , Mice
3.
Nat Commun ; 11(1): 468, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980603

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome/immunology , Osteoporosis/etiology , Parathyroid Hormone/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Animals , Fecal Microbiota Transplantation , Female , Germ-Free Life , Gram-Positive Endospore-Forming Rods/immunology , Hyperparathyroidism, Primary/complications , Hyperparathyroidism, Primary/immunology , Hyperparathyroidism, Primary/microbiology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoporosis/immunology , Osteoporosis/microbiology , Tumor Necrosis Factor-alpha/immunology
4.
J Clin Invest ; 130(4): 1767-1781, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917685

ABSTRACT

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.


Subject(s)
Butyrates/metabolism , Gastrointestinal Microbiome , Osteogenesis , Parathyroid Hormone/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Mice , Mice, Knockout , Parathyroid Hormone/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes, Regulatory/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
5.
J Bone Miner Res ; 34(2): 349-360, 2019 02.
Article in English | MEDLINE | ID: mdl-30399207

ABSTRACT

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.


Subject(s)
Bone Resorption/metabolism , Osteocytes/metabolism , Parathyroid Hormone/metabolism , RANK Ligand/biosynthesis , Receptors, Interleukin-17/metabolism , Signal Transduction , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Hyperparathyroidism, Primary/genetics , Hyperparathyroidism, Primary/metabolism , Hyperparathyroidism, Primary/pathology , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Knockout , Osteocytes/pathology , Parathyroid Hormone/genetics , RANK Ligand/genetics , Receptors, Interleukin-17/genetics
6.
Immunity ; 49(6): 1116-1131.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30446387

ABSTRACT

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.


Subject(s)
Butyrates/pharmacology , Osteogenesis/drug effects , T-Lymphocytes, Regulatory/metabolism , Wnt Proteins/metabolism , Animals , Butyrates/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication , Cell Proliferation/drug effects , Female , Lacticaseibacillus rhamnosus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Probiotics/administration & dosage , Probiotics/metabolism , T-Lymphocytes, Regulatory/cytology , Wnt Proteins/genetics
7.
EMBO Rep ; 19(1): 156-171, 2018 01.
Article in English | MEDLINE | ID: mdl-29158349

ABSTRACT

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.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Calcium-Regulating Hormones and Agents/therapeutic use , Osteoporosis, Postmenopausal/drug therapy , T-Lymphocytes, Regulatory/drug effects , Teriparatide/therapeutic use , Aged , Animals , Biomarkers/metabolism , Calcium/therapeutic use , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Lymphocyte Count , Mice , Osteocalcin/genetics , Osteocalcin/metabolism , Osteoporosis, Postmenopausal/genetics , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/pathology , Ovariectomy , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Treatment Outcome , Vitamin D/analogs & derivatives , Vitamin D/therapeutic use
8.
J Clin Invest ; 126(6): 2049-63, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27111232

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome/physiology , Gonadal Steroid Hormones/deficiency , Osteoporosis, Postmenopausal/etiology , Osteoporosis, Postmenopausal/prevention & control , Probiotics/pharmacology , Animals , Bone Remodeling , Digestive System Physiological Phenomena , Disease Models, Animal , Female , Germ-Free Life , Humans , Interleukin-17/metabolism , Lacticaseibacillus rhamnosus , Mice , Mice, Inbred C57BL , Osteoporosis, Postmenopausal/pathology , Permeability , RANK Ligand/metabolism , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
J Bone Miner Res ; 31(5): 949-63, 2016 05.
Article in English | MEDLINE | ID: mdl-26614970

ABSTRACT

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.


Subject(s)
Estrogens/deficiency , Hydrogen Sulfide/metabolism , Osteogenesis , Osteoporosis, Postmenopausal/metabolism , Wnt Signaling Pathway , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Female , Humans , Mice , Osteoporosis, Postmenopausal/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Wnt Proteins/metabolism
10.
Cell Metab ; 22(5): 799-810, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26456334

ABSTRACT

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.


Subject(s)
Bone Diseases, Metabolic/metabolism , Hyperparathyroidism, Primary/metabolism , Interleukin-17/metabolism , Animals , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/pathology , Calcium Channel Blockers/therapeutic use , Humans , Hyperparathyroidism, Primary/complications , Hyperparathyroidism, Primary/drug therapy , Hyperparathyroidism, Primary/pathology , Interleukin-17/biosynthesis , Mice , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/biosynthesis
11.
J Bone Miner Res ; 30(4): 695-705, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25359628

ABSTRACT

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.


Subject(s)
Anabolic Agents/pharmacology , Bone and Bones/drug effects , CD40 Ligand/immunology , Parathyroid Hormone/pharmacology , T-Lymphocytes/immunology , Anabolic Agents/administration & dosage , Animals , CD40 Ligand/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Parathyroid Hormone/administration & dosage
12.
J Bone Miner Res ; 29(1): 43-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24357520

ABSTRACT

Both blunted osteocytic production of the Wnt inhibitor sclerostin (Scl) and increased T-cell production of the Wnt ligand Wnt10b contribute to the bone anabolic activity of intermittent parathyroid hormone (iPTH) treatment. However, the relative contribution of these mechanisms is unknown. In this study, we modeled the repressive effects of iPTH on Scl production in mice by treatment with a neutralizing anti-Scl antibody (Scl-Ab) to determine the contribution of T-cell-produced Wnt10b to the Scl-independent modalities of action of iPTH. We report that combined treatment with Scl-Ab and iPTH was more potent than either iPTH or Scl-Ab alone in increasing stromal cell production of OPG, osteoblastogenesis, osteoblast life span, bone turnover, bone mineral density, and trabecular bone volume and structure in mice with T cells capable of producing Wnt10b. In T-cell-null mice and mice lacking T-cell production of Wnt10b, combined treatment increased bone turnover significantly more than iPTH or Scl-Ab alone. However, in these mice, combined treatment with Scl-Ab and iPTH was equally effective as Scl-Ab alone in increasing the osteoblastic pool, bone volume, density, and structure. These findings demonstrate that the Scl-independent activity of iPTH on osteoblasts and bone mass is mediated by T-cell-produced Wnt10b. The data provide a proof of concept of a more potent therapeutic effect of combined treatment with iPTH and Scl-Ab than either alone.


Subject(s)
Glycoproteins/antagonists & inhibitors , Parathyroid Hormone/administration & dosage , Wnt Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Antibodies/pharmacology , Bone Density/drug effects , Bone and Bones/drug effects , Female , Glycoproteins/biosynthesis , Glycoproteins/immunology , Intercellular Signaling Peptides and Proteins , Mice , Osteoblasts/drug effects , Osteogenesis/drug effects , T-Lymphocytes/metabolism
13.
Blood ; 122(14): 2346-57, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23954891

ABSTRACT

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.


Subject(s)
CD40 Ligand/biosynthesis , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Ovariectomy , T-Lymphocytes/metabolism , Wnt Proteins/biosynthesis , Animals , Bone Marrow Transplantation/immunology , Female , Flow Cytometry , Hematopoietic Stem Cells/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology , Wnt Proteins/immunology
14.
Blood ; 120(22): 4352-62, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22955916

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Hematopoietic Stem Cells/drug effects , Parathyroid Hormone/pharmacology , T-Lymphocytes/physiology , Animals , Bone Marrow Transplantation , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Female , Genes, T-Cell Receptor beta/physiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/drug effects , Time Factors , Wnt Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 109(12): E725-33, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22393015

ABSTRACT

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.


Subject(s)
Bone and Bones/metabolism , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Animals , Bone Density , Female , Gene Silencing , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Osteoblasts/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , X-Ray Microtomography/methods
16.
Proc Natl Acad Sci U S A ; 108(2): 768-73, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187391

ABSTRACT

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.


Subject(s)
CD40 Ligand/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , T-Lymphocytes/cytology , Animals , Coculture Techniques , Estrogens/metabolism , Humans , Ligands , Mice , NF-kappa B/metabolism , Osteoporosis/metabolism , Osteoprotegerin/metabolism , Ovariectomy/methods , Tumor Necrosis Factor-alpha/metabolism
17.
PLoS One ; 5(8): e12290, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20808842

ABSTRACT

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.


Subject(s)
Bone Resorption/chemically induced , Bone Resorption/metabolism , Gene Silencing , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/deficiency , Receptor, Parathyroid Hormone, Type 1/genetics , T-Lymphocytes/metabolism , Animals , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Female , Humans , Male , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
18.
Ann N Y Acad Sci ; 1192: 215-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392239

ABSTRACT

T cells are required for continuous parathyroid hormone (cPTH) treatment to induce bone loss as they sensitize stromal cells to PTH through CD40 ligand (CD40L), a surface molecule of activated T cells. Since CD40L expression is a feature of activated T cells, we investigated whether antigen (Ag)-mediated T cell activation is required for PTH to exert its catabolic activity. We report that inhibition of Ag presentation through silencing of either class I or class II MHC-T cell receptor (TCR) interaction prevents the cortical bone loss induced by in vivo cPTH treatment. We also show that the bone loss and the stimulation of bone resorption induced by cPTH treatment are prevented by CTLA4-Ig, an inhibitor of T cell costimulation approved for the treatment of rheumatoid arthritis. Since inhibition of antigen-driven T cell activation by blockade of either TCR signaling or T cell costimulation is sufficient to silence the catabolic activity of cPTH, antigen-presenting cells and T lymphocyte interactions therefore play a critical role in the mechanism of action of PTH.


Subject(s)
Antigen Presentation/drug effects , Bone Resorption/chemically induced , Bone Resorption/prevention & control , Immunoconjugates/pharmacology , Lymphocyte Activation/drug effects , Parathyroid Hormone/adverse effects , T-Lymphocytes/drug effects , Abatacept , Animals , Antigen Presentation/genetics , Antigen Presentation/physiology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Female , Immunoconjugates/therapeutic use , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/immunology
19.
Cell Metab ; 10(3): 229-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723499

ABSTRACT

Intermittent administration of parathyroid hormone (iPTH) is used to treat osteoporosis because it improves bone architecture and strength, but the underlying cellular and molecular mechanisms are unclear. Here, we show that iPTH increases the production of Wnt10b by bone marrow CD8+ T cells and induces these lymphocytes to activate canonical Wnt signaling in preosteoblasts. Accordingly, in responses to iPTH, T cell null mice display diminished Wnt signaling in preosteoblasts and blunted osteoblastic commitment, proliferation, differentiation, and life span, which result in decreased trabecular bone anabolism and no increase in strength. Demonstrating the specific role of lymphocytic Wnt10b, iPTH has no anabolic activity in mice lacking T-cell-produced Wnt10b. Therefore, T-cell-mediated activation of Wnt signaling in osteoblastic cells plays a key permissive role in the mechanism by which iPTH increases bone strength, suggesting that T cell osteoblast crosstalk pathways may provide pharmacological targets for bone anabolism.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Parathyroid Hormone/pharmacology , Wnt Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction
20.
Dev Cell ; 16(1): 35-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19154717

ABSTRACT

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.


Subject(s)
Ferritins/metabolism , Iron-Binding Proteins/metabolism , Iron/metabolism , Receptors, Cell Surface/metabolism , Receptors, Transferrin/metabolism , Scavenger Receptors, Class A/metabolism , Transferrin/metabolism , Animals , Biological Transport , Cell Line , Chimera/physiology , Endocytosis/physiology , Iron-Binding Proteins/genetics , Kidney/embryology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/physiology , Receptors, Cell Surface/genetics , Receptors, Transferrin/genetics , Scavenger Receptors, Class A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...