Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
Nature ; 453(7198): 1112-6, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18500333

RESUMO

Understanding the molecular underpinnings of cancer is of critical importance to the development of targeted intervention strategies. Identification of such targets, however, is notoriously difficult and unpredictable. Malignant cell transformation requires the cooperation of a few oncogenic mutations that cause substantial reorganization of many cell features and induce complex changes in gene expression patterns. Genes critical to this multifaceted cellular phenotype have therefore only been identified after signalling pathway analysis or on an ad hoc basis. Our observations that cell transformation by cooperating oncogenic lesions depends on synergistic modulation of downstream signalling circuitry suggest that malignant transformation is a highly cooperative process, involving synergy at multiple levels of regulation, including gene expression. Here we show that a large proportion of genes controlled synergistically by loss-of-function p53 and Ras activation are critical to the malignant state of murine and human colon cells. Notably, 14 out of 24 'cooperation response genes' were found to contribute to tumour formation in gene perturbation experiments. In contrast, only 1 in 14 perturbations of the genes responding in a non-synergistic manner had a similar effect. Synergistic control of gene expression by oncogenic mutations thus emerges as an underlying key to malignancy, and provides an attractive rationale for identifying intervention targets in gene networks downstream of oncogenic gain- and loss-of-function mutations.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias do Colo/genética , Mutação/genética , Oncogenes/genética , Animais , Linhagem Celular , Colo/citologia , Colo/patologia , Regulação Neoplásica da Expressão Gênica , Genes p53/genética , Genes ras/genética , Genótipo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fenótipo
3.
Arthritis Rheum ; 64(8): 2611-23, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22422036

RESUMO

OBJECTIVE: The incidence of low back pain is extremely high and is often linked to intervertebral disc (IVD) degeneration. The mechanism of this disease is currently unknown. This study was undertaken to investigate the role of ß-catenin signaling in IVD tissue function. METHODS: ß-catenin protein levels were measured by immunohistochemical analysis of disc samples obtained from patients with disc degeneration and from normal subjects. To generate ß-catenin conditional activation (cAct) mice, Col2a1-CreER(T2) -transgenic mice were bred with ß-catenin(fx(Ex3)/fx(Ex3)) mice. Changes in disc tissue morphology and function were examined by micro-computed tomography, histologic analysis, and real-time polymerase chain reaction assays. RESULTS: ß-catenin protein was up-regulated in disc tissue samples from patients with disc degeneration. To assess the effects of increased ß-catenin levels on disc tissue, we generated ß-catenin cAct mice. Overexpression of ß-catenin in disc cells led to extensive osteophyte formation in 3- and 6-month-old ß-catenin cAct mice, which were associated with significant changes in the cells and extracellular matrix of disc tissue and growth plate. Gene expression analysis demonstrated that activation of ß-catenin enhanced runt-related transcription factor 2-dependent Mmp13 and Adamts5 expression. Moreover, genetic ablation of Mmp13 or Adamts5 on the ß-catenin cAct background, or treatment of ß-catenin cAct mice with a specific matrix metalloproteinase 13 inhibitor, ameliorated the mutant phenotype. CONCLUSION: Our findings indicate that the ß-catenin signaling pathway plays a critical role in disc tissue function.


Assuntos
Degeneração do Disco Intervertebral/fisiopatologia , Deslocamento do Disco Intervertebral/fisiopatologia , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , beta Catenina/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAMTS5 , Animais , Colágeno Tipo II/genética , Modelos Animais de Doenças , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais
4.
J Cell Biochem ; 113(6): 2156-66, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22461172

RESUMO

Since transforming growing factor-ß (TGF-ß)/Smad signaling inhibits chondrocyte maturation, endogenous negative regulators of TGF-ß signaling are likely also important regulators of the chondrocyte differentiation process. One such negative regulator, Ski, is an oncoprotein that is known to inhibit TGF-ß/Smad3 signaling via its interaction with phospho-Smad3 and recruitment of histone deacetylases (HDACs) to the DNA binding complex. Based on this, we hypothesized that Ski inhibits TGF-ß signaling and accelerates maturation in chondrocytes via recruitment of HDACs to transcriptional complexes containing Smads. We tested this hypothesis in chick upper sternal chondrocytes (USCs), where gain and loss of Ski expression experiments were performed. Over-expression of Ski not only reversed the inhibitory effect of TGF-ß on the expression of hypertrophic marker genes such as type X collagen (colX) and osteocalcin, it induced these genes basally as well. Conversely, knockdown of Ski by RNA interference led to a reduction of colX and osteocalcin expression under basal conditions. Furthermore, Ski blocked TGF-ß induction of cyclinD1 and caused a basal up-regulation of Runx2, consistent with the observed acceleration of hypertrophy. Regarding mechanism, not only does Ski associate with phospho-Smad2 and 3, but its association with phospho-Smad3 is required for recruitment of HDAC4 and 5. Implicating this recruitment of HDACs in the phenotypic effects of Ski in chondrocytes, the HDAC inhibitor SAHA reversed the up-regulation of colX and osteocalcin in Ski over-expressing cells. These results suggest that inhibition of TGF-ß signaling by Ski, which involves its association with phospho-Smad3 and recruitment of HDAC4 and 5, leads to accelerated chondrocyte differentiation.


Assuntos
Condrócitos/citologia , Condrócitos/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Galinha , Colágeno Tipo X/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Ciclina D1/biossíntese , Histona Desacetilases/metabolismo , Osteocalcina/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Proteína Smad2/metabolismo
5.
Arthritis Rheum ; 62(8): 2359-69, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20506210

RESUMO

OBJECTIVE: To investigate the biologic significance of Smad3 in the progression of osteoarthritis (OA), the crosstalk between Smad3 and activating transcription factor 2 (ATF-2) in the transforming growth factor beta (TGFbeta) signaling pathway, and the effects of ATF-2 overexpression and p38 activation in chondrocyte differentiation. METHODS: Joint disease in Smad3-knockout (Smad3(-/-)) mice was examined by microfocal computed tomography and histologic analysis. Numerous in vitro methods including immunostaining, real-time polymerase chain reaction, Western blotting, an ATF-2 DNA-binding assay, and a p38 kinase activity assay were used to study the various signaling responses and protein interactions underlying the altered chondrocyte phenotype in Smad3(-/-) mice. RESULTS: In Smad3(-/-) mice, an end-stage OA phenotype gradually developed. TGFbeta-activated kinase 1 (TAK1)/ATF-2 signaling was disrupted in Smad3(-/-) mouse chondrocytes at the level of p38 MAP kinase (MAPK) activation, resulting in reduced ATF-2 phosphorylation and transcriptional activity. Reintroduction of Smad3 into Smad3(-/-) cells restored the normal p38 response to TGFbeta. Phosphorylated p38 formed a complex with Smad3 by binding to a portion of Smad3 containing both the MAD homology 1 and linker domains. Additionally, Smad3 inhibited the dephosphorylation of p38 by MAPK phosphatase 1 (MKP-1). Both ATF-2 overexpression and p38 activation repressed type X collagen expression in wild-type and Smad3(-/-) chondrocytes. P38 was detected in articular cartilage and perichondrium; articular and sternal chondrocytes expressed p38 isoforms alpha, beta, and gamma, but not delta. CONCLUSION: Smad3 is involved in both the onset and progression of OA. Loss of Smad3 abrogates TAK1/ATF-2 signaling, most likely by disrupting the Smad3-phosphorylated p38 complex, thereby promoting p38 dephosphorylation and inactivation by MKP-1. ATF-2 and p38 activation inhibit chondrocyte hypertrophy. Modulation of p38 isoform activity may provide a new therapeutic approach for OA.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Condrócitos/patologia , Osteoartrite/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator 2 Ativador da Transcrição/genética , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Progressão da Doença , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Osteoartrite/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Smad3/genética , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Front Immunol ; 12: 630204, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717161

RESUMO

Regulatory T (Treg) cells are essential to maintain immune homeostasis in the intestine and Treg cell dysfunction is associated with several inflammatory and autoimmune disorders including inflammatory bowel disease (IBD). Efforts using low-dose (LD) interleukin-2 (IL-2) to expand autologous Treg cells show therapeutic efficacy for several inflammatory conditions. Whether LD IL-2 is an effective strategy for treating patients with IBD is unknown. Recently, we demonstrated that LD IL-2 was protective against experimental colitis in immune humanized mice in which human CD4+ T cells were restricted to human leukocyte antigen (HLA). Whether HLA restriction is required for human Treg cells to ameliorate colitis following LD IL-2 therapy has not been demonstrated. Here, we show that treatment with LD IL-2 reduced 2,4,6-trinitrobenzensulfonic acid (TNBS) colitis severity in NOD.PrkdcscidIl2rg-/- (NSG) mice reconstituted with human CD34+ hematopoietic stem cells. These data demonstrate the utility of standard immune humanized NSG mice as a pre-clinical model system to evaluate therapeutics targeting human Treg cells to treat IBD.


Assuntos
Antígenos HLA/imunologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Interleucina-2/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Humanos , Doenças Inflamatórias Intestinais/imunologia , Interleucina-2/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Ácido Trinitrobenzenossulfônico/farmacologia
7.
Exp Cell Res ; 315(14): 2386-98, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19481076

RESUMO

We have previously demonstrated that Smurf2 is highly expressed in human osteoarthritis (OA) tissue, and overexpression of Smurf2 under the control of the type II collagen promoter (Col2a1) induces an OA-like phenotype in aged Col2a1-Smurf2 transgenic mice, suggesting that Smurf2 is located upstream of a signal cascade which initiates OA development. However, the factors downstream of Smurf2 in this signal cascade and how Smurf2-induced OA is initiated are largely unknown. In this study, we further characterized the phenotypic changes in Col2a1-Smurf2 transgenic and WT articular cartilage from the postnatal stage to adulthood. We found that the articular cartilage degeneration occurring at the cartilage surface in 6 month-old Col2a1-Smurf2 transgenic mice progressed from an expanded hypertrophic domain in the basal layer of the deep articular cartilage at 2.5 weeks of age, which may lead to an accelerated calcification and ectopic ossification of this region at 1 month of age, and aggregation and maturation of articular chondrocytes in the middle and deep zones at 2 months and 4.5 months of age, respectively. Furthermore, we discovered that ectopically expressed Smurf2 interacted with GSK-3beta and induced its ubiquitination and subsequent proteasomal degradation, and hence upregulated beta-catenin in Col2a1-Smurf2 transgenic chondrocytes ex vivo. It is therefore likely that Smurf2-mediated upregulation of beta-catenin through induction of proteasomal degradation of GSK-beta in chondrocytes may activate articular chondrocyte maturation and associated alteration of gene expression, the early events of OA.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Osteoartrite/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , beta Catenina/metabolismo , Animais , Cartilagem Articular/patologia , Condrócitos/patologia , Colágeno Tipo II/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Camundongos Transgênicos , Osteoartrite/patologia , Regulação para Cima/genética
8.
Methods Mol Biol ; 1130: 61-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24482165

RESUMO

Given the prevalence and the scope of the personal and societal burden of OA, investigators have become increasingly interested in understanding the pathogenic basis of disease and developing novel disease-modifying OA therapies. Because of the well-documented central role that joint trauma plays in the initiation of knee OA, large animal and rodent models of knee injury that accurately recapitulate the OA disease process have become increasingly widespread over the past decade. To enable study in the context of defined genetic backgrounds, investigative teams have informally developed standardized protocols for injuring the mouse knee that aim to induce a reproducible degenerative process both in terms of severity and temporal pacing of disease progression. One such procedure, the meniscal/ligamentous injury (MLI) model of posttraumatic OA, is described in detail in this chapter. The description provided here sets the stage for both inexperienced and established investigators to employ the MLI procedure, or other similar surgical destabilization methods, to initiate the development of posttraumatic OA in the mouse. Successful application of this method provides a preclinical platform to study the mechanisms driving the pathogenesis of OA and to develop chondroprotective/regenerative strategies to treat it.


Assuntos
Osteoartrite/etiologia , Osteoartrite/cirurgia , Ferimentos e Lesões/complicações , Animais , Cartilagem/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Procedimentos Ortopédicos , Osteoartrite/patologia
9.
Arthritis Res Ther ; 15(1): R5, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23298463

RESUMO

INTRODUCTION: Osteoarthritis (OA) is a degenerative joint disease affecting a large population of people. The mechanism of this highly prevalent disease is not fully understood. Currently there is no effective disease-modifying treatment for OA. The purpose of this study was two-fold: 1) to investigate the role of MMP13 in the development of OA; and 2) to evaluate the efficacy of the MMP13 inhibitor CL82198 as a pharmacologic treatment for preventing OA progression. METHODS: To investigate the role of the endogenous Mmp13 gene in OA development, tamoxifen was administered to two-week-old Col2CreER;Mmp13fx/fx (Mmp13Col2ER) and Cre-negative control mice for five days. OA was induced by meniscal-ligamentous injury (MLI) when the mice were 10 weeks old and MLI or sham-operated joints were harvested 4, 8, 12, or 16 weeks after surgery. To evaluate the efficacy of CL82198, MLI surgery was performed on 10-week-old wild type mice. CL82198 or saline was administered to the mice daily beginning immediately after the surgery for up to 16 weeks. The joint tissues collected from both experiments were evaluated by cartilage grading, histology/histomorphometry, immunohistochemistry (IHC), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The ability of CL82198 to inhibit MMP13 activity in vitro was confirmed by ELISA. RESULTS: The OA progression was decelerated in Mmp13Col2ER mice 8, 12, and 16 weeks post-surgery. Cartilage grading by blinded observers confirmed decreased articular cartilage degeneration in Mmp13Col2ER mice at 8, 12 and 16 weeks compared to Cre-negative mice. Histomorphometric analysis demonstrated that Mmp13Col2ER mice had a higher articular cartilage area and thickness at 12 and 16 weeks post-surgery compared to the control mice. Results of IHC revealed greater type II collagen and proteoglycan expression in Mmp13Col2ER mice. Chondrocyte apoptosis, as determined by TUNEL staining, was higher in control mice compared to Mmp13Col2ER mice. CL82198 inhibited MMP13 activity in conditioned media from vehicle (>85%) or bone morphogenetic protein 2 (BMP2)-treated (>90%) primary murine sternal chondrocytes. Intraperitoneal injection of CL82198 decelerated MLI-induced OA progression, increased type II collagen and proteoglycan levels, and inhibited chondrocyte apoptosis compared to saline treatment as determined by OA grading, histology, histomorphometry, IHC, and TUNEL staining, respectively. CONCLUSIONS: Mmp13 is critical for OA progression and pharmacologic inhibition of MMP13 is an effective strategy to decelerate articular cartilage loss in a murine model of injury-induced knee OA.


Assuntos
Metaloproteinase 13 da Matriz/metabolismo , Osteoartrite/enzimologia , Animais , Anti-Inflamatórios/farmacologia , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Osteoartrite/genética , Osteoartrite/patologia
10.
J Orthop Res ; 30(2): 325-33, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21809376

RESUMO

Management of various tumor metastases to bone has dramatically improved, but this is not so for renal cell carcinoma (RCC), which is a difficult surgical problem due to its great vascularity. Furthermore, the unique mechanisms that mediate RCC vasculogenesis in bone remain unknown. To understand this process we developed a xenograft model that recapitulates highly vascular RCC versus less vascular tumors that metastasize to bone. Human tumor cell lines of RCC (786-O), prostate cancer (PC3), lung cancer (A549), breast cancer (MDA-MB231), and melanoma (A375) were transduced with firefly luciferase (Luc), injected into the tibiae of nude mice, and differences in growth, osteolysis, and vascularity were assessed by longitudinal bioluminescent imaging, micro-CT for measurement of calcified tissues and vascularity and histology. The results showed that while RCC-Luc has reduced growth and osteolytic potential versus the other tumor lines, it displayed a significant increase in vascular volume (p < 0.05). This expansion was due to 3- and 5-fold increases in small and large vessel numbers respectively. In vitro gene expression profiling revealed that RCC-Luc expresses significantly (p < 0.05) more vegf-a (10-fold) and 20- to 30-fold less ang-1 versus the other lines. These data demonstrate the utility of this model to study the unique vasculogenic properties of RCC bone metastases.


Assuntos
Angiopoietina-1/fisiologia , Neoplasias Ósseas/irrigação sanguínea , Neoplasias Ósseas/secundário , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Neovascularização Patológica/etiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Angiopoietina-1/genética , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Transplante de Neoplasias , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/genética
11.
J Orthop Res ; 30(11): 1760-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22517267

RESUMO

Lead remains a significant environmental toxin, and we believe we may have identified a novel target of lead toxicity in articular chondrocytes. These cells are responsible for the maintenance of joint matrix, and do so under the regulation of TGF-ß signaling. As lead is concentrated in articular cartilage, we hypothesize that it can disrupt normal chondrocyte phenotype through suppression of TGF-ß signaling. These experiments examine the effects of lead exposure in vivo and in vitro at biologically relevant levels, from 1 nM to 10 µM on viability, collagen levels, matrix degrading enzyme activity, TGF-ß signaling, and articular surface morphology. Our results indicate that viability was unchanged at levels ≤100 µM Pb, but low and high level lead in vivo exposure resulted in fibrillation and degeneration of the articular surface. Lead treatment also decreased levels of type II collagen and increased type X collagen, in vivo and in vitro. Additionally, MMP13 activity increased in a dose-dependent manner. Active caspase 3 and 8 were dose-dependently elevated, and treatment with 10 µM Pb resulted in increases of 30% and 500%, respectively. Increasing lead treatment resulted in a corresponding reduction in TGF-ß reporter activity, with a 95% reduction at 10µM. Levels of phosphoSmad2 and 3 were suppressed in vitro and in vivo and lead dose-dependently increased Smurf2. These changes closely parallel those seen in osteoarthritis. Over time this phenotypic shift could compromise maintenance of the joint matrix.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Chumbo/toxicidade , Osteoartrite/induzido quimicamente , Fator de Crescimento Transformador beta/metabolismo , Animais , Cartilagem Articular/metabolismo , Linhagem Celular , Galinhas , Condrócitos/metabolismo , Fenótipo , Ratos , Transdução de Sinais/efeitos dos fármacos , Testes de Toxicidade Aguda
12.
J Orthop Res ; 29(4): 623-32, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20957741

RESUMO

Soft tissue sarcoma (STS) is a rare malignancy that is generally resistant to chemotherapy. We investigated the ability of the histone deacetylase inhibitor vorinostat to sensitize STS cells versus normal fibroblasts to chemotherapy. Fibrosarcoma, leiomyosarcoma, and liposarcoma cells and normal fibroblasts were treated with vorinostat to determine effects on proliferation and basal apoptosis as measured by total cell number and cleaved caspase 3 staining. Effects on histone deacetylases (HDAC) activity were confirmed by Western blotting for acetylated histone H3. A clinically relevant dose of vorinostat that had no effect on basal apoptosis was selected to examine altered sensitivity to doxorubicin. The effects of vorinostat, doxorubicin, or the combination on fibrosarcoma growth in vivo were determined in a xenograft model. Tumor volume was measured biweekly and HDAC activity and cell death were assessed by immunohistochemical analysis of acetylated histone H3, cleaved caspase 3, and TUNEL staining. Vorinostat inhibited proliferation and induced histone acetylation without affecting basal apoptosis levels. Combined treatment with vorinostat and doxorubicin synergistically induced apoptosis in vitro in fibrosarcoma but not leiomyosarcoma, liposarcoma, or normal fibroblasts. In nude mice, the combination of vorinostat and doxorubicin inhibited fibrosarcoma xenograft growth further than either agent alone. Cell death, as measured by cleaved caspase 3 and TUNEL staining, was greatest in xenografts from mice treated with vorinostat and doxorubicin. Vorinostat inhibits growth and induces chemosensitivity in fibrosarcoma cells in vitro and in vivo, suggesting that the combination of vorinostat and chemotherapy may represent a novel treatment option for this STS subtype. © 2010 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 29:623-632, 2011.


Assuntos
Antineoplásicos/farmacologia , Fibrossarcoma/tratamento farmacológico , Histona Acetiltransferases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias de Tecidos Moles/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibrossarcoma/enzimologia , Fibrossarcoma/patologia , Histona Acetiltransferases/antagonistas & inibidores , Humanos , Camundongos , Camundongos Nus , Neoplasias de Tecidos Moles/enzimologia , Neoplasias de Tecidos Moles/patologia , Vorinostat , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Bone Miner Res ; 26(6): 1283-94, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21308771

RESUMO

Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. Ninety percent of patients who present with metastatic and 30% to 40% of patients with nonmetastatic disease experience relapse, creating an urgent need for novel therapeutic strategies. The Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), are important for mitosis, motility, and cell survival. Upregulation of Met/HGF signaling via receptor overexpression, amplification, or mutation drives the proliferation, invasiveness, and metastasis of a variety of cancer cells, including OS, prompting the development of Met/HGF inhibitors. OS cells depend on Met overexpression because introduction of dominant-negative Met inhibits in vivo tumorigenicity. Despite the importance of Met/HGF signaling in the development and maintenance of OS, the potential efficacy of pharmacologic Met inhibition in OS has been addressed only in in vitro studies. PF-2341066 is an orally bioavailable, selective ATP-competitive Met inhibitor that showed promising results recently in a phase I clinical trial in non-small cell lung cancer (NSCLC) patients. We tested the ability of PF-2341066 to inhibit malignant properties of osteosarcoma cells in vitro and orthotopic xenograft growth in vivo. In vitro, PF-2341066 inhibited osteosarcoma behavior associated with primary tumor growth (eg, proliferation and survival) as well as metastasis (eg, invasion and clonogenicity). In nude mice treated with PF-2341066 via oral gavage, the growth and associated osteolysis and extracortical bone matrix formation of osteosarcoma xenografts were inhibited by PF-2341066. PF-2341066 may represent an effective new systemic therapy for localized and potentially disseminated osteosarcoma.


Assuntos
Matriz Óssea/metabolismo , Osteólise/patologia , Osteossarcoma/patologia , Piperidinas/administração & dosagem , Piperidinas/farmacocinética , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Piridinas/administração & dosagem , Piridinas/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Disponibilidade Biológica , Matriz Óssea/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Clonais , Crizotinibe , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Osteogênese/efeitos dos fármacos , Osteólise/complicações , Osteossarcoma/complicações , Fosforilação/efeitos dos fármacos , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirazóis , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
14.
Arthritis Res Ther ; 13(6): R198, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22152451

RESUMO

INTRODUCTION: Increasing obesity and type 2 diabetes, in part due to the high-fat (HF) Western diet, parallels an increased incidence of osteoarthritis (OA). This study was undertaken to establish a causal relation between the HF diet and accelerated OA progression in a mouse model and to determine the relative roles of weight gain and metabolic dysregulation in this progression. METHODS: Five-week-old C57BL/6 mice were placed on HF (60% kcal) or low-fat (lean, 10% kcal) diets for 8 or 12 weeks before transecting the medial collateral ligament and excising a segment of the medial meniscus of the knee to initiate OA. One group was switched from lean to HF diet at the time of surgery. RESULTS: Body weight of mice on the HF diet peaked at 45.9 ± 2.1 g compared with 29.9 ± 1.8 g for lean diets, with only those on the HF becoming diabetic. Severity of OA was greater in HF mice, evidenced by the Osteoarthritis Research Society International (OARSI) histopathology initiative scoring method for mice and articular cartilage thickness and area. To assess the importance of weight gain, short- and long-term HF diets were compared with the lean diet. Short- and long-term HF groups outweighed lean controls by 6.2 g and 20.5 g, respectively. Both HF groups became diabetic, and OA progression, evidenced by increased OARSI score, decreased cartilage thickness, and increased osteophyte diameter, was comparably accelerated relative to those of lean controls. CONCLUSIONS: These results demonstrate that the HF diet accelerates progression of OA in a type 2 diabetic mouse model without correlation to weight gain, suggesting that metabolic dysregulation is a comorbid factor in OA-related cartilage degeneration.


Assuntos
Ligamentos Colaterais/cirurgia , Dieta Hiperlipídica/efeitos adversos , Meniscos Tibiais/cirurgia , Osteoartrite do Joelho/etiologia , Animais , Peso Corporal/efeitos dos fármacos , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Diabetes Mellitus Tipo 2/etiologia , Gorduras na Dieta/administração & dosagem , Progressão da Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Osteoartrite do Joelho/diagnóstico por imagem , Fatores de Tempo , Aumento de Peso/efeitos dos fármacos , Microtomografia por Raio-X
15.
J Orthop Res ; 29(8): 1145-51, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21374709

RESUMO

The goals of our study were to establish quantitative outcomes for assessing murine knee arthritis and develop an Arthritis Index that incorporates multiple outcomes into a single calculation that provides enhanced sensitivity. Using an accepted model of meniscal/ligamentous injury (MLI)-induced osteoarthritis (OA), we assessed mouse knee arthritis using several approaches. Histology-based methods were performed to visualize joint tissues including articular cartilage and subchondral bone. Accepted histologic scoring methods and histomorphometry were performed to grade cartilage degeneration and determine articular cartilage area, respectively. MicroCT was used to visualize and quantify the bony structures of the joint including osteophytes and joint bone volume. A statistical algorithm was then developed that combined histologic scores and cartilage areas into a single Arthritis Index. MLI induced progressive, OA-like articular cartilage degeneration characterized by increasing (worsening) histologic score and decreasing cartilage area. MicroCT revealed osteophytes and increased joint bone volume between the femoral and tibial physes following MLI. Lastly, an Arthritis Index calculation was established, which incorporated histologic scoring and cartilage area. The Arthritis Index provided enhanced quantitative sensitivity in assessing the level of joint degeneration compared to either histologic scoring or cartilage area determination alone; when using the Index, between 29% and 43% fewer samples are needed to establish statistical significance in studies of murine arthritis. Arthritis in the mouse knee can be quantitatively assessed by histologic scoring, measuring cartilage area, and determining joint bone volume. Enhanced sensitivity can be achieved by performing the Arthritis Index calculation, a novel method for quantitatively assessing mouse knee arthritis.


Assuntos
Cartilagem Articular/patologia , Membro Posterior/patologia , Osteoartrite do Joelho/patologia , Índice de Gravidade de Doença , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
Sci Transl Med ; 3(101): 101ra93, 2011 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-21937758

RESUMO

There is no disease-modifying therapy for osteoarthritis, a degenerative joint disease that is projected to afflict more than 67 million individuals in the United States alone by 2030. Because disease pathogenesis is associated with inappropriate articular chondrocyte maturation resembling that seen during normal endochondral ossification, pathways that govern the maturation of articular chondrocytes are candidate therapeutic targets. It is well established that parathyroid hormone (PTH) acting via the type 1 PTH receptor induces matrix synthesis and suppresses maturation of chondrocytes. We report that the PTH receptor is up-regulated in articular chondrocytes after meniscal injury and in osteoarthritis in humans and in a mouse model of injury-induced knee osteoarthritis. To test whether recombinant human PTH(1-34) (teriparatide) would inhibit aberrant chondrocyte maturation and associated articular cartilage degeneration, we administered systemic teriparatide (Forteo), a Food and Drug Administration-approved treatment for osteoporosis, either immediately after or 8 weeks after meniscal/ligamentous injury in mice. Knee joints were harvested at 4, 8, or 12 weeks after injury to examine the effects of teriparatide on cartilage degeneration and articular chondrocyte maturation. Microcomputed tomography revealed increased bone volume within joints from teriparatide-treated mice compared to saline-treated control animals. Immediate systemic administration of teriparatide increased proteoglycan content and inhibited articular cartilage degeneration, whereas delayed treatment beginning 8 weeks after injury induced a regenerative effect. The chondroprotective and chondroregenerative effects of teriparatide correlated with decreased expression of type X collagen, RUNX2 (runt-related transcription factor 2), matrix metalloproteinase 13, and the carboxyl-terminal aggrecan cleavage product NITEGE. These preclinical findings provide proof of concept that Forteo may be useful for decelerating cartilage degeneration and inducing matrix regeneration in patients with osteoarthritis.


Assuntos
Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Osteoartrite/tratamento farmacológico , Osteoartrite/patologia , Regeneração/efeitos dos fármacos , Teriparatida/farmacologia , Teriparatida/uso terapêutico , Anabolizantes/farmacologia , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Articulações/efeitos dos fármacos , Articulações/enzimologia , Articulações/patologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Meniscos Tibiais/efeitos dos fármacos , Meniscos Tibiais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/complicações , Osteoartrite/metabolismo , Osteófito/complicações , Osteófito/patologia , Proteoglicanas/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Medicina Regenerativa , Proteínas Serrate-Jagged , Teriparatida/administração & dosagem , Lesões do Menisco Tibial
17.
Arthritis Rheum ; 58(10): 3132-44, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18821706

RESUMO

OBJECTIVE: To determine whether Smurf2, an E3 ubiquitin ligase known to inhibit transforming growth factor beta (TGFbeta) signaling, is expressed in human osteoarthritic (OA) cartilage and can initiate OA in mice. METHODS: Human OA cartilage was obtained from patients undergoing knee arthroplasty. Samples were graded histologically using the Mankin scale and were examined immunohistochemically for Smurf2 expression. A transgene driven by the collagen 2alpha1 promoter was used to overexpress Smurf2 in mice. Smurf2 overexpression in mouse sternal chondrocytes was confirmed by reverse transcription-polymerase chain reaction and Western blotting. Changes in articular cartilage area, chondrocyte number, and chondrocyte diameter were assessed histomorphometrically using OsteoMeasure software. Alterations in type X collagen and matrix metalloproteinase 13 (MMP-13) in articular chondrocytes were examined by in situ hybridization and immunohistochemistry, respectively. Joint bone phenotypes were evaluated by microfocal computed tomography. The effects of Smurf2 overexpression on TGFbeta signaling were examined using a luciferase-based reporter and immunoprecipitation/Western blotting. RESULTS: Human OA cartilage strongly expressed Smurf2 as compared with nonarthritic human cartilage. By 8 months of age, Smurf2-transgenic mice exhibited decreased articular cartilage area, fibrillation, clefting, eburnation, subchondral sclerosis, and osteophytes. Increased expression of type X collagen and MMP-13 were also detected in articular cartilage from transgenic mice. Transgenic sternal chondrocytes showed reduced TGFbeta signaling as well as decreased expression and increased ubiquitination of pSmad3. CONCLUSION: Smurf2 is up-regulated during OA in humans, and Smurf2-transgenic mice spontaneously develop an OA-like phenotype that correlates with decreased TGFbeta signaling and increased pSmad3 degradation. Overall, these results suggest a role of Smurf2 in the pathogenesis of OA.


Assuntos
Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Proteína Smad3/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células Cultivadas , Condrócitos/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Ubiquitina-Proteína Ligases/fisiologia , Regulação para Cima
18.
J Biol Chem ; 277(39): 36499-508, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12068007

RESUMO

The influence of estrogen on the development of the male reproductive system may be interrupted in a subset of partial androgen insensitivity syndrome (PAIS) patients. PAIS describes a wide range of male undermasculinization resulting from mutations in the androgen receptor (AR) or steroid metabolism enzymes that perturb androgen-AR regulation of male sex organ development. In this study, we are interested in determining if PAIS-derived AR mutants that respond normally to androgen have altered responses to estrogen in the presence of ARA70, a coregulator previously shown to enhance 17beta-estradiol E2-induced AR transactivation. The wild-type AR (wtAR) and two PAIS AR mutants, AR(S703G) and AR(E709K), all bind to androgen and E2 and subsequently translocate to the nucleus. Whereas ARA70 functionally interacts with the wtAR and the PAIS AR mutants in response to androgen, E2 only promotes the functional interaction between ARA70 and the wtAR but not the PAIS AR mutants. ARA70 increases E2 competitive binding to the wtAR in the presence of low level androgen and also retards E2 dissociation from the wtAR. ARA70 is present in both the cytoplasm and the nucleus of various mouse testicular cells during early embryogenesis day 16, at postpartum day 0 during estradiol synthesis and in the Leydig cells at postpartum day 49. ARA70 may be unable to modulate the PAIS AR mutants-E2 binding, diminishing the effect of E2 via AR during male reproductive system development in patients with such mutations. Therefore, the presence of ARA70 in the testosterone and E2-producing Leydig cells may enhance the overall activity of AR during critical stages of male sex organ development.


Assuntos
Estradiol/metabolismo , Mutação , Proteínas Oncogênicas , Receptores Androgênicos/metabolismo , Transativadores/genética , Fatores de Transcrição , Ativação Transcricional , Animais , Células COS , Núcleo Celular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Células Intersticiais do Testículo/metabolismo , Ligantes , Masculino , Microscopia de Fluorescência , Coativadores de Receptor Nuclear , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Receptores Androgênicos/genética , Fatores de Tempo , Transativadores/metabolismo , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
19.
J Biol Chem ; 277(18): 15426-31, 2002 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-11856738

RESUMO

The proline-rich tyrosine kinase 2 (Pyk2) was first identified as a key kinase linked to the MAP kinase and JNK signaling pathways that play important roles in cell growth and adhesion. The linkage between Pyk2 and the androgen receptor (AR), an important transcription factor in prostate cancer progression, however, remains unclear. Here we report that using the full-length androgen receptor-associated protein, ARA55, coregulator as bait, we were able to isolate an ARA55-interacting protein, Pyk2, and demonstrated that Pyk2 could repress AR transactivation via inactivation of ARA55. This inactivation may result from the direct phosphorylation of ARA55 by Pyk2 at tyrosine 43, impairing the coactivator activity of ARA55 and/or sequestering ARA55 to reduce its interaction with AR. Our finding that Pyk2 can indirectly modulate AR function via interaction and/or phosphorylation of ARA55 not only expands the role of Pyk2 in AR-mediated prostate cancer growth but also strengthens the role of ARA55 as an AR coregulator.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Receptores Androgênicos/genética , Transativadores/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Quinase 2 de Adesão Focal , Biblioteca Gênica , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Domínio LIM , Masculino , Mutagênese Sítio-Dirigida , Fosforilação , Próstata/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Ativação Transcricional , Transfecção
20.
J Biol Chem ; 279(32): 33438-46, 2004 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-15166229

RESUMO

Androgen receptor (AR)-associated coregulator 70 (ARA70) was the first identified AR coregulator. However, its molecular mechanism and biological relevance to prostate cancer remain unclear. Here we show that ARA70 interacts with and promotes AR activity via the consensus FXXLF motif within the ARA70-N2 domain (amino acids 176-401). However, it does not promote AR activity via the classic LXXLL motif located at amino acids 92-96, although this classic LXXLL motif is important for ARA70 to interact with other receptors, such as PPARgamma. The molecular mechanisms by which ARA70 enhances AR transactivation involve the increase of AR expression, protein stability, and nuclear translocation. Furthermore, ARA70 protein is more frequently detected in prostate cancer specimens (91.74%) than in benign tissues (64.64%, p < 0.0001). ARA70 expression is also increased in high-grade prostate cancer tissues as well as the hormone-refractory LNCaP xenografts and prostate cancer cell lines. Because ARA70 can promote the antiandrogen hydroxyflutamide (HF)-enhanced AR transactivation, the increased ARA70 expression in hormone-refractory prostate tumors may confer the development of HF withdrawal syndrome, commonly diagnosed in patients with the later stages of prostate cancer. Because ARA70-N2 containing the AR-interacting FXXLF motif without coactivation function can suppress HF-enhanced AR transactivation in the hormone-refractory LNCaP cells, using the ARA70-N2 inhibitory peptide at the hormone refractory stage to battle the HF withdrawal syndrome may become an alternative strategy to treat prostate cancer.


Assuntos
Flutamida/análogos & derivados , Expressão Gênica , Proteínas Oncogênicas/química , Proteínas Oncogênicas/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/fisiologia , Fatores de Transcrição/química , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Antagonistas de Androgênios/farmacologia , Animais , Western Blotting , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Sequência Consenso , Estabilidade de Medicamentos , Imunofluorescência , Flutamida/farmacologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Coativadores de Receptor Nuclear , Proteínas Oncogênicas/genética , RNA Mensageiro/análise , Receptores Androgênicos/genética , Relação Estrutura-Atividade , Fatores de Transcrição/genética , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA