Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Sci Rep ; 13(1): 17067, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37816950

RESUMO

Humanized mice have been used to study human immunodeficiency virus type 1 (HIV-1) transmission, pathogenesis, and treatment. The ability of pediatric thymus tissue implanted either in the leg (Leg PedThy) or under the renal capsule (Renal PedThy) with allogeneic CD34+ hematopoietic cells (HSCs) in NSG mice was evaluated for reconstitution of human immune cells and for rectal transmission of HIV-1. These mice were compared to traditional BLT mice implanted with fetal liver and thymus under the renal capsule and mice injected only with HSCs. Renal PedThy mice had similar immune reconstitution in the blood, spleen and intestine as BLT mice, while Leg PedThy mice had transient detection of immune cells, particularly CD4+ T cells and macrophages, the target cells for HIV-1 infection. Rectal transmission and replication of HIV-1 was efficient in BLT mice but lower and more variable in Renal PedThy mice. HIV-1 was poorly transmitted in HSC mice and not transmitted in Leg PedThy mice, which correlated with the frequencies of target cells in the spleen and intestine. Humanization of NSG mice with pediatric thymus was successful when implanted under the kidney capsule, but led to less efficient HIV-1 rectal transmission and replication compared to BLT mice.


Assuntos
Infecções por HIV , HIV-1 , Camundongos , Humanos , Animais , Criança , Modelos Animais de Doenças , Timo/patologia , Linfócitos T CD4-Positivos , Camundongos SCID , Camundongos Endogâmicos NOD
2.
Viruses ; 14(9)2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36146831

RESUMO

While antiretroviral drugs have transformed the lives of HIV-infected individuals, chronic treatment is required to prevent rebound from viral reservoir cells. People living with HIV also are at higher risk for cardiovascular and neurocognitive complications, as well as cancer. Finding a cure for HIV-1 infection is therefore an essential goal of current AIDS research. This review is focused on the discovery of pharmacological inhibitors of the HIV-1 Nef accessory protein. Nef is well known to enhance HIV-1 infectivity and replication, and to promote immune escape of HIV-infected cells by preventing cell surface MHC-I display of HIV-1 antigens. Recent progress shows that Nef inhibitors not only suppress HIV-1 replication, but also restore sufficient MHC-I to the surface of infected cells to trigger a cytotoxic T lymphocyte response. Combining Nef inhibitors with latency reversal agents and therapeutic vaccines may provide a path to clearance of viral reservoirs.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Antirretrovirais/uso terapêutico , Descoberta de Drogas , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Humanos , Fatores de Virulência , Produtos do Gene nef do Vírus da Imunodeficiência Humana
3.
Bio Protoc ; 11(13): e4068, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34327265

RESUMO

Non-receptor protein-tyrosine kinases regulate cellular responses to many external signals and are important drug discovery targets for cancer and infectious diseases. While many assays exist for the assessment of kinase activity in vitro, methods that report changes in tyrosine kinase activity in single cells have the potential to provide information about kinase responses at the cell population level. In this protocol, we combined bimolecular fluorescence complementation (BiFC), an established method for the assessment of protein-protein interactions, and immunofluorescence staining with phosphospecific antibodies to characterize changes in host cell tyrosine kinase activity in the presence of an HIV-1 virulence factor, Nef. Specifically, two Tec family kinases (Itk and Btk) as well as Nef were fused to complementary, non-fluorescent fragments of the Venus variant of YFP. Each kinase was expressed in 293T cells in the presence or absence of Nef and immunostained for protein expression and activity with anti-phosphotyrosine (pTyr) antibodies. Multi-color confocal microscopy revealed the interaction of Nef with each kinase (BiFC), kinase activity, and kinase protein expression. Strong BiFC signals were observed when Nef was co-expressed with both Itk and Btk, indicative of interaction, and a strong anti-pTyr immunoreactivity was also seen. The BiFC, pTyr, and kinase expression signals co-localized to the plasma membrane, consistent with Nef-mediated kinase activation in this subcellular compartment. Image analysis allowed calculation of pTyr-to-kinase protein ratios, which showed a range of responses in individual cells across the population that shifted upward in the presence of Nef and back down in the presence of a kinase inhibitor. This method has the potential to reveal changes in steady-state non-receptor tyrosine kinase activity and subcellular localization in a cell population in response to other protein-kinase interactions, information that is not attainable from immunoblotting or other in vitro methods.

4.
Leuk Lymphoma ; 61(2): 409-419, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31592701

RESUMO

Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1α (MIP-1α) are important factors that increase bone resorption and hypercalcemia in adult T-cell leukemia (ATL). We investigated the role of PTHrP and MIP-1α in the development of local osteolytic lesions in T-cell leukemia through overexpression in Jurkat T-cells. Injections of Jurkat-PTHrP and Jurkat-MIP-1α into the tibia and the left ventricle of NSG mice were performed to evaluate tumor growth and metastasis in vivo. Jurkat-pcDNA tibial neoplasms grew at a significantly greater rate and total tibial tumor burden was significantly greater than Jurkat-PTHrP neoplasms. Despite the lower tibial tumor burden, Jurkat-PTHrP bone neoplasms had significantly greater osteolysis than Jurkat-pcDNA and Jurkat-MIP-1α neoplasms. Jurkat-PTHrP and Jurkat-pcDNA cells preferentially metastasized to bone following intracardiac injection, though the overall metastatic burden was lower in Jurkat-PTHrP mice. These findings demonstrate that PTHrP induced pathologic osteolysis in T-cell leukemia but did not increase the incidence of skeletal metastasis.


Assuntos
Neoplasias Ósseas , Hipercalcemia , Leucemia-Linfoma de Células T do Adulto , Osteólise , Adulto , Animais , Humanos , Hipercalcemia/etiologia , Camundongos , Osteólise/etiologia , Proteína Relacionada ao Hormônio Paratireóideo/genética
5.
ACS Chem Biol ; 13(6): 1551-1559, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29763550

RESUMO

Acute myelogenous leukemia (AML) is the most common hematologic malignancy in adults and is often associated with constitutive tyrosine kinase signaling. These pathways involve the nonreceptor tyrosine kinases Fes, Syk, and the three Src-family kinases expressed in myeloid cells (Fgr, Hck, and Lyn). In this study, we report remarkable anti-AML efficacy of an N-phenylbenzamide kinase inhibitor, TL02-59. This compound potently suppressed the proliferation of bone marrow samples from 20 of 26 AML patients, with a striking correlation between inhibitor sensitivity and expression levels of the myeloid Src family kinases Fgr, Hck, and Lyn. No correlation was observed with Flt3 expression or mutational status, with the four most sensitive patient samples being wild-type for Flt3. Kinome-wide target specificity profiling coupled with in vitro kinase assays demonstrated a narrow overall target specificity profile for TL02-59, with picomolar potency against the myeloid Src-family member Fgr. In a mouse xenograft model of AML, oral administration of TL02-59 for 3 weeks at 10 mg/kg completely eliminated leukemic cells from the spleen and peripheral blood while significantly reducing bone marrow engraftment. These results identify Fgr as a previously unrecognized kinase inhibitor target in AML and TL02-59 as a possible lead compound for clinical development in AML cases that overexpress this kinase independent of Flt3 mutations.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Quinazolinas/uso terapêutico , Quinases da Família src/antagonistas & inibidores , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Benzamidas/administração & dosagem , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-hck/antagonistas & inibidores , Quinazolinas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Bioorg Med Chem Lett ; 26(5): 1480-1484, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26852364

RESUMO

The HIV-1 Nef accessory factor enhances viral replication and promotes immune system evasion of HIV-infected cells, making it an attractive target for drug discovery. Recently we described a novel class of diphenylpyrazolodiazene compounds that bind directly to Nef in vitro and inhibit Nef-dependent HIV-1 infectivity and replication in cell culture. However, these first-generation Nef antagonists have several structural liabilities, including an azo linkage that led to poor oral bioavailability. The azo group was therefore replaced with either a one- or two-carbon linker. The resulting set of non-azo analogs retained nanomolar binding affinity for Nef by surface plasmon resonance, while inhibiting HIV-1 replication with micromolar potency in cell-based assays without cytotoxicity. Computational docking studies show that these non-azo analogs occupy the same predicted binding site within the HIV-1 Nef dimer interface as the original azo compound. Computational methods also identified a hot spot for inhibitor binding within this site that is defined by conserved HIV-1 Nef residues Asp108, Leu112, and Pro122. Pharmacokinetic evaluation of the non-azo B9 analogs in mice showed that replacement of the azo linkage dramatically enhanced oral bioavailability without substantially affecting plasma half-life or clearance. The improved oral bioavailability of non-azo diphenylpyrazolo Nef antagonists provides a starting point for further drug lead optimization in support of future efficacy testing in animal models of HIV/AIDS.


Assuntos
Fármacos Anti-HIV/síntese química , Fármacos Anti-HIV/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Administração Oral , Animais , Fármacos Anti-HIV/administração & dosagem , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , HIV-1/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Molecular , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/síntese química , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Replicação Viral/efeitos dos fármacos
7.
Chem Biol ; 20(1): 82-91, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23352142

RESUMO

HIV-1 Nef, a critical AIDS progression factor, represents an important target protein for antiretroviral drug discovery. Because Nef lacks intrinsic enzymatic activity, we developed an assay that couples Nef to the activation of Hck, a Src family member and Nef effector protein. Using this assay, we screened a large, diverse chemical library and identified small molecules that block Nef-dependent Hck activity with low micromolar potency. Of these, a diphenylpyrazolo compound demonstrated submicromolar potency in HIV-1 replication assays against a broad range of primary Nef variants. This compound binds directly to Nef via a pocket formed by the Nef dimerization interface and disrupts Nef dimerization in cells. Coupling of nonenzymatic viral accessory factors to host cell effector proteins amenable to high-throughput screening may represent a general strategy for the discovery of new antimicrobial agents.


Assuntos
Antirretrovirais/química , Antirretrovirais/farmacologia , HIV-1/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Linhagem Celular , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Humanos , Simulação de Acoplamento Molecular , Multimerização Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-hck/metabolismo , Pirazóis/química , Pirazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
8.
Oral Oncol ; 48(6): 491-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22265717

RESUMO

Oral squamous cell carcinoma (OSCC) is the most commonly diagnosed oral malignancy in humans and cats and frequently invades bone. The objective of this study was to determine if feline OSCC serves as a relevant model of human OSCC in terms of osteolytic behavior and expression of bone resorption agonists. Novel feline OSCC cell lines (SCCF2 and SCCF3) were derived from spontaneous carcinomas. Gene expression and osteolytic behavior were compared to an established feline OSCC cell line (SCCF1) and three human OSCC cell lines (UMSCC-12, A253 and SCC25). Interaction of OSCC with bone and murine pre-osteoblasts (MC3T3) was investigated using in vitro co-culture techniques. In vivo bioluminescent imaging, Faxitron radiography and microscopy were used to measure xenograft growth and bone invasion in nude mice. Human and feline OSCC expressing the highest levels of parathyroid hormone-related protein (PTHrP) were associated with in vitro and in vivo bone resorption and osteoclastogenesis. MC3T3 cells had increased receptor activator of nuclear factor κB ligand (RANKL) expression and reduced osteoprotegerin (OPG) expression in conditioned medium from bone-invasive SCCF2 cells compared to minimally bone invasive SCCF3 cells, which was partially reversed with a neutralizing anti-PTHrP antibody. Human and feline OSCC cells cultured in bone-conditioned medium had increased PTHrP secretion and proliferation. Feline OSCC-induced bone resorption was associated with tumor cell secretion of PTHrP and with increased RANKL:OPG expression ratio in mouse preosteoblasts. Bone-CM increased OSCC proliferation and secretion of PTHrP. The preclinical models of feline OSCC recapitulated the bone-invasive phenotype characteristic of spontaneous OSCC and will be useful to future preclinical and mechanistic studies of bone invasive behavior.


Assuntos
Reabsorção Óssea/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Bucais/metabolismo , Animais , Reabsorção Óssea/veterinária , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/veterinária , Gatos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias Bucais/patologia , Neoplasias Bucais/veterinária , Osteoprotegerina/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Ligante RANK/metabolismo
9.
Leuk Lymphoma ; 53(4): 688-98, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21942940

RESUMO

Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1α (MIP-1α) have been implicated in the pathogenesis of adult T-cell leukemia/lymphoma, but their effects on T-cells have not been well studied. Here we analyzed the functions of PTHrP and MIP-1α on T-cell growth and death both in vitro and in vivo by overexpressing either factor in human Jurkat T-cells. PTHrP or MIP-1α did not affect Jurkat cell growth in vitro, but PTHrP increased their sensitivity to apoptosis. Importantly, PTHrP and MIP-1α decreased both tumor incidence and growth in vivo. To investigate possible mechanisms, polymerase chain reaction (PCR) arrays and real-time reverse transcription (RT)-PCR assays were performed. Both PTHrP and MIP-1α increased the expression of several factors including signal transducer and activator of transcription 4, tumor necrosis factor α, receptor activator of nuclear factor κB ligand and death-associated protein kinase 1, and decreased the expression of inhibitor of DNA binding 1, interferon γ and CD40 ligand in Jurkat cells. In addition, MIP-1α also increased the expression of transcription factor AP-2α and PTHrP increased expression of the vitamin D3 receptor. These data demonstrate that PTHrP and MIP-1α exert a profound antitumor effect presumably by increasing the sensitivity to apoptotic signals through modulation of transcription and apoptosis factors in T-cells.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Quimiocina CCL3/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Experimental/genética , Proteína Relacionada ao Hormônio Paratireóideo/genética , Animais , Apoptose/genética , Ligante de CD40/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Linhagem Celular Tumoral , Proteínas Quinases Associadas com Morte Celular , Humanos , Interferon gama/genética , Células Jurkat , Leucemia Experimental/patologia , Leucemia de Células T/genética , Leucemia de Células T/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Ligante RANK/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT4/genética , Linfócitos T/metabolismo , Linfócitos T/patologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/genética
10.
Prostate ; 71(12): 1251-63, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21321976

RESUMO

BACKGROUND: Prostate cancer in men has a high mortality and morbidity due to metastatic disease. The pathobiology of prostate cancer metastasis is not well understood and cell lines and animal models that recapitulate the complex nature of the disease are needed. Therefore, the goal of the study was to establish and characterize a new prostate cancer line derived from a dog with spontaneous prostate cancer. METHODS: A new cell line (Leo) was derived from a dog with spontaneous prostate cancer. Immunohistochemistry and PCR were used to characterize the primary prostate cancer and xenografts in nude mice. Subcutaneous tumor growth and metastases in nude mice were evaluated by bioluminescent imaging, radiography and histopathology. In vitro chemosensitivity of Leo cells to therapeutic agents was measured. RESULTS: Leo cells expressed the secretory epithelial cytokeratins (CK)8, 18, and ductal cell marker, CK7. The cell line grew in vitro (over 75 passages) and was tumorigenic in the subcutis of nude mice. Following intracardiac injection, Leo cells metastasized to the brain, spinal cord, bone, and adrenal gland. The incidence of metastases was greatest to the central nervous system (80%) with a lower incidence to bone (20%) and the adrenal glands (16%). In vitro chemosensitivity assays demonstrated that Leo cells were sensitive to Velcade and an HDAC-42 inhibitor with IC(50) concentrations of 1.9 nm and 0.95 µm, respectively. CONCLUSION: The new prostate cancer cell line (Leo) will be a valuable model to investigate the mechanisms of the brain and bone metastases.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Carcinoma/patologia , Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias da Próstata/patologia , Neoplasias das Glândulas Suprarrenais/epidemiologia , Neoplasias das Glândulas Suprarrenais/secundário , Animais , Antineoplásicos/farmacologia , Neoplasias Ósseas/epidemiologia , Neoplasias Ósseas/secundário , Ácidos Borônicos/farmacologia , Bortezomib , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/metabolismo , Testes de Carcinogenicidade , Carcinoma/epidemiologia , Carcinoma/metabolismo , Divisão Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/patologia , Cães , Imuno-Histoquímica , Incidência , Injeções Subcutâneas , Queratina-18/metabolismo , Queratina-7/metabolismo , Queratina-8/metabolismo , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias de Tecido Conjuntivo/secundário , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Fenilbutiratos/antagonistas & inibidores , Pirazinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Medula Espinal/epidemiologia , Neoplasias da Medula Espinal/secundário , Tela Subcutânea , Transplante Heterólogo
11.
Prostate ; 71(6): 615-25, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20957670

RESUMO

BACKGROUND: Osteoblastic bone metastasis is the predominant phenotype observed in prostate cancer patients and is associated with high patient mortality and morbidity. However, the mechanisms determining the development of this phenotype are not well understood. Prostate cancer cells secrete several osteogenic factors including Wnt proteins, which are not only osteoinductive but also oncogenic. Therefore, the purpose of the study was to investigate the contribution of the Wnt signaling pathway in prostate cancer growth, incidence of bone metastases, and osteoblastic phenotype of bone metastases. The strategy involved overexpressing the Wnt antagonist, DKK-1, in the mixed osteoblastic and osteolytic Ace-1 prostate cancer cells. METHODS: Ace-1 prostate cancer cells stably expressing human DKK-1 or empty vector were established and transduced with lentiviral yellow fluorescent protein (YFP)-luciferase (Luc). The Ace-1/vector(YFP-LUC) and Ace-1/DKK-1(YFP-LUC) cells were injected subcutaneously, intratibially, or in the left cardiac ventricle in athymic mice. RESULTS: Unexpectedly, DKK-1 significantly increased Ace-1 subcutaneous tumor mass and the incidence of bone metastases after intracardiac injection of Ace-1 cells. DKK-1 increased Ace-1 tumor growth associated with increased phospho46 c-Jun amino-terminal kinase by the Wnt noncanonical pathway. As expected, DKK-1 decreased the Ace-1 osteoblastic phenotype of bone metastases, as confirmed by radiographic, histopathologic, and microcomputed tomographic analysis. DKK-1 decreased osteoblastic activity via the Wnt canonical pathway evidenced by an inhibition of T-cell factor activity in murine osteoblast precursor ST2 cells. CONCLUSION: The present study showed that DKK-1 is a potent inhibitor of bone growth in prostate cancer-induced osteoblastic metastases.


Assuntos
Neoplasias Ósseas/secundário , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias da Próstata/patologia , Proteínas Wnt/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Cães , Histocitoquímica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Camundongos , Camundongos Nus , Osteogênese/fisiologia , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Estatísticas não Paramétricas , Tomografia Computadorizada por Raios X , Proteínas Wnt/antagonistas & inibidores
12.
Anticancer Res ; 30(6): 1937-46, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20651337

RESUMO

BACKGROUND: Protein phosphorylation is one of the essential steps in cell signaling, and aberrant phosphorylation is a common event in human cancer. The expression of receptor type protein tyrosine phosphatase gamma (PTPRG) in normal breast is found to be approximately 50-60% higher than that of breast tumor tissue. Overexpression of PTPRG inhibits anchorage-independent growth and proliferation of breast cancer cells. To understand the tumor suppression characteristics of PTPRG, we studied its tumor suppressive function in an athymic mouse model and evaluated factors that can potentially regulate its expression in breast cancer cells. MATERIALS AND METHODS: To investigate the function of PTPRG in vivo, athymic nude mice were implanted with MCF-7 cells overexpressing PTPRG. For in vitro study, protein levels of cell cycle regulators, cell cycle re-entry, and the phosphorylation levels of extracellular signal-regulated protein kinases 1/2 (ERK1/2) were examined. In addition, methylation assays were conducted to investigate the epigenetic modification on the promoter of PTPRG. RESULTS: Athymic nude mice bearing MCF-7 cells overexpressing PTPRG showed a reduction in tumor burden in comparison to animals implanted with MCF-7 cells transfected with vector alone. When these two cell lines were studied in an in vitro system, elevated mRNA and protein levels of cell cycle regulators, p21(cip) and p27(kip) were detected in MCF-7 cells overexpressing PTPRG compared to cells transfected with vector alone. Similarly, overexpression of PTPRG also delayed the re-entry of breast cancer cells into the cell cycle after serum starvation, and reduced the phosphorylation levels ERK1/2 in MCF-7 cells. In addition, methylation assays in PTPRG promoter in breast cancer cell lines (including SK-Br-3) revealed an aberrant methylation pattern. When SK-Br-3 and MCF-7 cells were treated with deoxy-5-azacytidine (DAC) and trichostatin A (TSA), these compounds reactivated the expression of PTPRG, suggesting an epigenetic control on its expression. CONCLUSION: Our results indicated that PTPRG inhibited breast tumor formation in vivo; PTPRG may up-regulate p21(cip) and p27(kip) proteins through the ERK1/2 pathway. This study also showed methylation-mediated silencing of PTPRG in breast cancer cell lines. These data indicate that PTPRG exhibits the characteristics of a breast tumor suppressor.


Assuntos
Neoplasias da Mama/prevenção & controle , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/fisiologia , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias da Mama/patologia , Ciclo Celular , Linhagem Celular Tumoral , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p21/análise , Inibidor de Quinase Dependente de Ciclina p27 , Metilação de DNA , Decitabina , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Ácidos Hidroxâmicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/análise , Camundongos , Fosforilação
13.
Int J Mol Med ; 26(1): 113-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20514430

RESUMO

Induction of apoptosis is one of the mechanisms of chemotherapeutic agents against breast cancer. In addition, recent studies have shown that diets containing polyphenolic components possess anticancer activities either in vitro or in vivo by inhibiting cell proliferation and inducing apoptosis. The aim of our study was to explore the effects of (-)-gossypol-enriched cottonseed oil [(-)-GPCSO], a polyphenolic compound, on the proliferation of the breast cancer cell line MCF-7 as well as primary cultured human breast cancer epithelial cells (PCHBCEC). We investigated whether the mechanism of the effects of (-)-GPCSO was mediated via the induction of cell apoptosis and the regulation of Bcl-2 gene expression at both the mRNA and protein levels. Our results showed that (-)-GPCSO inhibited the proliferation of MCF-7 and PCHBCEC in a dose-dependent manner. (-)-GPCSO (0.1 and 0.2%) induced DNA fragmentation in both MCF-7 cells and PCHBCEC. (-)-GPCSO suppressed the expression of Bcl-2 at both the mRNA and protein levels in MCF-7 cells and PCHBCEC in a dose-dependent fashion. Our results suggest that the growth inhibitory effect of (-)-GPCSO on MCF-7 and PCHBCEC is due, at least partially, to the induction of cell apoptosis, which is mediated by down-regulation of Bcl-2 expression at both the mRNA and protein levels. It might be possible for (-)-GPCSO to be developed as a novel chemotherapeutic agent for breast cancer patients.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Óleo de Sementes de Algodão/farmacologia , Gossipol/farmacologia , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Células Cultivadas , Óleo de Sementes de Algodão/química , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletroforese em Gel de Ágar , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Gossipol/química , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Leuk Lymphoma ; 51(4): 702-14, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20214446

RESUMO

Adult T-cell leukemia/lymphoma (ATLL) is caused by human T lymphotropic virus type 1 (HTLV-1). Patients with ATLL frequently develop humoral hypercalcemia of malignancy (HHM) resulting from increased osteoclastic bone resorption. Our goal was to investigate the mechanisms of ATLL-induced osteoclastic bone resorption. Murine calvaria co-cultured with HTLV-1-infected cells directly or conditioned media from cell cultures had increased osteoclast activity that was dependent on RANKL, indicating that factors secreted from ATLL cells had a stimulatory effect on bone resorption. Factors released from resorbing bone stimulated proliferation of HTLV-1-infected T-cells. Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1alpha (MIP-1alpha), both osteoclast stimulators, were expressed in HTLV-1-infected T-cell lines. Interestingly, when HTLV-1-infected T-cells were co-cultured with pre-osteoblasts, the expression of osteoprotegerin (OPG), an osteoclast inhibitory factor, was significantly down-regulated in the pre-osteoblasts. When OPG was added into the ex vivo osteoclastogenesis assay induced by HTLV-1-infected T-cells, osteoclastogenesis was strongly inhibited. In addition, HTLV-1-infected T-cells inhibited expression of early osteoblast genes and induced late genes. These regulators will serve as future therapeutic targets for the treatments of HHM in ATLL.


Assuntos
Reabsorção Óssea/complicações , Leucemia-Linfoma de Células T do Adulto/complicações , Adulto , Animais , Animais Recém-Nascidos , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Infecções por HTLV-I/complicações , Infecções por HTLV-I/genética , Infecções por HTLV-I/patologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Células Jurkat , Leucemia-Linfoma de Células T do Adulto/genética , Leucemia-Linfoma de Células T do Adulto/patologia , Leucemia-Linfoma de Células T do Adulto/virologia , Camundongos , Osteólise/complicações , Osteólise/genética , Osteólise/metabolismo
15.
FASEB J ; 24(6): 1947-57, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20145205

RESUMO

The functions of parathyroid hormone-related protein (PTHrP) on morphogenesis, cell proliferation, apoptosis, and calcium homeostasis have been attributed to its N terminus. Evidence suggests that many of these effects are not mediated by the N terminus but by the midregion, a nuclear localization sequence (NLS), and C terminus of the protein. A knock-in mouse lacking the midregion, NLS, and C terminus of PTHrP (Pthrp(Delta/Delta)) was developed. Pthrp(Delta/Delta) mice had craniofacial dysplasia, chondrodysplasia, and kyphosis, with most mice dying by d 5 of age. In bone, there were fewer chondrocytes and osteoblasts per area, bone mass was decreased, and the marrow was less cellular, with erythroid hypoplasia. Cellular proliferation was impaired, and apoptosis was increased. Runx2, Ocn, Sox9, Crtl1, beta-catenin, Runx1, ephrin B2, cyclin D1, and Gata1 were underexpressed while P16/Ink4a, P21, GSK-3beta, Il-6, Ffg3, and Ihh were overexpressed. Mammary gland development was aberrant, and energy metabolism was deregulated. These results establish that the midregion, NLS, and C terminus of PTHrP are crucial for the commitment of osteogenic and hematopoietic precursors to their lineages, and for survival, and many of the effects of PTHrP on development are not mediated by its N terminus. The down-regulation of Runx1, Runx2, and Sox9 indicates that PTHrP is a modulator of transcriptional activation during stem cell commitment.


Assuntos
Apoptose , Osso e Ossos/citologia , Genes Letais , Hematopoese , Sinais de Localização Nuclear/deficiência , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Animais , Western Blotting , Osso e Ossos/patologia , Condrócitos/citologia , Condrócitos/patologia , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Técnicas Imunoenzimáticas , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida
16.
J Biol Chem ; 284(7): 4658-66, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19059914

RESUMO

Interferon-gamma (IFN-gamma) has been shown to enhance anti-tumor immunity and inhibit the formation of bone-resorbing osteoclasts. We evaluated the role of IFN-gamma in bone metastases, tumor-associated bone destruction, and hypercalcemia in human T cell lymphotrophic virus type 1-Tax transgenic mice. Compared with Tax(+)IFN-gamma(+/+) mice, Tax(+)IFN-gamma(-/-) mice developed increased osteolytic bone lesions and soft tissue tumors, as well as increased osteoclast formation and activity. In vivo administration of IFN-gamma to tumor-bearing Tax(+)IFN-gamma(-/-) mice prevented new tumor development and resulted in decreased bromodeoxyuridine uptake by established tumors. In vitro, IFN-gamma directly decreased the viability of Tax(+) tumor cells through inhibition of proliferation, suppression of ERK phosphorylation, and induction of apoptosis and caspase 3 cleavage. IFN-gamma also inhibited macrophage colonystimulating factor-mediated proliferation and survival of osteoclast progenitors in vitro. Administration of IFN-gamma to C57BL/6 mice decreased Tax(+) tumor growth and prevented tumor-associated bone loss and hypercalcemia. In contrast, IFN-gamma treatment failed to protect IFN-gammaR1(-/-) mice from Tax(+) tumor-induced skeletal complications, despite decreasing tumor growth. These data demonstrate that IFN-gamma suppressed tumor-induced bone loss and hypercalcemia in Tax(+) mice by inhibiting both Tax(+) tumor cell growth and host-induced osteolysis. These data suggest a protective role for IFN-gamma in patients with bone metastases and hypercalcemia of malignancy.


Assuntos
Neoplasias Ósseas/metabolismo , Reabsorção Óssea/metabolismo , Vírus Linfotrópico T Tipo 1 Humano , Interferon gama/farmacologia , Osteoclastos/metabolismo , Neoplasias de Tecidos Moles/metabolismo , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Reabsorção Óssea/etiologia , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Produtos do Gene tax/genética , Produtos do Gene tax/metabolismo , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/etiologia , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hipercalcemia/patologia , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Camundongos Knockout , Metástase Neoplásica , Osteoclastos/patologia , Neoplasias de Tecidos Moles/complicações , Neoplasias de Tecidos Moles/tratamento farmacológico , Neoplasias de Tecidos Moles/patologia
17.
Retrovirology ; 5: 46, 2008 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-18541021

RESUMO

BACKGROUND: Adult T-cell leukemia/lymphoma (ATLL) is initiated by infection with human T-lymphotropic virus type-1 (HTLV-1); however, additional host factors are also required for T-cell transformation and development of ATLL. The HTLV-1 Tax protein plays an important role in the transformation of T-cells although the exact mechanisms remain unclear. Parathyroid hormone-related protein (PTHrP) plays an important role in the pathogenesis of humoral hypercalcemia of malignancy (HHM) that occurs in the majority of ATLL patients. However, PTHrP is also up-regulated in HTLV-1-carriers and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients without hypercalcemia, indicating that PTHrP is expressed before transformation of T-cells. The expression of PTHrP and the PTH/PTHrP receptor during immortalization or transformation of lymphocytes by HTLV-1 has not been investigated. RESULTS: We report that PTHrP was up-regulated during immortalization of lymphocytes from peripheral blood mononuclear cells by HTLV-1 infection in long-term co-culture assays. There was preferential utilization of the PTHrP-P2 promoter in the immortalized cells compared to the HTLV-1-transformed MT-2 cells. PTHrP expression did not correlate temporally with expression of HTLV-1 tax. HTLV-1 infection up-regulated the PTHrP receptor (PTH1R) in lymphocytes indicating a potential autocrine role for PTHrP. Furthermore, co-transfection of HTLV-1 expression plasmids and PTHrP P2/P3-promoter luciferase reporter plasmids demonstrated that HTLV-1 up-regulated PTHrP expression only mildly, indicating that other cellular factors and/or events are required for the very high PTHrP expression observed in ATLL cells. We also report that macrophage inflammatory protein-1alpha (MIP-1alpha), a cellular gene known to play an important role in the pathogenesis of HHM in ATLL patients, was highly expressed during early HTLV-1 infection indicating that, unlike PTHrP, its expression was enhanced due to activation of lymphocytes by HTLV-1 infection. CONCLUSION: These data demonstrate that PTHrP and its receptor are up-regulated specifically during immortalization of T-lymphocytes by HTLV-1 infection and may facilitate the transformation process.


Assuntos
Transformação Celular Viral , Vírus Linfotrópico T Tipo 1 Humano/crescimento & desenvolvimento , Leucócitos Mononucleares/virologia , Proteína Relacionada ao Hormônio Paratireóideo/biossíntese , Sobrevivência Celular , Células Cultivadas , Quimiocina CCL3/biossíntese , Técnicas de Cocultura , Produtos do Gene tax/biossíntese , Humanos , Receptor Tipo 1 de Hormônio Paratireóideo/biossíntese , Fatores de Tempo , Regulação para Cima
18.
Cancer Res ; 67(24): 11859-66, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18089816

RESUMO

Adult T-cell /lymphomaleukemia (ATLL) is caused by human T-cell lymphotropic virus type 1 (HTLV-1). Approximately 80% of ATLL patients develop humoral hypercalcemia of malignancy (HHM), a life-threatening complication leading to a poor prognosis. Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1 alpha (MIP-1 alpha) are important factors in the pathogenesis of HHM in ATLL and the expression of PTHrP can be activated by nuclear factor kappaB (NF-kappaB). NF-kappaB is constitutively activated in ATLL cells and is essential for leukemogenesis including transformation of lymphocytes infected by HTLV-1. Our goal was to evaluate the effects of NF-kappaB disruption by a proteasomal inhibitor (PS-341) and osteoclastic inhibition by zoledronic acid (Zol) on the development of ATLL and HHM using a novel bioluminescent mouse model. We found that PS-341 decreased cell viability, increased apoptosis, and down-regulated PTHrP expression in ATLL cells in vitro. To investigate the in vivo efficacy, nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice were xenografted with ATLL cells and treated with vehicle control, PS-341, Zol, or a combination of PS-341 and Zol. Bioluminescent imaging and tumor cell count showed a significant reduction in tumor burden in mice from all treatment groups. All treatments also significantly reduced the plasma calcium concentrations. Zol treatment increased trabecular bone volume and decreased osteoclast parameters. PS-341 reduced PTHrP and MIP-1 alpha expression in tumor cells in vivo. Our results indicate that both PS-341 and Zol are effective treatments for ATLL and HHM, which are refractory to conventional therapy.


Assuntos
Infecções por HTLV-I/tratamento farmacológico , Infecções por HTLV-I/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Ácidos Borônicos/uso terapêutico , Bortezomib , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Genes Reporter , Humanos , Células Jurkat , Luciferases/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Inibidores de Proteases/uso terapêutico , Pirazinas/uso terapêutico
19.
Anticancer Res ; 27(1A): 107-16, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17352222

RESUMO

BACKGROUND: Multidrug resistance (MDR) is a major impediment to successful cancer chemotherapy. P-glycoprotein (P-gp), the product of the multidrug resistance 1 (MDR1) gene, acts as an efflux pump and prevents sufficient intracellular accumulation of several anticancer agents, thus, playing a major role in MDR. Tamoxifen (Tam), ICI 182 780 (ICI) and Adriamycin (Adr) alone or with (-)-gossypol-enriched cottonseed oil [(-)-GPCSO] possible effects on cell growth inhibition and regulation of MDR1, mRNA and P-gp expression were examined in both an MDR human breast cancer cell line, MCF-7/Adr cells, and primary cultured human breast cancer epithelial cells (PCHBCEC). MATERIALS AND METHODS: Cells were treated with 0.05% of (-)-GPCSO either in the absence or presence of either 0.1 microM Tam, ICI or Adr for 24 h. RESULTS: Using the non-radioactive cell proliferation MTS assay, none of these chemotherapeutic agents alone inhibited MCF-7/Adr cell and PCHBCEC proliferation; meanwhile, the combination of 0.1 microM Tam, ICI or Adr with 0.05% (-)-GPCSO significantly reduced MCF-7/Adr cell growth by approximately 34%, 32% and 23%, respectively, of that of the vehicle-treated cells. For PCHBCEC, the combination of 0.05% (-)-GPCSO with 0.1 microM of Tam, ICI and Adr reduced cell growth to about 94%, 90%, and 71% respectively, of the vehicle treated PCHBCEC. Furthermore, (-)-GPCSO inhibited MDR1/P-gp expression in both MCF- 7/Adr and PCHBCEC in a dose-dependent manner. Our results provide insight into the MDR-reversing potential of (-)-GPCSO in human breast cancer cells resistant to current chemotherapeutic agents.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Óleo de Sementes de Algodão/farmacologia , Genes MDR/efeitos dos fármacos , Gossipol/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Óleo de Sementes de Algodão/administração & dosagem , Óleo de Sementes de Algodão/química , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Gossipol/administração & dosagem , Gossipol/química , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Tamoxifeno/administração & dosagem , Tamoxifeno/farmacologia , Células Tumorais Cultivadas
20.
J Steroid Biochem Mol Biol ; 100(1-3): 24-33, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16854582

RESUMO

In normal prostate, keratinocyte growth factor (KGF), also known as fibroblast growth factor-7 (FGF-7) serves as a paracrine growth factor synthesized in stromal cells that acts on epithelial cells through its receptor, KGFR. KGF and KGFR were found in human cancer epithelial cells as well as stromal cells. Since KGF expressed in epithelial cells of benign prostatic hyperplasia (BPH) and in prostate cancer, it has been suggested that KGF might act as an autocrine factor in BPH and prostate cancer. To investigate the roles of KGF in cancerous stroma, primary cultured human prostate cancer stromal cells (PCSCs) were isolated and evaluated. These PCSCs possessed estrogen receptors and KGFR, but not androgen receptor as determined by RT-PCR and Western blot, respectively. KGF exhibited mitogenic and anti-apoptotic effects that correlated with induction of cyclin-D1, Bcl-2, Bcl-xL and phospho-Akt expression in PCSCs, where treatment with KGF antiserum abolished cell proliferation and anti-apoptotic protein expression. PCSCs exposed to KGF for various time periods resulted in phosphorylation of Akt and subsequent up-regulation of Bcl-2. KGF modulated dynamic protein expression indicated that KGF triggered cell cycle machinery and then activated anti-apoptotic actions in PCSCs. Cell proliferation analysis indicated that tamoxifen or ICI 182,780 reduced cell viability in a dose-dependent manner; however, KGF prevented this inhibition, which further demonstrated KGF triggered anti-apoptotic machinery through activating Bcl-2 and phospho-Akt expression. In summary, KGF has an autocrine effect and serves as a survival factor in primary cultured human prostate cancer stromal cells.


Assuntos
Fator 7 de Crescimento de Fibroblastos/farmacologia , Neoplasias da Próstata/patologia , Células Estromais/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Imuno-Histoquímica , Masculino , Receptores de Estrogênio/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Células Estromais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA