Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 77(6): 1331-1344, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28108510

RESUMO

Identification of factors that mediate visceral and bone metastatic spread and subsequent bone remodeling events is highly relevant to successful therapeutic intervention in advanced human prostate cancer. TBX2, a T-box family transcription factor that negatively regulates cell-cycle inhibitor p21, plays critical roles during embryonic development, and recent studies have highlighted its role in cancer. Here, we report that TBX2 is overexpressed in human prostate cancer specimens and bone metastases from xenograft mouse models of human prostate cancer. Blocking endogenous TBX2 expression in PC3 and ARCaPM prostate cancer cell models using a dominant-negative construct resulted in decreased tumor cell proliferation, colony formation, and invasion in vitro Blocking endogenous TBX2 in human prostate cancer mouse xenografts decreased invasion and abrogation of bone and soft tissue metastasis. Furthermore, blocking endogenous TBX2 in prostate cancer cells dramatically reduced bone-colonizing capability through reduced tumor cell growth and bone remodeling in an intratibial mouse model. TBX2 acted in trans by promoting transcription of the canonical WNT (WNT3A) promoter. Genetically rescuing WNT3A levels in prostate cancer cells with endogenously blocked TBX2 partially restored the TBX2-induced prostate cancer metastatic capability in mice. Conversely, WNT3A-neutralizing antibodies or WNT antagonist SFRP-2 blocked TBX2-induced invasion. Our findings highlight TBX2 as a novel therapeutic target upstream of WNT3A, where WNT3A antagonists could be novel agents for the treatment of metastasis and for skeletal complications in prostate cancer patients. Cancer Res; 77(6); 1331-44. ©2017 AACR.


Assuntos
Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias Ósseas/prevenção & controle , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias da Próstata/prevenção & controle , Proteínas com Domínio T/antagonistas & inibidores , Proteína Wnt3A/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Gradação de Tumores , Prognóstico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Células Tumorais Cultivadas , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Oncotarget ; 7(9): 10433-47, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26862737

RESUMO

Prostate cancer (PCa) cells use matrix metalloproteinases (MMPs) to degrade tissue during invasion. Perlecan/HSPG2 is degraded at basement membranes, in reactive stroma and in bone marrow during metastasis. We previously showed MMP-7 efficiently degrades perlecan. We now analyzed PCa tissue and serum from 288 prostatectomy patients of various Gleason grades to decipher the relationship between perlecan and MMP-7 in invasive PCa. In 157 prostatectomy specimens examined by tissue microarray, perlecan levels were 18% higher than their normal counterparts. In Gleason grade 4 tissues, MMP-7 and perlecan immunostaining levels were highly correlated with each other (average correlation coefficient of 0.52) in PCa tissue, regardless of grade. Serial sections showed intense, but non-overlapping, immunostaining for MMP-7 and perlecan at adjacent borders, reflecting the protease-substrate relationship. Using a capture assay, analysis of 288 PCa sera collected at prostatectomy showed elevated levels of perlecan fragments, with most derived from domain IV. Perlecan fragments in PCa sera were associated with overall MMP-7 staining levels in PCa tissues. Domain IV perlecan fragments were present in stage IV, but absent in normal, sera, suggesting perlecan degradation during metastasis. Together, perlecan fragments in sera and MMP-7 in tissues of PCa patients are measures of invasive PCa.


Assuntos
Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/metabolismo , Proteoglicanas de Heparan Sulfato/sangue , Metaloproteinase 7 da Matriz/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Invasividade Neoplásica/patologia , Prostatectomia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/cirurgia , Microglobulina beta-2/sangue , Microglobulina beta-2/metabolismo
4.
Nat Commun ; 6: 8282, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26392000

RESUMO

HIV infection causes bone loss. We previously reported that immunosuppression-mediated B-cell production of receptor activator of NF-κB ligand (RANKL) coupled with decline in osteoprotegerin correlate with decreased bone mineral density (BMD) in untreated HIV infection. Paradoxically, antiretroviral therapy (ART) worsens bone loss although existing data suggest that such loss is largely independent of specific antiretroviral regimen. This led us to hypothesize that skeletal deterioration following HIV disease reversal with ART may be related to T-cell repopulation and/or immune reconstitution. Here we transplant T cells into immunocompromised mice to mimic ART-induced T-cell expansion. T-cell reconstitution elicits RANKL and TNFα production by B cells and/or T cells, accompanied by enhanced bone resorption and BMD loss. Reconstitution of TNFα- or RANKL-null T-cells and pharmacological TNFα antagonist all protect cortical, but not trabecular bone, revealing complex effects of T-cell reconstitution on bone turnover. These findings suggest T-cell repopulation and/or immune reconstitution as putative mechanisms for bone loss following ART initiation.


Assuntos
Fármacos Anti-HIV/efeitos adversos , HIV-1 , Osteoporose/induzido quimicamente , Linfócitos T/fisiologia , Transferência Adotiva , Animais , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/imunologia , Reabsorção Óssea , Contagem de Linfócito CD4 , Feminino , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
PLoS One ; 10(4): e0121568, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25860662

RESUMO

Osteosarcoma (OS) is a hyperproliferative malignant tumor that requires a high vascular density to maintain its large volume. Vascular Endothelial Growth Factor (VEGF) plays a crucial role in angiogenesis and acts as a paracrine and autocrine agent affecting both endothelial and tumor cells. The alpha-Ca2+/Calmodulin kinase two (α-CaMKII) protein is an important regulator of OS growth. Here, we investigate the role of α-CaMKII-induced VEGF in the growth and tumorigenicity of OS. We show that the pharmacologic and genetic inhibition of α-CaMKII results in decreases in VEGF gene expression (50%) and protein secretion (55%), while α- CaMKII overexpression increases VEGF gene expression (250%) and protein secretion (1,200%). We show that aggressive OS cells (143B) express high levels of VEGF receptor 2 (VEGFR-2) and respond to exogenous VEGF (100nm) by increasing intracellular calcium (30%). This response is ameliorated by the VEGFR inhibitor CBO-P11, suggesting that secreted VEGF results in autocrine stimulated α-CaMKII activation. Furthermore, we show that VEGF and α-CaMKII inhibition decreases the transactivation of the HIF-1α and AP-1 reporter constructs. Additionally, chromatin immunoprecipitation assay shows significantly decreased binding of HIF-1α and AP-1 to their responsive elements in the VEGF promoter. These data suggest that α-CaMKII regulates VEGF transcription by controlling HIF-1α and AP-1 transcriptional activities. Finally, CBO-P11, KN-93 (CaMKII inhibitor) and combination therapy significantly reduced tumor burden in vivo. Our results suggest that VEGF-induced OS tumor growth is controlled by CaMKII and dual therapy by CaMKII and VEGF inhibitors could be a promising therapy against this devastating adolescent disease.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Benzilaminas/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fatores de Crescimento Endotelial/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Invasividade Neoplásica , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Peptídeos Cíclicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Ativação Transcricional/efeitos dos fármacos , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
PLoS One ; 9(8): e104987, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25122124

RESUMO

Snail transcription factor is up-regulated in several cancers and associated with increased tumor migration and invasion via induction of epithelial-to-mesenchymal transition (EMT). MAPK (ERK1/2) signaling regulates cellular processes including cell motility, adhesion, and invasion. We investigated the regulation of ERK1/2 by Snail in breast cancer cells. ERK1/2 activity (p-ERK) was higher in breast cancer patient tissue as compared to normal tissue. Snail and p-ERK were increased in several breast cancer cell lines as compared to normal mammary epithelial cells. Snail knockdown in MDA-MB-231 and T47-D breast cancer cells decreased or re-localized p-ERK from the nuclear compartment to the cytoplasm. Snail overexpression in MCF-7 breast cancer cells induced EMT, increased cell migration, decreased cell adhesion and also increased tumorigenicity. Snail induced nuclear translocation of p-ERK, and the activation of its subcellular downstream effector, Elk-1. Inhibiting MAPK activity with UO126 or knockdown of ERK2 isoform with siRNA in MCF-7 Snail cells reverted EMT induced by Snail as shown by decreased Snail and vimentin expression, decreased cell migration and increased cell adhesion. Overall, our data suggest that ERK2 isoform activation by Snail in aggressive breast cancer cells leads to EMT associated with increased cell migration and decreased cell adhesion. This regulation is enhanced by positive feedback regulation of Snail by ERK2. Therefore, therapeutic targeting of ERK2 isoform may be beneficial for breast cancer.


Assuntos
Neoplasias da Mama/patologia , Núcleo Celular/enzimologia , Transição Epitelial-Mesenquimal/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fatores de Transcrição/fisiologia , Animais , Neoplasias da Mama/metabolismo , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico , Fatores de Transcrição da Família Snail
7.
Arthritis Rheumatol ; 66(4): 990-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24757150

RESUMO

OBJECTIVE: Rheumatoid arthritis (RA) is an inflammatory autoimmune disease characterized by severe joint erosion and systemic osteoporosis. Chronic T cell activation is a hallmark of RA, and agents that target the CD28 receptor on T cells, which is required for T cell activation, are being increasingly used as therapies for RA and other inflammatory diseases. Lymphocytes play complex roles in the regulation of the skeleton, and although activated T cells and B cells secrete cytokines that promote skeletal decline, under physiologic conditions lymphocytes also have key protective roles in the stabilization of skeletal mass. Consequently, disruption of T cell costimulation may have unforeseen consequences for physiologic bone turnover. This study was undertaken to investigate the impact of pharmacologic CD28 T cell costimulation blockade on physiologic bone turnover and structure. METHODS: C57BL6 mice were treated with CTLA-4Ig, a pharmacologic CD28 antagonist or with irrelevant control antibody (Ig), and serum biochemical markers of bone turnover were quantified by enzyme-linked immunosorbent assay. Bone mineral density and indices of bone structure were further measured by dual x-ray absorptiometry and micro-computed tomography, respectively, and static and dynamic indices of bone formation were quantified using bone histomorphometry. RESULTS: Pharmacologic disruption of CD28 T cell costimulation in mice significantly increased bone mass and enhanced indices of bone structure, a consequence of enhanced bone formation, concurrent with enhanced secretion of the bone anabolic factor Wnt-10b by T cells. CONCLUSION: Inhibition of CD28 costimulation by CTLA-4Ig promotes T cell Wnt-10b production and bone formation and may represent a novel anabolic strategy for increasing bone mass in osteoporotic conditions.


Assuntos
Artrite Reumatoide/metabolismo , Densidade Óssea/fisiologia , Imunoconjugados/farmacologia , Osteogênese/fisiologia , Linfócitos T/metabolismo , Proteínas Wnt/metabolismo , Abatacepte , Animais , Artrite Reumatoide/imunologia , Densidade Óssea/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Osteogênese/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
8.
Endocr Relat Cancer ; 21(2): 311-26, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24478054

RESUMO

Prostate cancer (PCa) metastasis to bone is lethal and there is no adequate animal model for studying the mechanisms underlying the metastatic process. Here, we report that receptor activator of NF-κB ligand (RANKL) expressed by PCa cells consistently induced colonization or metastasis to bone in animal models. RANK-mediated signaling established a premetastatic niche through a feed-forward loop, involving the induction of RANKL and c-Met, but repression of androgen receptor (AR) expression and AR signaling pathways. Site-directed mutagenesis and transcription factor (TF) deletion/interference assays identified common TF complexes, c-Myc/Max, and AP4 as critical regulatory nodes. RANKL-RANK signaling activated a number of master regulator TFs that control the epithelial-to-mesenchymal transition (Twist1, Slug, Zeb1, and Zeb2), stem cell properties (Sox2, Myc, Oct3/4, and Nanog), neuroendocrine differentiation (Sox9, HIF1α, and FoxA2), and osteomimicry (c-Myc/Max, Sox2, Sox9, HIF1α, and Runx2). Abrogating RANK or its downstream c-Myc/Max or c-Met signaling network minimized or abolished skeletal metastasis in mice. RANKL-expressing LNCaP cells recruited and induced neighboring non metastatic LNCaP cells to express RANKL, c-Met/activated c-Met, while downregulating AR expression. These initially non-metastatic cells, once retrieved from the tumors, acquired the potential to colonize and grow in bone. These findings identify a novel mechanism of tumor growth in bone that involves tumor cell reprogramming via RANK-RANKL signaling, as well as a form of signal amplification that mediates recruitment and stable transformation of non-metastatic bystander dormant cells.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-met/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Transdução de Sinais , Análise Serial de Tecidos
9.
Clin Dev Immunol ; 2013: 107321, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023563

RESUMO

Osteoblasts support hematopoietic cell development, including B lymphopoiesis. We have previously shown that the nuclear factor of activated T cells (NFAT) negatively regulates osteoblast differentiation and bone formation. Interestingly, in smooth muscle, NFAT has been shown to regulate the expression of vascular cellular adhesion molecule-1 (VCAM-1), a mediator of cell adhesion and signaling during leukocyte development. To examine whether NFAT signaling in osteoblasts regulates hematopoietic development in vivo, we generated a mouse model expressing dominant-negative NFAT driven by the 2.3 kb fragment of the collagen-αI promoter to disrupt NFAT activity in osteoblasts (dnNFAT(OB)). Bone histomorphometry showed that dnNFAT(OB) mice have significant increases in bone volume (44%) and mineral apposition rate (131%) and decreased trabecular thickness (18%). In the bone microenvironment, dnNFAT(OB) mice displayed a significant increase (87%) in Lineage(-)cKit(+)Sca-1(+) (LSK) cells and significant decreases in B220(+)CD19(-)IgM(-) pre-pro-B cells (41%) and B220(+)CD19(+)IgM(+) immature B cells (40%). Concurrent with these findings, LSK cell differentiation into B220(+) cells was inhibited when cocultured on differentiated primary osteoblasts harvested from dnNFAT(OB) mice. Gene expression and protein levels of VCAM-1 in osteoblasts decreased in dnNFAT(OB) mice compared to controls. These data suggest that osteoblast-specific NFAT activity mediates early B lymphopoiesis, possibly by regulating VCAM-1 expression on osteoblasts.


Assuntos
Osso e Ossos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoblastos/metabolismo , Transdução de Sinais , Nicho de Células-Tronco/fisiologia , Animais , Diferenciação Celular , Microambiente Celular/fisiologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFATC/genética , Osteoblastos/citologia , Osteogênese/fisiologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
10.
PLoS One ; 8(7): e68366, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874600

RESUMO

BACKGROUND: Bone metastasis is the most lethal form of several cancers. The ß2-microglobulin (ß2-M)/hemochromatosis (HFE) complex plays an important role in cancer development and bone metastasis. We demonstrated previously that overexpression of ß2-M in prostate, breast, lung and renal cancer leads to increased bone metastasis in mouse models. Therefore, we hypothesized that ß2-M is a rational target to treat prostate cancer bone metastasis. RESULTS: In this study, we demonstrate the role of ß2-M and its binding partner, HFE, in modulating radiation sensitivity and chemo-sensitivity of prostate cancer. By genetic deletion of ß2-M or HFE or using an anti-ß2-M antibody (Ab), we demonstrate that prostate cancer cells are sensitive to radiation in vitro and in vivo. Inhibition of ß2-M or HFE sensitized prostate cancer cells to radiation by increasing iron and reactive oxygen species and decreasing DNA repair and stress response proteins. Using xenograft mouse model, we demonstrate that anti-ß2-M Ab sensitizes prostate cancer cells to radiation treatment. Additionally, anti-ß2-M Ab was able to prevent tumor growth in an immunocompetent spontaneous prostate cancer mouse model. Since bone metastasis is lethal, we used a bone xenograft model to test the ability of anti-ß2-M Ab and radiation to block tumor growth in the bone. Combination treatment significantly prevented tumor growth in the bone xenograft model by inhibiting ß2-M and inducing iron overload. In addition to radiation sensitive effects, inhibition of ß2-M sensitized prostate cancer cells to chemotherapeutic agents. CONCLUSION: Since prostate cancer bone metastatic patients have high ß2-M in the tumor tissue and in the secreted form, targeting ß2-M with anti-ß2-M Ab is a promising therapeutic agent. Additionally, inhibition of ß2-M sensitizes cancer cells to clinically used therapies such as radiation by inducing iron overload and decreasing DNA repair enzymes.


Assuntos
Anticorpos/farmacologia , Sobrecarga de Ferro/induzido quimicamente , Proteínas de Membrana/antagonistas & inibidores , Neoplasias da Próstata/terapia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Microglobulina beta-2/antagonistas & inibidores , Animais , Anticorpos/uso terapêutico , Terapia Combinada , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Masculino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/imunologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Tolerância a Radiação/genética , Radiossensibilizantes/uso terapêutico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Microglobulina beta-2/imunologia , Microglobulina beta-2/metabolismo
11.
Obesity (Silver Spring) ; 21(7): 1459-66, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23512354

RESUMO

OBJECTIVE: To investigate the risk of bone fracture sustained by obese children exposed to falls. The bone fracture risk of obese children would be greater than that of their nonobese counterparts was hypothesized. DESIGN AND METHODS: Finite element-based computational models for children that reflected various levels of obesity by varying body mass and the thickness of the subcutaneous adipose tissue layer was developed. The models took account of both the momentum effect of variation of body mass and the cushion effect of variation of soft tissue thickness and examined these two contradictory effects on pelvic bone fracture risk through a set of sideways fall simulations with a range of impact speeds. RESULTS: The critical impact speed that yielded pelvic bone fracture decreased as the levels of obesity increased, which meant that the momentum effect of a greater body mass took precedence over the cushion effect of the soft tissue layer. CONCLUSIONS: The result suggests that obese children have a greater risk of pelvic bone fracture than do their nonobese counterparts in sideways falls. A further implication is that current child safety devices, systems, and regulations will need to be revisited as the prevalence of child obesity increases.


Assuntos
Fraturas Ósseas/epidemiologia , Obesidade Infantil/epidemiologia , Tecido Adiposo/fisiologia , Índice de Massa Corporal , Criança , Estudos de Coortes , Simulação por Computador , Fraturas Ósseas/fisiopatologia , Humanos , Músculo Esquelético/fisiologia , Obesidade Infantil/fisiopatologia , Ossos Pélvicos/lesões , Projetos Piloto , Reprodutibilidade dos Testes , Fatores de Risco
12.
Mol Cancer Res ; 11(4): 349-59, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23364534

RESUMO

Osteosarcoma is among the most frequently occurring primary bone tumors, primarily affecting adolescents and young adults. Despite improvements in osteosarcoma treatment, more specific molecular targets are needed as potential therapeutic options. One target of interest is α-Ca(2+)/calmodulin-dependent protein kinase II (α-CaMKII), a ubiquitous mediator of Ca(2+)-linked signaling, which has been shown to regulate tumor cell proliferation and differentiation. Here, we investigate the role of α-CaMKII in the growth and tumorigenicity of human osteosarcoma. We show that α-CaMKII is highly expressed in primary osteosarcoma tissue derived from 114 patients, and is expressed in varying levels in different human osteosarcoma (OS) cell lines [MG-63, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)/HOS, and 143B). To examine whether α-CaMKII regulates osteosarcoma tumorigenic properties, we genetically inhibited α-CaMKII in two osteosarcoma cell lines using two different α-CaMKII shRNAs delivered by lentiviral vectors and overexpressed α-CaMKII by retrovirus. The genetic deletion of α-CaMKII by short hairpin RNA (shRNA) in MG-63 and 143B cells resulted in decreased proliferation (50% and 41%), migration (22% and 25%), and invasion (95% and 90%), respectively. The overexpression of α-CaMKII in HOS cells resulted in increased proliferation (240%), migration (640%), and invasion (10,000%). Furthermore, α-CaMKII deletion in MG-63 cells significantly reduced tumor burden in vivo (65%), whereas α-CaMKII overexpression resulted in tumor formation in a previously nontumor forming osteosarcoma cell line (HOS). Our results suggest that α-CaMKII plays a critical role in determining the aggressive phenotype of osteosarcoma, and its inhibition could be an attractive therapeutic target to combat this devastating adolescent disease.


Assuntos
Neoplasias Ósseas/enzimologia , Neoplasias Ósseas/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Animais , Neoplasias Ósseas/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Osteossarcoma/genética , Fosforilação , Transdução de Sinais
13.
Biochem Biophys Res Commun ; 422(3): 488-93, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22583901

RESUMO

Transforming growth factor-ß (TGF-ß) is a critical regulator of bone development and remodeling. TGF-ß must be activated from its latent form in order to signal. Thrombospondin-1 (TSP1) is a major regulator of latent TGF-ß activation and TSP1 control of TGF-ß activation is critical for regulation of TGF-ß activity in multiple diseases. Bone marrow-derived mesenchymal stem cells (MSCs) have osteogenic potential and they participate in bone remodeling in injury and in response to tumor metastasis. Since both TSP1 and TGF-ß inhibit osteoblast differentiation, we asked whether TSP1 blocks osteoblast differentiation of MSCs through its ability to stimulate TGF-ß activation. TSP1 added to human bone marrow-derived MSCs under growth conditions increases active TGF-ß. Cultured MSCs express TSP1 and both TSP1 expression and TGF-ß activity decrease during osteoblast differentiation. TSP1 and active TGF-ß block osteoblast differentiation of MSCs grown in osteogenic media as measured by decreased Runx2 and alkaline phosphatase expression. The inhibitory effect of TSP1 on osteoblast differentiation is due to its ability to activate latent TGF-ß, since a peptide which blocks TSP1 TGF-ß activation reduced TGF-ß activity and restored osteoblast differentiation as measured by increased Runx2 and alkaline phosphatase expression. Anti-TGF-ß neutralizing antibody also increased alkaline phosphatase expression in the presence of TSP1. These studies show that TSP1 regulated TGF-ß activity is a critical determinant of osteoblast differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Trombospondina 1/fisiologia , Fator de Crescimento Transformador beta/agonistas , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/química , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Peptídeos/farmacologia , Trombospondina 1/análise , Trombospondina 1/farmacologia , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/metabolismo
14.
PLoS One ; 6(11): e27720, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110740

RESUMO

LIV-1, a zinc transporter, is an effector molecule downstream from soluble growth factors. This protein has been shown to promote epithelial-to-mesenchymal transition (EMT) in human pancreatic, breast, and prostate cancer cells. Despite the implication of LIV-1 in cancer growth and metastasis, there has been no study to determine the role of LIV-1 in prostate cancer progression. Moreover, there was no clear delineation of the molecular mechanism underlying LIV-1 function in cancer cells. In the present communication, we found increased LIV-1 expression in benign, PIN, primary and bone metastatic human prostate cancer. We characterized the mechanism by which LIV-1 drives human prostate cancer EMT in an androgen-refractory prostate cancer cells (ARCaP) prostate cancer bone metastasis model. LIV-1, when overexpressed in ARCaP(E) (derivative cells of ARCaP with epithelial phenotype) cells, promoted EMT irreversibly. LIV-1 overexpressed ARCaP(E) cells had elevated levels of HB-EGF and matrix metalloproteinase (MMP) 2 and MMP 9 proteolytic enzyme activities, without affecting intracellular zinc concentration. The activation of MMPs resulted in the shedding of heparin binding-epidermal growth factor (HB-EGF) from ARCaP(E) cells that elicited constitutive epidermal growth factor receptor (EGFR) phosphorylation and its downstream extracellular signal regulated kinase (ERK) signaling. These results suggest that LIV-1 is involved in prostate cancer progression as an intracellular target of growth factor receptor signaling which promoted EMT and cancer metastasis. LIV-1 could be an attractive therapeutic target for the eradication of pre-existing human prostate cancer and bone and soft tissue metastases.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Animais , Anticorpos/imunologia , Neoplasias Ósseas/secundário , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/imunologia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Neoplasias da Próstata/genética , Neoplasias de Tecidos Moles/secundário
15.
Am J Clin Pathol ; 136(2): 239-46, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21757596

RESUMO

Our aim was to compare the usefulness of fatty acid synthase (FASn) with that of α-methylacyl coenzyme-A racemase (AMACR) in the diagnosis of prostatic adenocarcinoma. The expression of these 2 markers was compared in a tissue microarray containing 62 foci of benign glands and 36 foci of prostatic adenocarcinoma. Similar to AMACR, there was significantly higher FASn expression in adenocarcinoma compared with that in benign glands. The optimal accuracy rate and area under curve (AUC) by receiver operating characteristic analysis for FASn were not significantly different from those for AMACR (accuracy, 80% vs 87%; AUC, 0.942 vs 0.956; P for both, > .05). Moreover, in cases with coexistent malignant and benign glands on the same core, FASn could selectively distinguish a proportion of cases (17/21 [81%]) similar to using AMACR. We conclude that FASn may aid in the diagnosis of prostatic adenocarcinoma, at least to supplement AMACR as another positive marker of carcinoma and potentially increase diagnostic accuracy.


Assuntos
Adenocarcinoma/diagnóstico , Biomarcadores Tumorais/análise , Ácido Graxo Sintases/análise , Neoplasias da Próstata/diagnóstico , Adenocarcinoma/enzimologia , Área Sob a Curva , Ácido Graxo Sintases/biossíntese , Humanos , Masculino , Neoplasias da Próstata/enzimologia , Curva ROC , Racemases e Epimerases/análise , Racemases e Epimerases/biossíntese , Sensibilidade e Especificidade , Análise Serial de Tecidos
16.
Clin Cancer Res ; 17(8): 2159-69, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21355075

RESUMO

PURPOSE: Prostate tumor cells frequently show the features of osteoblasts, which are differentiated from bone marrow mesenchymal stem cells. We examined human prostate cancer cell lines and clinical prostate cancer specimens for additional bone marrow mesenchymal stem cell properties. EXPERIMENTAL DESIGN: Prostate cancer cell lines were induced for osteoblastogenic and adipogenic differentiation, detected by standard staining methods and confirmed by lineage-specific marker expression. Abnormal expression of the markers was then assessed in clinical prostate cancer specimens. RESULTS: After osteoblastogenic induction, cells of the LNCaP lineage, PC-3 lineage, and DU145 displayed osteoblastic features. Upon adipogenic induction, PC-3 lineage and DU145 cells differentiated into adipocyte-like cells. The adipocyte-like cancer cells expressed brown adipocyte-specific markers, suggesting differentiation along the brown adipocyte lineage. The adipogenic differentiation was accompanied by growth inhibition, and most of the adipocyte-like cancer cells were committed to apoptotic death. During cyclic treatments with adipogenic differentiation medium and then with control medium, the cancer cells could commit to repeated adipogenic differentiation and retrodifferentiation. In clinical prostate cancer specimens, the expression of uncoupling protein 1 (UCP1), a brown fat-specific marker, was enhanced with the level of expression correlated to disease progression from primary to bone metastatic cancers. CONCLUSIONS: This study thus revealed that prostate cancer cells harbor the stem cell properties of bone marrow mesenchymal stem cells. The abnormally expressed adipogenic UCP1 protein may serve as a unique marker, while adipogenic induction can be explored as a differentiation therapy for prostate cancer progression and bone metastasis.


Assuntos
Células da Medula Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Linhagem da Célula , Meios de Cultura/química , Meios de Cultura/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Proteína Desacopladora 1
17.
Cancer Res ; 71(7): 2600-10, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21427356

RESUMO

Bone metastasis is one of the predominant causes of cancer lethality. This study demonstrates for the first time how ß2-microglobulin (ß2-M) supports lethal metastasis in vivo in human prostate, breast, lung, and renal cancer cells. ß2-M mediates this process by activating epithelial to mesenchymal transition (EMT) to promote lethal bone and soft tissue metastases in host mice. ß2-M interacts with its receptor, hemochromatosis (HFE) protein, to modulate iron responsive pathways in cancer cells. Inhibition of either ß2-M or HFE results in reversion of EMT. These results demonstrate the role of ß2-M in cancer metastasis and lethality. Thus, ß2-M and its downstream signaling pathways are promising prognostic markers of cancer metastases and novel therapeutic targets for cancer therapy.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias/metabolismo , Neoplasias/patologia , Microglobulina beta-2/metabolismo , Animais , Neoplasias Ósseas/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Técnicas de Silenciamento de Genes , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Hospedeiro Imunocomprometido , Imuno-Histoquímica , Ferro/metabolismo , Neoplasias Renais/imunologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Neoplasias/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transplante Heterólogo , Microglobulina beta-2/antagonistas & inibidores , Microglobulina beta-2/biossíntese , Microglobulina beta-2/imunologia
18.
Proc Natl Acad Sci U S A ; 108(2): 768-73, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187391

RESUMO

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


Assuntos
Ligante de CD40/metabolismo , Osteoblastos/citologia , Osteoclastos/citologia , Linfócitos T/citologia , Animais , Técnicas de Cocultura , Estrogênios/metabolismo , Humanos , Ligantes , Camundongos , NF-kappa B/metabolismo , Osteoporose/metabolismo , Osteoprotegerina/metabolismo , Ovariectomia/métodos , Fator de Necrose Tumoral alfa/metabolismo
19.
Biochem Biophys Res Commun ; 404(1): 34-9, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21093414

RESUMO

Reactive oxygen species increases in various diseases including cancer and has been associated with induction of epithelial-mesenchymal transition (EMT), as evidenced by decrease in cell adhesion-associated molecules like E-cadherin, and increase in mesenchymal markers like vimentin. We investigated the molecular mechanisms by which Snail transcription factor, an inducer of EMT, promotes tumor aggressiveness utilizing ARCaP prostate cancer cell line. An EMT model created by Snail overexpression in ARCaP cells was associated with decreased E-cadherin and increased vimentin. Moreover, Snail-expressing cells displayed increased concentration of reactive oxygen species (ROS), specifically, superoxide and hydrogen peroxide, in vitro and in vivo. Real Time PCR profiling demonstrated increased expression of oxidative stress-responsive genes, such as aldehyde oxidase I, in response to Snail. The ROS scavenger, N-acetyl cysteine partially reversed Snail-mediated EMT after 7 days characterized by increased E-cadherin levels and decreased ERK activity, while treatment with the MEK inhibitor, UO126, resulted in a more marked effect by 3 days, characterized by cells returning back to the epithelial morphology and increased E-cadherin. In conclusion, this study shows for the first time that Snail transcription factor can regulate oxidative stress enzymes and increase ROS-mediated EMT regulated in part by ERK activation. Therefore, Snail may be an attractive molecule for therapeutic targeting to prevent tumor progression in human prostate cancer.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Estresse Oxidativo/genética , Neoplasias da Próstata/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Transfecção
20.
Proc Natl Acad Sci U S A ; 107(31): 13848-53, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20643942

RESUMO

Osteoporosis and bone fractures are increasingly recognized complications of HIV-1 infection. Although antiretroviral therapy itself has complex effects on bone turnover, it is now evident that the majority of HIV-infected individuals already exhibit reduced bone mineral density before therapy. The mechanisms responsible are likely multifactorial and have been difficult to delineate in humans. The HIV-1 transgenic rat recapitulates many key features of human AIDS. We now demonstrate that, like their human counterparts, HIV-1 transgenic rats undergo severe osteoclastic bone resorption, a consequence of an imbalance in the ratio of receptor activator of NF-kappaB ligand, the key osteoclastogenic cytokine, to that of its physiological decoy receptor osteoprotegerin. This imbalance stemmed from a switch in production of osteoprotegerin to that of receptor activator of NF-kappaB ligand by B cells, and was further compounded by a significantly elevated number of osteoclast precursors. With the advancing age of individuals living with HIV/AIDS, low bone mineral density associated with HIV infection is likely to collide with the pathophysiology of skeletal aging, leading to increased fracture risk. Understanding the mechanisms driving bone loss in HIV-infected individuals will be critical to developing effective therapeutic strategies.


Assuntos
Reabsorção Óssea/imunologia , HIV-1/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Densidade Óssea , Reabsorção Óssea/metabolismo , Reabsorção Óssea/fisiopatologia , Masculino , Ligante RANK/biossíntese , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...