Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 11(4): e1005160, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25859855

RESUMO

RECQL4 mutations are associated with Rothmund Thomson Syndrome (RTS), RAPADILINO Syndrome and Baller-Gerold Syndrome. These patients display a range of benign skeletal abnormalities such as low bone mass. In addition, RTS patients have a highly increased incidence of osteosarcoma (OS). The role of RECQL4 in normal adult bone development and homeostasis is largely uncharacterized and how mutation of RECQL4 contributes to OS susceptibility is not known. We hypothesised that Recql4 was required for normal skeletal development and both benign and malignant osteoblast function, which we have tested in the mouse. Recql4 deletion in vivo at the osteoblastic progenitor stage of differentiation resulted in mice with shorter bones and reduced bone volume, assessed at 9 weeks of age. This was associated with an osteoblast intrinsic decrease in mineral apposition rate and bone formation rate in the Recql4-deficient cohorts. Deletion of Recql4 in mature osteoblasts/osteocytes in vivo, however, did not cause a detectable phenotype. Acute deletion of Recql4 in primary osteoblasts or shRNA knockdown in an osteoblastic cell line caused failed proliferation, accompanied by cell cycle arrest, induction of apoptosis and impaired differentiation. When cohorts of animals were aged long term, the loss of Recql4 alone was not sufficient to initiate OS. We then crossed the Recql4fl/fl allele to a fully penetrant OS model (Osx-Cre p53fl/fl). Unexpectedly, the Osx-Cre p53fl/flRecql4fl/fl (dKO) animals had a significantly increased OS-free survival compared to Osx-Cre p53fl/fl or Osx-Cre p53fl/flRecql4fl/+ (het) animals. The extended survival was explained when the Recql4 status in the tumors that arose was assessed, and in no case was there complete deletion of Recql4 in the dKO OS. These data provide a mechanism for the benign skeletal phenotypes of RECQL4 mutation syndromes. We propose that tumor suppression and osteosarcoma susceptibility are most likely a function of mutant, not null, alleles of RECQL4.


Assuntos
Neoplasias Ósseas/genética , Osteoblastos/metabolismo , Osteogênese , Osteossarcoma/genética , RecQ Helicases/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Proliferação de Células , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Osteossarcoma/metabolismo , RecQ Helicases/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
2.
EMBO Rep ; 16(6): 753-68, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25916856

RESUMO

T-cell-dependent antigenic stimulation drives the differentiation of B cells into antibody-secreting plasma cells and memory B cells, but how B cells regulate this process is unclear. We show that LKB1 expression in B cells maintains B-cell quiescence and prevents the premature formation of germinal centers (GCs). Lkb1-deficient B cells (BKO) undergo spontaneous B-cell activation and secretion of multiple inflammatory cytokines, which leads to splenomegaly caused by an unexpected expansion of T cells. Within this cytokine response, increased IL-6 production results from heightened activation of NF-κB, which is suppressed by active LKB1. Secreted IL-6 drives T-cell activation and IL-21 production, promoting T follicular helper (TFH ) cell differentiation and expansion to support a ~100-fold increase in steady-state GC B cells. Blockade of IL-6 secretion by BKO B cells inhibits IL-21 expression, a known inducer of TFH -cell differentiation and expansion. Together, these data reveal cell intrinsic and surprising cell extrinsic roles for LKB1 in B cells that control TFH -cell differentiation and GC formation, and place LKB1 as a central regulator of T-cell-dependent humoral immunity.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Centro Germinativo/fisiologia , Ativação Linfocitária , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas Quinases Ativadas por AMP , Animais , Diferenciação Celular , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucinas/imunologia , Camundongos , NF-kappa B/genética , Linfócitos T Auxiliares-Indutores/fisiologia
3.
Proc Natl Acad Sci U S A ; 111(16): 6040-5, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24733914

RESUMO

Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.


Assuntos
Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Artrite/virologia , Reabsorção Óssea/patologia , Reabsorção Óssea/virologia , Osteoblastos/patologia , Ross River virus/fisiologia , Fosfatase Ácida/sangue , Adulto , Infecções por Alphavirus/sangue , Animais , Anticorpos Neutralizantes/farmacologia , Artrite/sangue , Artrite/patologia , Reabsorção Óssea/sangue , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Osso e Ossos/virologia , Feminino , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/patologia , Lâmina de Crescimento/virologia , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Isoenzimas/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Osteoblastos/efeitos dos fármacos , Osteoblastos/virologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteoclastos/virologia , Osteogênese/efeitos dos fármacos , Osteoprotegerina/metabolismo , Fenótipo , Ligante RANK/metabolismo , Ross River virus/efeitos dos fármacos , Líquido Sinovial/metabolismo , Fosfatase Ácida Resistente a Tartarato , Replicação Viral/efeitos dos fármacos , Microtomografia por Raio-X
4.
J Virol ; 89(1): 581-93, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25339772

RESUMO

UNLABELLED: The recent global resurgence of arthritogenic alphaviruses, in particular chikungunya virus (CHIKV), highlights an urgent need for the development of therapeutic intervention strategies. While there has been significant progress in defining the pathophysiology of alphaviral disease, relatively little is known about the mechanisms involved in CHIKV-induced arthritis or potential therapeutic options to treat the severe arthritic symptoms associated with infection. Here, we used microcomputed tomographic (µCT) and histomorphometric analyses to provide previously undescribed evidence of reduced bone volume in the proximal tibial epiphysis of CHIKV-infected mice compared to the results for mock controls. This was associated with a significant increase in the receptor activator of nuclear factor-κB ligand/osteoprotegerin (RANKL/OPG) ratio in infected murine joints and in the serum of CHIKV patients. The expression levels of the monocyte chemoattractant proteins (MCPs), including MCP-1/CCL2, MCP-2/CCL8, and MCP-3/CCL7, were also highly elevated in joints of CHIKV-infected mice, accompanied by increased cellularity within the bone marrow in tibial epiphysis and ankle joints. Both this effect and CHIKV-induced bone loss were significantly reduced by treatment with the MCP inhibitor bindarit. Collectively, these findings demonstrate a unique role for MCPs in promoting CHIKV-induced osteoclastogenesis and bone loss during disease and suggest that inhibition of MCPs with bindarit may be an effective therapy for patients affected with alphavirus-induced bone loss. IMPORTANCE: Arthritogenic alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), cause worldwide outbreaks of polyarthritis, which can persist in patients for months following infection. Previous studies have shown that host proinflammatory soluble factors are associated with CHIKV disease severity. Furthermore, it is established that chemokine (C-C motif) ligand 2 (CCL2/MCP-1) is important in cellular recruitment and inducing bone-resorbing osteoclast (OC) formation. Here, we show that CHIKV replicates in bone and triggers bone loss by increasing the RANKL/OPG ratio. CHIKV infection results in MCP-induced cellular infiltration in the inflamed joints, and bone loss can be ameliorated by treatment with an MCP-inhibiting drug, bindarit. Taken together, our data reveal a previously undescribed role for MCPs in CHIKV-induced bone loss: one of recruiting monocytes/OC precursors to joint sites and thereby favoring a pro-osteoclastic microenvironment. This suggests that bindarit may be an effective treatment for alphavirus-induced bone loss and arthritis in humans.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Reabsorção Óssea/prevenção & controle , Quimiocina CCL2/antagonistas & inibidores , Febre de Chikungunya/complicações , Indazóis/administração & dosagem , Propionatos/administração & dosagem , Adulto , Idoso , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
5.
Cytokine ; 68(2): 101-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24767864

RESUMO

OBJECTIVE: To identify how the gp130-signaling cytokine oncostatin M (OSM), acting alone or in concert with IL-1ß or TNFα, affects synovial fibroblast expression of genes relevant to inflammation and bone erosion in inflammatory arthritis. METHODS: Synovial fibroblasts (SFs) were isolated from non-arthritic wild type (WT) or OSM receptor deficient (OSMR(-/-)) mice and stimulated with OSM, IL-1ß or TNFα and their combinations. Cytokine gene expression was assessed by quantitative RT-PCR. ELISA, flow cytometry and immunohistochemistry identified protein expression. Gene expression patterns were confirmed in SFs isolated from patients with osteoarthritis (OASFs) and rheumatoid arthritis (RASFs). RESULTS: Expression of OSM and its receptors, gp130, OSMR and LIFR, was increased in synovial tissue from the mouse antigen-induced arthritis model. In isolated WT mouse synovial fibroblasts OSM alone, or in synergy with IL-1ß, or together with TNFα, potently induced expression of the pro-inflammatory cytokine IL-6. OSM also induced a sustained increase in mRNA levels of the pro-osteoclastic cytokine RANKL. Combining OSM with IL-1ß, but not with TNFα, further increased RANKL expression. Importantly these effects of OSM were all dependent on the expression of OSMR. Furthermore, OSM also increased expression of its own receptors, gp130 and OSMR and the IL-1 receptor, IL1-R1; the latter effects were also observed in both human OASFs and RASFs. CONCLUSION: Together our data suggests that OSM signaling via OSMR in SFs has the potential to contribute significantly to joint destruction in inflammatory arthritis. It not only induces expression of pro-inflammatory and pro-osteoclastic cytokines but can also augment its own actions and that of IL-1 by inducing expression of OSMR and IL-1R1.


Assuntos
Fibroblastos/metabolismo , Interleucina-1beta/metabolismo , Oncostatina M/metabolismo , Receptores de Oncostatina M/metabolismo , Membrana Sinovial/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Regulação da Expressão Gênica , Humanos , Interleucina-1beta/genética , Camundongos Endogâmicos C57BL , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Oncostatina M/deficiência
6.
Proc Natl Acad Sci U S A ; 108(7): 2873-8, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282644

RESUMO

We utilized gene expression profiling of a comprehensive panel of purified developmentally defined normal murine B cells to identify unique transcriptional signatures for each subset. To elucidate transcription factor activities that function in a stage-specific fashion, we used gene sets that share transcription factor targets and found that germinal center B cells had a robust enrichment of up-regulated and down-regulated signatures compared with the other B-cell subsets. Notably, we found Yy1 and its targets to be central regulators of the germinal center B (GCB)-specific transcriptional program with binding of Yy1 to select signature genes in GCB cells, and translation of the Yy1 signatures to human GCB cells. We then tested whether our newly generated, stage-specific transcriptional signatures could be used to link murine lymphoma models to stages of normal B-cell development. Although each of the molecularly defined murine lymphoma models conserved certain stage-specific features of normal B-cell development, there was a significant alteration of the normal differentiation signature following malignant transformation. These findings offer important tools and insights for elucidating differences between normal and malignant B cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Regulação da Expressão Gênica/imunologia , Centro Germinativo/metabolismo , Linfoma/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Subpopulações de Linfócitos B/citologia , Diferenciação Celular/imunologia , Imunoprecipitação da Cromatina , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Camundongos
7.
Immunol Rev ; 233(1): 301-12, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20193007

RESUMO

The past decade has observed an explosion of new information regarding the impact of inflammation on bone. In rheumatic diseases, several factors that act as both immune modulators and regulators of bone homeostasis have been shown to mediate an imbalance in bone resorption and bone formation resulting in joint degeneration. In rheumatoid arthritis (RA), focal bone loss is due to excess bone resorption by osteoclasts. Resorption is mediated in part by increased local expression of the cytokine receptor activator of nuclear factor-kappaB ligand (RANKL) compared with expression of its decoy receptor osteoprotegerin (OPG). Bone formation by osteoblasts is also impaired at erosion sites in RA, and inhibitors of the canonical Wingless (Wnt) signaling pathway, including DKK1, have been implicated in the suppression of normal osteoblast function at these sites. Inhibition of DKK1 in an animal model of RA attenuated bone erosion by increasing OPG expression as well as promoting bone formation. In contrast to RA, inflammation in the spondyloarthropathies often results in excess periosteal bone formation, highlighting that the net impact of inflammation on bone is specific to the site at which inflammation occurs, and the cell types, cytokines, and factors present within the local bone microenvironment. This fertile area of research bears watching for the identification of novel targets for the prevention of abnormal bone remodeling in inflammatory diseases.


Assuntos
Artrite Reumatoide/fisiopatologia , Remodelação Óssea , Articulações/fisiopatologia , Espondiloartropatias/fisiopatologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/imunologia , Remodelação Óssea/imunologia , Citocinas/imunologia , Humanos , Mediadores da Inflamação/imunologia , Articulações/imunologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Transdução de Sinais , Espondiloartropatias/imunologia , Proteínas Wnt/imunologia
8.
Arthritis Rheum ; 64(5): 1540-50, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22139865

RESUMO

OBJECTIVE: Inflammation in the bone microenvironment stimulates osteoclast differentiation, resulting in uncoupling of resorption and formation. Mechanisms contributing to the inhibition of osteoblast function in inflammatory diseases, however, have not been elucidated. Rheumatoid arthritis (RA) is a prototype of an inflammatory arthritis that results in focal loss of articular bone. The paucity of bone repair in inflammatory diseases such as RA raises compelling questions regarding the impact of inflammation on bone formation. The aim of this study was to establish the mechanisms by which inflammation regulates osteoblast activity. METHODS: We characterized an innovative variant of a murine model of arthritis in which inflammation is induced in C57BL/6J mice by transfer of arthritogenic K/BxN serum and allowed to resolve. RESULTS: In the setting of resolving inflammation, bone resorption ceased and appositional osteoblast-mediated bone formation was induced, resulting in repair of eroded bone. Resolution of inflammation was accompanied by striking changes in the expression of regulators of the Wnt/ß-catenin pathway, which is critical for osteoblast differentiation and function. Down-regulation of the Wnt antagonists secreted frizzled-related protein 1 (sFRP1) and sFRP2 during the resolution phase paralleled induction of the anabolic and pro-matrix mineralization factors Wnt10b and DKK2, demonstrating the role of inflammation in regulating Wnt signaling. CONCLUSION: Repair of articular bone erosion occurs in the setting of resolving inflammation, accompanied by alterations in the Wnt signaling pathway. These data imply that in inflammatory diseases that result in persistent articular bone loss, strict control of inflammation may not be achieved and may be essential for the generation of an anabolic microenvironment that supports bone formation and repair.


Assuntos
Artrite Experimental/metabolismo , Inflamação/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Proteínas Wnt/biossíntese , Via de Sinalização Wnt , Fosfatase Ácida/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Regeneração Óssea/fisiologia , Proteínas Relacionadas à Folistatina/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Isoenzimas/metabolismo , Articulações/metabolismo , Articulações/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Fosfatase Ácida Resistente a Tartarato
9.
Curr Osteoporos Rep ; 10(2): 109-17, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22427140

RESUMO

Intercellular communication within the bone microenvironment is critical for the maintenance of normal bone structure. Osteoblast-lineage cells at all stages of differentiation, from pluripotent precursors to matrix-embedded osteocytes, produce regulatory factors that modulate the differentiation and activity of both bone-forming osteoblasts and bone-resorbing osteoclasts. Osteoclasts can also release factors that feed back to regulate osteoblast activity. Intercellular cross-talk within the bone microenvironment is not restricted only to these bone cells. Other cells within the bone marrow microenvironment, including adipocytes, T cells, and macrophages, play key roles that influence the processes of bone formation and resorption. This review discusses recent work that provides new insights into some of these communication networks and the factors involved, including osteocytic production of receptor activator of nuclear factor-κB ligand (RANKL) and sclerostin, osteoblastic production of interleukin-33, osteoclast-derived Semaphorin 4D, ephrin signaling, and signals from T helper cells and resident osteal macrophages (osteomacs).


Assuntos
Osso e Ossos/citologia , Osso e Ossos/fisiologia , Comunicação Celular/fisiologia , Transdução de Sinais/fisiologia , Animais , Humanos , Modelos Animais , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteoclastos/citologia , Osteoclastos/fisiologia , Ligante RANK/fisiologia , Linfócitos T/citologia , Linfócitos T/fisiologia
11.
Annu Rev Pathol ; 12: 187-215, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-27959627

RESUMO

Immunodeficient mice engrafted with functional human cells and tissues, that is, humanized mice, have become increasingly important as small, preclinical animal models for the study of human diseases. Since the description of immunodeficient mice bearing mutations in the IL2 receptor common gamma chain (IL2rgnull) in the early 2000s, investigators have been able to engraft murine recipients with human hematopoietic stem cells that develop into functional human immune systems. These mice can also be engrafted with human tissues such as islets, liver, skin, and most solid and hematologic cancers. Humanized mice are permitting significant progress in studies of human infectious disease, cancer, regenerative medicine, graft-versus-host disease, allergies, and immunity. Ultimately, use of humanized mice may lead to the implementation of truly personalized medicine in the clinic. This review discusses recent progress in the development and use of humanized mice and highlights their utility for the study of human diseases.


Assuntos
Doenças Transmissíveis/terapia , Modelos Animais de Doenças , Sistema Imunitário/imunologia , Animais , Doenças Transmissíveis/imunologia , Humanos , Camundongos , Camundongos SCID
12.
PLoS One ; 12(6): e0178641, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28605395

RESUMO

BACKGROUND/AIMS: Studies of human cadaveric pancreas specimens indicate that pancreas inflammation plays an important role in type 1 diabetes pathogenesis. Due to the inaccessibility of pancreas in living patients, imaging technology to visualize pancreas inflammation is much in need. In this study, we investigated the feasibility of utilizing ultrasound imaging to assess pancreas inflammation longitudinally in living rats during the progression leading to type 1 diabetes onset. METHODS: The virus-inducible BBDR type 1 diabetes rat model was used to systematically investigate pancreas changes that occur prior to and during development of autoimmunity. The nearly 100% diabetes incidence upon virus induction and the highly consistent time course of this rat model make longitudinal imaging examination possible. A combination of histology, immunoblotting, flow cytometry, and ultrasound imaging technology was used to identify stage-specific pancreas changes. RESULTS: Our histology data indicated that exocrine pancreas tissue of the diabetes-induced rats underwent dramatic changes, including blood vessel dilation and increased CD8+ cell infiltration, at a very early stage of disease initiation. Ultrasound imaging data revealed significant acute and persistent pancreas inflammation in the diabetes-induced rats. The pancreas micro-vasculature was significantly dilated one day after diabetes induction, and large blood vessel (superior mesenteric artery in this study) dilation and inflammation occurred several days later, but still prior to any observable autoimmune cell infiltration of the pancreatic islets. CONCLUSIONS: Our data demonstrate that ultrasound imaging technology can detect pancreas inflammation in living rats during the development of type 1 diabetes. Due to ultrasound's established use as a non-invasive diagnostic tool, it may prove useful in a clinical setting for type 1 diabetes risk prediction prior to autoimmunity and to assess the effectiveness of potential therapeutics.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Diabetes Mellitus Tipo 1/patologia , Pancreatite/diagnóstico por imagem , Pancreatite/patologia , Ultrassonografia , Animais , Apoptose , Resistência Capilar , Caspase 3/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/etiologia , Modelos Animais de Doenças , Humanos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Microvasos , Pâncreas/irrigação sanguínea , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/complicações , Pancreatite/metabolismo , Prognóstico , Ratos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Ultrassonografia/métodos
13.
Gene ; 372: 92-102, 2006 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-16513293

RESUMO

The transcription factor NFATc1 plays an essential role in transducing signals from RANKL in osteoclast differentiation. To date, however, the specific transcriptional targets of NFATc1 are unknown. Expression of the beta3 integrin is required for normal osteoclast function. We therefore examined the role of NFATc1 in human beta3 integrin expression in osteoclast differentiation. Analysis of the mouse and human beta3 gene promoters revealed considerable sequence homology across a 1.3 kb region upstream of the transcription start site (TSS), with conserved NFAT binding elements present. The region -1242 to +29 (relative to the TSS) was cloned as a luciferase reporter construct (pB3-1.3) and a deletion construct removing to -997 (pB3-1) made. The deletion of 245 bp 5' removed three conserved NFAT sites including a consensus NFAT:AP-1 site. The pB3-1.3 reporter construct was induced by treatment with RANKL in the range 2.5-40 ng/ml and dose-dependently induced by co-transfection with human NFATc1 in RAW264.7 cells. The pB3-1 deletion construct was minimally induced with RANKL treatment and unresponsive to co-transfected NFATc1. Direct NFAT binding to two of the consensus NFAT sites within this 245 bp 5' region was demonstrated by EMSA and supershift with anti-NFAT antibodies. Mutation of two of the conserved NFAT sites in the -1242 to -997 fragment was required to prevent binding. The double NFAT mutant, in the context of the full-length promoter was unresponsive to RANKL treatment or co-transfected NFATc1. We generated cell-permeable TAT-dominant-negative (dn)NFATc1 fusion proteins to assess the effect of blockade of NFAT signaling. Transduction with dnNFAT inhibited RANKL induction of the human beta3 integrin promoter. Involvement of the NFATc1-calcineurin pathway in regulating the human beta3 integrin promoter was further confirmed using the calcineurin pathway inhibitory peptide 11R-VIVIT. Together these results establish the beta3 gene as a direct target of NFATc1 in RANKL-dependent osteoclast formation.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Integrina beta3/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Regiões Promotoras Genéticas/genética , Animais , Sítios de Ligação/genética , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Permeabilidade da Membrana Celular , Sequência Conservada/genética , Relação Dose-Resposta a Droga , Produtos do Gene tat , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Camundongos , Mutação/genética , Oligopeptídeos/farmacologia , Ligação Proteica , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Elementos de Resposta/genética , Ativação Transcricional/efeitos dos fármacos
14.
J Mech Behav Biomed Mater ; 61: 96-109, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26851527

RESUMO

Isotropic hyperelastic models have been used to determine the material properties of normal human cartilage, but there remains an incomplete understanding of how these properties may be altered by osteoarthritis. The aims of this study were to (1) measure the material constants of normal and osteoarthritic human knee cartilage using isotropic hyperelastic models; (2) determine whether the material constants correlate with histological measures of structure and/or cartilage tissue damage; and (3) quantify the abilities of two common isotropic hyperelastic material models, the neo-Hookean and Yeoh models, to describe articular cartilage contact force, area, and pressure. Small osteochondral specimens of normal and osteoarthritic condition were retrieved from human cadaveric knees and from the knees of patients undergoing total knee arthroplasty and tested in unconfined compression at loading rates and large strains representative of weight-bearing activity. Articular surface contact area and lateral deformation were measured concurrently and specimen-specific finite element models then were used to determine the hyperelastic material constants. Structural parameters were measured using histological techniques while the severity of cartilage damage was quantified using the OARSI grading scale. The hyperelastic material constants correlated significantly with OARSI grade, indicating that the mechanical properties of cartilage for large strains change with tissue damage. The measurements of contact area described anisotropy of the tissue constituting the superficial zone. The Yeoh model described contact force and pressure more accurately than the neo-Hookean model, whereas both models under-predicted contact area and poorly described the anisotropy of cartilage within the superficial zone. These results identify the limits by which isotropic hyperelastic material models may be used to describe cartilage contact variables. This study provides novel data for the mechanical properties of normal and osteoarthritic human articular cartilage and enhances our ability to model this tissue using simple isotropic hyperelastic materials.


Assuntos
Cartilagem Articular/fisiologia , Cartilagem Articular/fisiopatologia , Osteoartrite/fisiopatologia , Anisotropia , Fenômenos Biomecânicos , Humanos , Modelos Biológicos , Estresse Mecânico , Suporte de Carga
15.
Sci Rep ; 6: 34491, 2016 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-27682431

RESUMO

Concomitant traumatic brain injury (TBI) and long bone fracture are commonly observed in multitrauma and polytrauma. Despite clinical observations of enhanced bone healing in patients with TBI, the relationship between TBI and fracture healing remains poorly understood, with clinical data limited by the presence of several confounding variables. Here we developed a novel trauma model featuring closed-skull weight-drop TBI and concomitant tibial fracture in order to investigate the effect of TBI on fracture healing. Male mice were assigned into Fracture + Sham TBI (FX) or Fracture + TBI (MULTI) groups and sacrificed at 21 and 35 days post-injury for analysis of healing fractures by micro computed tomography (µCT) and histomorphometry. µCT analysis revealed calluses from MULTI mice had a greater bone and total tissue volume, and displayed higher mean polar moment of inertia when compared to calluses from FX mice at 21 days post-injury. Histomorphometric results demonstrated an increased amount of trabecular bone in MULTI calluses at 21 days post-injury. These findings indicate that closed head TBI results in calluses that are larger in size and have an increased bone volume, which is consistent with the notion that TBI induces the formation of a more robust callus.

16.
Bone ; 72: 34-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25460578

RESUMO

To define their gene expression and function, osteocytes are commonly isolated and purified by fluorescence-activated cell sorting (FACS) from mice expressing GFP directed by the dentin matrix protein 1 (Dmp1) promoter (DMP1-GFP). These cells express mRNA for osteocyte genes, including sclerostin (Sost) and Dmp1, and genes associated with the osteoclast phenotype: Dcstamp, Oscar, Cathepsin K (Ctsk), tartrate resistant acid phosphatase (TRAP/Acp5) and calcitonin receptor (Calcr). This suggests either that osteoclasts and osteocytes share genes and functions or that DMP1-GFP(+) preparations contain haematopoietic osteoclasts. To resolve this we stained DMP1-GFP cells for haematopoietic lineage (Lin) surface markers (CD2, CD3e, CD4, CD45, CD5, CD8, CD11b, B220, Gr1, Ter119) and CD31. Lin(-)CD31(-) (Lin(-)) and Lin(+)CD31(+) (Lin(+)) populations were analysed for GFP, and the four resulting populations assessed by quantitative real-time PCR. Lin(-)GFP(+) cells expressed mRNAs for Sost, Dmp1, and Mepe, confirming their osteocyte identity. Dcstamp and Oscar mRNAs were restricted to haematopoietic (Lin(+)) cells, but Calcr, Ctsk and Acp5 were readily detected in purified osteocytes (Lin(-)GFP(+)). The capacity of these purified osteocytes to support osteoclastogenesis was assessed: no TRAP+ cells with >2 nuclei were formed when purified osteocytes were cultured with bone marrow macrophages and stimulated with 1,25-dihydroxyvitamin-D3/prostaglandin E2. Lin(-)GFP(+) osteocytes also expressed lower levels of Tnfsf11 (RANKL) mRNA than the osteoblast-enriched population (Lin(-)GFP(-)). This demonstrates the importance of haematopoietic depletion in generating highly purified osteocytes and shows that osteocytes express Acp5, Ctsk and Calcr, but not other osteoclast markers, and do not fully support osteoclast formation in vitro.


Assuntos
Proteínas da Matriz Extracelular/genética , Células-Tronco Hematopoéticas/citologia , Osteócitos/citologia , Fosfatase Ácida/metabolismo , Animais , Catepsina K/metabolismo , Linhagem da Célula , Proliferação de Células , Separação Celular , Citometria de Fluxo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/química , Isoenzimas/metabolismo , Camundongos , Osteoblastos/metabolismo , Osteoclastos/citologia , Fenótipo , Ligante RANK/metabolismo , Receptores da Calcitonina/metabolismo , Fosfatase Ácida Resistente a Tartarato
17.
Trends Microbiol ; 23(1): 35-43, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25449049

RESUMO

Arthritogenic alphaviral infection begins as a febrile illness and often progresses to joint pain and rheumatic symptoms that are described as polyarthritis. Alphaviral arthritis and classical arthritides share many similar cellular and immune mediators involved in their pathogenesis. Recent in vitro and in vivo evidence suggests that bone loss resulting from increased expression of bone resorption mediators may accompany alphaviral infection. In addition, several longitudinal studies have reported more severe and delayed recovery of alphaviral disease in patients with pre-existing arthritic conditions. This review aims to provide insights into alphavirus-induced bone loss and focuses on aspects of disease exacerbation in patients with underlying arthritis and on possible therapeutic targets.


Assuntos
Alphavirus/genética , Artrite Infecciosa/virologia , Reabsorção Óssea/patologia , Animais , Artrite Infecciosa/complicações , Reabsorção Óssea/etiologia , Reabsorção Óssea/virologia , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Inflamação/virologia , Interleucina-17/imunologia , Interleucina-6/imunologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Fatores de Risco , Células Th17/imunologia
18.
J Bone Miner Res ; 30(5): 886-97, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25418357

RESUMO

Skeletal-related events resulting from accelerated bone loss are common complications in patients treated for a range of cancers. However, the mechanisms and rate of bone loss after myelosuppression are unclear. We, therefore, investigated this in mice and humans. We treated mice with different myelosuppressive therapies (chemotherapy or irradiation with or without transplantation) and studied their effects on bone structure. Myelosuppression of mice rapidly caused an increase in bone resorption that was not matched by bone formation. The resultant significant and persistent bone loss early after therapy was associated with increased inflammatory cytokines, in particular, monocyte chemoattractant protein 1 (MCP1). Therapy-induced bone loss was prevented with a single dose of the bisphosphonate zoledronic acid (ZA), administered before myelosuppression. Importantly, ZA treatment of mice did not impair hematopoiesis, including hematopoietic stem cell function. Furthermore, examination of serum from patients before and after autologous or allogeneic stem cell transplantion (SCT) revealed altered levels of bone turnover markers and elevated inflammatory cytokines. MCP1 levels in serum obtained between days 7 and 14 post-SCT positively correlated with bone loss observed at 100 days after allogeneic SCT. Similar to that observed in our studies in mice, the bone loss was long term, persisting at 12 months post-SCT. Furthermore, patients who received chemotherapy less than 100 days before SCT had significantly more bone loss at the hip. In these patients, serum levels of MCP1, but not routine biomarkers of bone turnover, including C-terminal cross-linking telopeptide of type-1 collagen (ß-CTx), positively correlated with their bone loss. Hence, myelosuppressive therapies increase inflammation and directly contribute to bone loss. Administration of an osteoclast inhibitor before the initiation of cancer therapy is likely to have the best outcome in preventing bone loss in patients with cancer.


Assuntos
Antineoplásicos/efeitos adversos , Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Animais , Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/patologia , Quimiocina CCL2/sangue , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Hematopoese/efeitos dos fármacos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Inflamação/patologia , Camundongos Endogâmicos C57BL , Transplante de Células-Tronco , Transplante Homólogo , Microtomografia por Raio-X , Ácido Zoledrônico
19.
Sci Rep ; 5: 10120, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25944566

RESUMO

Osteosarcoma (OS) survival rates have plateaued in part due to a lack of new therapeutic options. Here we demonstrate that bromodomain inhibitors (BETi), JQ1, I-BET151, I-BET762, exert potent anti-tumour activity against primary and established OS cell lines, mediated by inhibition of BRD4. Strikingly, unlike previous observations in long-term established human OS cell lines, the antiproliferative activity of JQ1 in primary OS cells was driven by the induction of apoptosis, not cell cycle arrest. In further contrast, JQ1 activity in OS was mediated independently of MYC downregulation. We identified that JQ1 suppresses the transcription factor FOSL1 by displacement of BRD4 from its locus. Loss of FOSL1 phenocopied the antiproliferative effects of JQ1, identifying FOSL1 suppression as a potential novel therapeutic approach for OS. As a monotherapy JQ1 demonstrated significant anti-tumour activity in vivo in an OS graft model. Further, combinatorial treatment approaches showed that JQ1 increased the sensitivity of OS cells to doxorubicin and induced potent synergistic activity when rationally combined with CDK inhibitors. The greater level of activity achieved with the combination of BETi with CDK inhibitors demonstrates the efficacy of this combination therapy. Taken together, our studies show that BET inhibitors are a promising new therapeutic for OS.


Assuntos
Apoptose/efeitos dos fármacos , Osteossarcoma/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Azepinas/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Osteossarcoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição/metabolismo , Triazóis/farmacologia
20.
Gene ; 307: 111-23, 2003 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-12706893

RESUMO

Tartrate-resistant acid phosphatase (TRAP) is highly expressed in osteoclasts and in a subset of tissue macrophages and dendritic cells. It is expressed at lower levels in the parenchymal cells of the liver, glomerular mesangial cells of the kidney and pancreatic acinar cells. We have identified novel TRAP mRNAs that differ in their 5'-untranslated region (5'-UTR) sequence, but align with the known murine TRAP mRNA from the first base of Exon 2. The novel 5'-UTRs represent alternative first exons located upstream of the known 5'-UTR. A similar genomic structure exists for the human TRAP gene with partial conservation of the exon and promoter sequences. Expression of the most distal 5'-UTR (Exon 1A) is restricted to adult bone and spleen tissue. Exon 1B is expressed primarily in tissues containing TRAP-positive non-haematopoietic cells. The known TRAP 5'-UTR (Exon 1C) is expressed in tissues characteristic of myeloid cell expression. In addition the Exon 1C promoter sequence is shown to comprise distinct transcription start regions, with an osteoclast-specific transcription initiation site identified downstream of a TATA-like element. Macrophages are shown to initiate transcription of the Exon 1C transcript from a purine-rich region located upstream of the osteoclast-specific transcription start point. The distinct expression patterns for each of the TRAP 5'-UTRs suggest that TRAP mRNA expression is regulated by the use of four alternative tissue- and cell-restricted promoters.


Assuntos
Fosfatase Ácida/genética , Isoenzimas/genética , Regiões Promotoras Genéticas/genética , Regiões 5' não Traduzidas/genética , Processamento Alternativo , Animais , Sequência de Bases , Células Cultivadas , Éxons , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes/genética , Humanos , Íntrons , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Fosfatase Ácida Resistente a Tartarato , Sítio de Iniciação de Transcrição , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA