Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Osteoporos Int ; 27(4): 1469-1476, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26556733

RESUMO

UNLABELLED: Treatment with zoledronic acid in osteoporotic patients with spinal fusion shortens the duration of time to fusion, improves the fusion rate, prevents the subsequent adjacent vertebral compression fractures, improves the clinical outcomes, and prevents immobilization-induced bone loss in the hip. INTRODUCTION: The objective of the study was to explore the effects of zoledronic acid on the healing process in osteoporotic patients following spinal fusion in a randomized, placebo-controlled, and triple-blinded study. METHODS: Seventy-nine osteoporotic patients with single-level degenerative spondylolisthesis were randomly assigned to receive either zoledronic acid infusion (zoledronic acid group) or saline infusion (controls) after spinal fusion. Functional radiography and CT scans were used to evaluate fusion status. Bone formation was graded into three categories: Grade A (bridging bone bonding with adjacent vertebral bodies), Grade B (bridging bone bonding with either superior or inferior vertebral body), or Grade C (incomplete bony bridging). A solid fusion was defined as less than 5° of angular motion with Grade A or B bone formation. Adjacent vertebral compression fractures (VCF) were assessed on MRI at 12 months after surgery. Serum level of carboxy terminal cross-linked telopeptide of type I collagen (ß-CTX) and amino-terminal propeptide of type I procollagen (PINP) was measured. Bone mineral density (BMD) was measured by DXA. Oswestry Disability Index (ODI) was used to assess the clinical outcomes. RESULTS: Grade A or B bridging bone was more frequently observed in zoledronic acid group at 3, 6, and 9 months post-operation compared to the control group (p < 0.05). At 12 -months post-operation, bridging bone and solid fusion were not significantly different between groups. No patients in zoledronic acid group showed aVCF, whereas six patients (17 %) in the control group did (p < 0.05). Both ß-CTX and PINP were suppressed in zoledronic acid group. BMD at the femoral neck decreased rapidly and did not return to the preoperative level in the controls at 3 (-1.4 %), 6 (-2.5 %), and 12 (-0.8 %) months after surgery. Zoledronic acid prevented this immobilization-induced bone loss and increased BMD. ODI showed the improved clinical outcomes compared with controls at 9 and 12 months post-surgery. CONCLUSION: Treatment with zoledronic acid in osteoporotic patients with spinal fusion shortens the time to fusion, improves the fusion rate, prevents subsequent aVCFs, and improves clinical outcomes.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Difosfonatos/farmacologia , Imidazóis/farmacologia , Vértebras Lombares/cirurgia , Osteoporose/tratamento farmacológico , Fusão Vertebral/métodos , Idoso , Biomarcadores/sangue , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/fisiologia , Conservadores da Densidade Óssea/uso terapêutico , Difosfonatos/uso terapêutico , Avaliação da Deficiência , Método Duplo-Cego , Feminino , Colo do Fêmur/fisiopatologia , Fraturas por Compressão/prevenção & controle , Humanos , Imidazóis/uso terapêutico , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/fisiopatologia , Masculino , Pessoa de Meia-Idade , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Osteoporose/fisiopatologia , Cuidados Pós-Operatórios/métodos , Estudos Prospectivos , Espondilolistese/cirurgia , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Ácido Zoledrônico
2.
Minerva Urol Nefrol ; 64(1): 19-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22402315

RESUMO

The cancer stem cell (CSC) model provides insights into pathophysiology of cancers and their therapeutic response. The CSC model has been both controversial, yet provides a foundation to explore cancer biology. In this review, we provide an overview of CSC concepts, biology and potential therapeutic avenues. We then focus on prostate CSC including 1) their purported origin as either basal-derived or luminal-derived cells; 2) markers used for prostate CSC identification; 3) alterations of signaling pathways in prostate CSCs; 4) involvement of prostate CSCs in metastasis of PCa; and 5) microRNA-mediated regulation of prostate CSCs. Although definitive evidence for the identification and characterization of prostate CSCs still remains unclear, future directions pursuing therapeutic targets of CSCs may provide novel insights for the treatment of PCa.


Assuntos
Biomarcadores Tumorais/genética , MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Humanos , Masculino , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/fisiopatologia , Transdução de Sinais/genética
3.
J Clin Invest ; 106(7): 887-95, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11018077

RESUMO

To investigate the role of IL-6 in alcohol-mediated osteoporosis, we measured a variety of bone remodeling parameters in wild-type (il6(+/+)) or IL-6 gene knockout (il6(-/-)) mice that were fed either control or ethanol liquid diets for 4 months. In the il6(+/+) mice, ethanol ingestion decreased bone mineral density, as determined by dual-energy densitometry; decreased cancellous bone volume and trabecular width and increased trabecular spacing and osteoclast surface, as determined by histomorphometry of the femur; increased urinary deoxypyridinolines, as determined by ELISA; and increased CFU-GM formation and osteoclastogenesis as determined ex vivo in bone marrow cell cultures. In contrast, ethanol ingestion did not alter any of these parameters in the il6(-/-) mice. Ethanol increased receptor activator of NF-kappaB ligand (RANKL) mRNA expression in the bone marrow of il6(+/+) but not il6(-/-) mice. Additionally, ethanol decreased several osteoblastic parameters including osteoblast perimeter and osteoblast culture calcium retention in both il6(+/+) and il6(-/-) mice. These findings demonstrate that ethanol induces bone loss through IL-6. Furthermore, they suggest that IL-6 achieves this effect by inducing RANKL and promoting CFU-GM formation and osteoclastogenesis.


Assuntos
Alcoolismo/complicações , Reabsorção Óssea/etiologia , Interleucina-6/metabolismo , Osteoclastos/citologia , Absorciometria de Fóton , Animais , Reabsorção Óssea/diagnóstico por imagem , Diferenciação Celular , Masculino , Camundongos , Camundongos Mutantes
4.
J Clin Invest ; 107(10): 1235-44, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11375413

RESUMO

Prostate cancer (CaP) forms osteoblastic skeletal metastases with an underlying osteoclastic component. However, the importance of osteoclastogenesis in the development of CaP skeletal lesions is unknown. In the present study, we demonstrate that CaP cells directly induce osteoclastogenesis from osteoclast precursors in the absence of underlying stroma in vitro. CaP cells produced a soluble form of receptor activator of NF-kappaB ligand (RANKL), which accounted for the CaP-mediated osteoclastogenesis. To evaluate for the importance of osteoclastogenesis on CaP tumor development in vivo, CaP cells were injected both intratibially and subcutaneously in the same mice, followed by administration of the decoy receptor for RANKL, osteoprotegerin (OPG). OPG completely prevented the establishment of mixed osteolytic/osteoblastic tibial tumors, as were observed in vehicle-treated animals, but it had no effect on subcutaneous tumor growth. Consistent with the role of osteoclasts in tumor development, osteoclast numbers were elevated at the bone/tumor interface in the vehicle-treated mice compared with the normal values in the OPG-treated mice. Furthermore, OPG had no effect on CaP cell viability, proliferation, or basal apoptotic rate in vitro. These results emphasize the important role that osteoclast activity plays in the establishment of CaP skeletal metastases, including those with an osteoblastic component.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Reabsorção Óssea/tratamento farmacológico , Glicoproteínas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Animais , Neoplasias Ósseas/patologia , Reabsorção Óssea/patologia , Proteínas de Transporte/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Metástase Neoplásica/tratamento farmacológico , Osteoprotegerina , Neoplasias da Próstata/secundário , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral , Tíbia
5.
Cytokine Growth Factor Rev ; 12(1): 33-40, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11312117

RESUMO

Prostate cancer, while initially dependent on androgens for proliferation, progresses to an androgen-independent state. Evidence has been accumulating that interleukin-6 (IL-6) may contribute to prostate cancer progression. Serum levels of IL-6 correlate with prostate tumor burden and patient morbidity. The prostate tissue itself appears to be a source of IL-6 and its receptor. Furthermore, experimental data suggest that IL-6 is an autocrine and paracrine growth factor for androgen-independent prostate cancer cell lines. For example, inhibition of IL-6, with anti-IL-6 antibody, sensitizes androgen-independent prostate cancer cells to chemotherapeutic agents in vitro. Finally, IL-6 activates a variety of signal transduction cascades, some which stimulate androgen receptor activity, in prostate cancer cells. These data suggest that targeting IL-6 may have multiple benefits in prostate cancer patients.


Assuntos
Interleucina-6/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Humanos , Interleucina-6/análise , Interleucina-6/farmacologia , Masculino , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/fisiopatologia , Receptores Androgênicos/metabolismo , Receptores de Interleucina-6/análise , Receptores de Interleucina-6/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
6.
Math Biosci ; 294: 1-14, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28919575

RESUMO

Despite advanced efforts in early diagnosis, aggressive surgical treatment, and use of targeted chemotherapies, the prognosis for many cancers is still dismal. This emphasizes the necessity to develop new strategies for understanding tumor growth and metastasis. Here we use a systems approach that combines mathematical modeling and numerical simulation to develop a predictive computational model for prostate cancer and its subversion of the bone microenvironment. This model simulates metastatic prostate cancer evolution, progressing from normal bone and hormone levels to quantifiable diseased states. The simulations clearly demonstrate phenomena similar to those found clinically in prostate cancer patients. In addition, the major prediction of this model is the existence of low and high osteogenic states that are markedly different from one another. The existence and potential realization of these steady states appear to be mediated by the Wnt signaling pathway and by the effects of PSA on TGF-ß, which encourages the bone microenvironment to evolve. The model is used to explore several potential therapeutic strategies, with some potential drug targets showing more promise than others: in particular, completely blocking Wnt and greatly increasing DKK-1 had significant positive effects, while blocking RANKL did not improve the outcome.


Assuntos
Neoplasias Ósseas/secundário , Simulação por Computador , Modelos Teóricos , Neoplasias da Próstata/patologia , Neoplasias Ósseas/tratamento farmacológico , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico
7.
Oncogene ; 36(37): 5243-5251, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28504717

RESUMO

Urothelial carcinoma is the most common type of bladder cancer and can be categorized as either non-muscle-invasive (Ta-T1) or muscle-invasive (⩾T2). The majority of bladder cancers are non-muscle-invasive at presentation; however, the recurrence rate for these tumors is high and a subset can progress to T2. In this study, we aimed to identify genes differentially expressed between T1 vs T2 bladder cancer to identify key regulators of bladder cancer progression and/or invasion. We performed RNA-Seq on T1 and T2 bladder cancer tissues and used publicly available bladder cancer profiling studies to prioritize differentially expressed genes for validation and functional assessment. This integrative approach nominated an extracellular matrix glycoprotein, fibulin-3 (FBLN3, also known as EFEMP1), as being highly expressed in T2 vs T1 bladder cancer and aggressive vs indolent disease. We confirmed the overexpression of fibulin-3 in ⩾T2 vs non-muscle-invasive bladder cancer (NMIBC) by quantitative reverse transcriptase-PCR. Consistent with these findings, fibulin-3 expression level correlated with the invasive ability of several bladder cancer cell lines and modulation of fibulin-3 expression directly affected invasion. Fibulin-3 knockdown in bladder cancer cells decreased the incidence of MIBCs in a murine orthotopic bladder cancer model and decreased the expression of insulin-like growth factor-binding protein-5 (IGFBP5). Restoring IGFBP5 in these cells rescued their invasive and migratory potential. These results indicate that fibulin-3 serves as a pro-invasive factor in bladder cancer, which may be mediated through modulation of IGFBP5 expression. This also suggests fibulin-3 and IGFBP5 may have potential as biomarkers of aggressive bladder cancer or therapeutic targets.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Animais , Linhagem Celular Tumoral , Progressão da Doença , Proteínas da Matriz Extracelular/genética , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Neoplasias da Bexiga Urinária/genética
8.
Oncogene ; 35(3): 366-76, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-25867060

RESUMO

The osteogenic transcription factor, Runx2, is abnormally expressed in prostate cancer (PCa) and associated with metastatic disease. During bone development, Runx2 is activated by signals known to be hyperactive in PCa including the RAS/MAP kinase pathway, which phosphorylates Runx2 on multiple serine residues including S301 and S319 (equivalent to S294 and S312 in human Runx2). This study examines the role of these phosphorylation sites in PCa. Runx2 was preferentially expressed in more invasive PCa cell lines (PC3>C4-2B>LNCaP). Furthermore, analysis using a P-S319-Runx2-specific antibody revealed that the ratio of P-S319-Runx2/total Runx2 as well as P-ERK/total ERK was highest in PC3 followed by C4-2B and LNCaP cells. These results were confirmed by immunofluorescence confocal microscopy, which showed a higher percentage of PC3 cells staining positive for P-S319-Runx2 relative to C4-2B and LNCaP cells. Phosphorylated Runx2 had an exclusively nuclear localization. When expressed in prostate cell lines, wild-type Runx2 increased metastasis-associated gene expression, in vitro migratory and invasive activity as well as in vivo growth of tumor cell xenografts. In contrast, S301A/S319A phosphorylation site mutations greatly attenuated these Runx2 responses. Analysis of tissue microarrays from 129 patients revealed strong nuclear staining with the P-S319-Runx2 antibody in primary PCas and metastases. P-S319-Runx2 staining was positively correlated with Gleason score and occurrence of lymph node metastases while little or no Runx2 phosphorylation was seen in normal prostate, benign prostate hyperplasia or prostatitis indicating that Runx2 S319 phosphorylation is closely associated with PCa induction and progression towards an aggressive phenotype. These studies establish the importance of Runx2 phosphorylation in prostate tumor growth and highlight its value as a potential diagnostic marker and therapeutic target.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Invasividade Neoplásica/genética , Metástase Neoplásica , Neoplasias da Próstata/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Fosforilação/genética , Neoplasias da Próstata/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Prostate Cancer Prostatic Dis ; 8(3): 253-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15999121

RESUMO

Zoledronic acid (ZA) has been shown to inhibit prostate tumor growth in vitro and have beneficial effects in patients with advanced prostate cancer (CaP). The aim of this study was to determine whether ZA exhibits direct anti-tumor effects on CaP cells in vivo. To distinguish the effects of inhibition of osteolysis and direct anti-tumor activity of ZA in vivo, we compared the results of treatment with ZA and osteoprotegerin (Fc-OPG), which inhibits osteolysis, but without significant direct anti-tumor effects. In vitro Fc-OPG had no significant effects on C4-2 proliferation, whereas ZA decreased proliferation. However, both agents decreased tumor growth in bone. Moreover, both increased bone volume and prevented the overall decreases in BMD associated with growth of C4-2 cells in bone. Our study provides novel and significant observations that the in vivo effects of ZA are consistent with indirect effects mediated by osteoclasts.


Assuntos
Anticorpos/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Difosfonatos/farmacologia , Glicoproteínas/química , Glicoproteínas/imunologia , Imidazóis/farmacologia , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/imunologia , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Apoptose , Densidade Óssea , Conservadores da Densidade Óssea/farmacologia , Proliferação de Células , Densitometria , Difosfonatos/química , Glicoproteínas/farmacologia , Humanos , Imidazóis/química , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Metástase Neoplásica , Osteoclastos/metabolismo , Osteólise , Osteoprotegerina , Tíbia/patologia , Fatores de Tempo , Ácido Zoledrônico
10.
Clin Cancer Res ; 7(6): 1773-81, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11410519

RESUMO

Interleukin-6 (IL-6) induces prostate cancer (CaP) cell proliferation in vitro. Several lines of evidence suggest that IL-6 may promote CaP progression through induction of an androgen response. In this work, we explored whether IL-6 induces androgen responsiveness through modulation of androgen receptor (AR) expression. We found that in the absence of androgen, IL-6 increased prostate-specific antigen (PSA) mRNA levels and activated several androgen-responsive promoters, but not the non-androgen responsive promoters in LNCaP cells. Bicalutamide, an antiandrogen, abolished the IL-6 effect and IL-6 could not activate the PSA and murine mammary tumor virus reporters in AR-negative DU-145 and PC3 cells. These data indicate the IL-6 induces an androgen response in CaP cells through the AR. Pretreatment of LNCaP cells with SB202190, PD98059, or tyrphostin AG879 [p38 mitogen-activated protein kinase (MAPK), MAP/extracellular signal-regulated protein kinase kinase 1/2, and ErbB2 MAPK inhibitors, respectively) but not wortmannin (PI3-kinase inhibitor) blocked IL-6-mediated induction of the PSA promoter, which demonstrates that IL-6 activity is dependent on a MAPK pathway. Finally, IL-6 activated the AR gene promoter, resulting in increased AR mRNA and protein levels in LNCaP cells. These results demonstrate that IL-6 induces AR expression and are the first report of cytokine-mediated induction of the AR promoter. Taken together, our results suggest that IL-6 induces AR activity through both increasing AR gene expression and activating the AR in the absence of androgen in CaP cells. These results provide a mechanism through which IL-6 may contribute to the development of androgen-independent CaP.


Assuntos
Interleucina-6/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Regulação para Cima , Anilidas/farmacologia , Western Blotting , Núcleo Celular/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Gammaretrovirus/metabolismo , Humanos , Imidazóis/farmacologia , Interleucina-6/biossíntese , Interleucina-6/farmacologia , Masculino , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Antígeno Prostático Específico/biossíntese , Antígeno Prostático Específico/genética , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Tosil , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Tirfostinas/farmacologia
11.
Clin Cancer Res ; 6(5): 1969-77, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10815922

RESUMO

Prostate cancer patients experiencing a relapse in disease often express high serum tumor necrosis factor-alpha (TNF-alpha) levels. Many androgen-insensitive prostate cancer cells are TNF-alpha insensitive because of the expression of antiapoptotic genes as part of the nuclear factor-kappaB (NF-kappaB) family of transcription factors. NF-kappaB stimulates gene transcription when expressed in the nucleus; however, in resting cells, this nuclear import is prevented by association with the cytoplasmic inhibitor IkappaBalpha. This cytoplasmic retention of NF-kappaB is uncoupled by many extracellular signals including low levels of TNF-alpha. During normal cell activation, nuclear translocation of NF-kappaB is preceded by phosphorylation and degradation of IkappaBalpha. When phosphorylation is blocked, IkappaBalpha remains intact, thereby blocking NF-kappaB translocation to the nucleus and subsequent activation of antiapoptotic genes that cause TNF-alpha insensitivity. We tested whether a "super-repressor" of NF-kappaB activity could be transfected into prostate cancer cells and make them TNF-alpha sensitive. PC-3 and LNCaP cells were stimulated with TNF-alpha (10 ng/ml) for 24 h in the presence or absence of the IkappaBalpha "super-repressor" (p6R-IkappaB(S32A + S36A)). NF-kappaB activity was measured by electrophoretic mobility shift assay and the steady state levels of the cytoplasmic IkappaBalpha protein were measured by Western blot. Secretory IL-6 and IL-6 mRNA were measured by ELISA. p6R-IkappaB(S32A + S36A) blocked the stimulation of NF-kappaB activity by TNF-alpha in prostate cancer cells. It also subsequently decreased IL-6 production by TNF-alpha. We conclude that these data demonstrate that inhibition of NF-kappaB selectively sensitizes previously insensitive prostate cancer cells to TNF-alpha.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , NF-kappa B/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Fator de Necrose Tumoral alfa/farmacologia , Sequência de Aminoácidos , Transporte Biológico , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/genética , Masculino , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
12.
Clin Cancer Res ; 1(12): 1595-601, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9815961

RESUMO

Two randomized, double-blind clinical trials in dogs with spontaneous appendicular osteosarcoma treated with combination chemoimmunotherapy are reported. In both trials, dogs without overt metastasis underwent complete amputation of the affected limb. In trial 1, 40 dogs were treated with cisplatin chemotherapy [(CDDP), 70 mg/m2 i.v. every 28 days x 4]. Following CDDP, dogs without evidence of overt metastasis (n = 25) were randomized to receive liposome-encapsulated muramyl tripeptide phosphatidylethanolamine ](L-MTP-PE), 2 mg/m2 i.v.) or placebo liposomes (lipid equivalent) twice weekly for 8 weeks. Of 14 dogs in the placebo group, 13 (93%) died of metastasis; the median survival time was 9.8 months. Of 11 dogs in the L-MTP-PE group, 8 (73%) developed metastasis; the median survival time was 14.4 months, which was significantly longer than that of the placebo group (P < 0.01). In trial 2, 64 dogs received CDDP (70 mg/m2 i.v. every 21 days x 4) and were randomized to concurrently receive L-MTP-PE (2 mg/m2 i.v.) twice or once weekly, or placebo liposomes once weekly for 8 weeks. Median survival times were 10.3, 10.5, and 7.6 months, respectively. There were no significant differences among the three treatment groups in trial 2. Survival times for dogs receiving L-MTP-PE in trial 1 were significantly longer than those for dogs in trial 2 that received four doses of CDDP concurrently with twice weekly L-MTP-PE (P < 0. 04). The results of the first trial confirm our previous observation that L-MTP-PE has antimetastatic activity in dogs with osteosarcoma when given following amputation. The results of the second trial demonstrate that there is no survival advantage of administering L-MTP-PE concurrently with CDDP.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/veterinária , Doenças do Cão/tratamento farmacológico , Osteossarcoma/veterinária , Acetilmuramil-Alanil-Isoglutamina/administração & dosagem , Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Quimioterapia Adjuvante/veterinária , Cisplatino/administração & dosagem , Doenças do Cão/patologia , Cães , Método Duplo-Cego , Portadores de Fármacos , Feminino , Lipossomos , Masculino , Osteossarcoma/tratamento farmacológico , Osteossarcoma/secundário
13.
Hum Gene Ther ; 9(13): 1851-61, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9741424

RESUMO

Cytokine gene-engineered tumor vaccines are currently an area of intense investigation in both basic research and clinical medicine. Our efforts to utilize tumor vaccines in an immunotherapeutic manner involve canines with spontaneous tumors. We hypothesized that canine tumor cells, transfected with human granulocyte-macrophage colony-stimulating factor (hGM-CSF) cDNA in a plasmid vector, would prove nontoxic following intradermal administration, generate biologically relevant levels of protein, effect local histological changes at the sites of vaccination, and create a systemic antitumor response. Sixteen tumor-bearing dogs were admitted to a study of ex vivo gene therapy. Tumor tissue was surgically removed, enzymatically and mechanically dissociated, irradiated, transfected, and intradermally injected back into the patients. The dogs were vaccinated with primary autologous tumor cells transfected with hGM-CSF or a reporter control gene. hGM-CSF protein was detected (0.07 to 14.15 ng/vaccination site) at 24 hr postinjection and dramatic histological changes were observed, characterized by neutrophil and macrophage infiltration at the sites of injection of hGM-CSF-transfected tumor cells. This was in stark contrast to the lesser neutrophilic and eosinophilic infiltrates found at control vaccination sites. Objective evidence of an antitumor response was observed in three animals. These data, in a large animal translational model of spontaneous tumors, demonstrate in vivo biological activity of hGM-CSF-transfected autologous tumor cell vaccines.


Assuntos
Vacinas Anticâncer/uso terapêutico , Doenças do Cão/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias/veterinária , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Transplante de Células/veterinária , DNA Complementar , Doenças do Cão/patologia , Cães , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Masculino , Transplante de Neoplasias/veterinária , Neoplasias/patologia , Neoplasias/terapia , Transfecção/métodos , Células Tumorais Cultivadas , Vacinação/veterinária
14.
Antioxid Redox Signal ; 3(3): 493-504, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11491660

RESUMO

Aging is associated not only with oxidant stress, but also with increased interleukin-6 (IL-6) levels. To determine if oxidative stress could contribute to the age-associated increase IL-6 expression, we exposed LNCaP prostate carcinoma cells and HeLa cervical carcinoma cells to H2O2 as an oxidant challenge. We found that H2O2 induced IL-6 expression through activation of the IL-6 promoter. Furthermore, H2O2-induced activation of the promoter was mediated through nuclear factor-kappaB (NFkappaB) secondary to H2O2-induced phosphorylation and degradation of IkappaBalpha. NFkappaB-inducing kinase (NIK) is upstream of the IkappaB kinase complex that induces IkappaBalpha degradation. Accordingly, we explored if H2O2 induces IL-6 expression through NIK. In addition to H2O2 inducing NIK autophosphorylation, transfection of LNCaP cells with a dominant negative NIK diminished H2O2-mediated NFkappaB and IL-6 promoter activity. Taken together, these results demonstrate that H2O2 induces the IL-6 promoter by activating NFkappaB through NIK. These data provide a candidate mechanism through which oxidant challenge induces IL-6 gene expression with age.


Assuntos
Peróxido de Hidrogênio/farmacologia , Proteínas I-kappa B , Interleucina-6/genética , NF-kappa B/metabolismo , Oxidantes/farmacologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/fisiologia , Transporte Ativo do Núcleo Celular , Envelhecimento , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/fisiologia , Relação Dose-Resposta a Droga , Feminino , Células HeLa , Humanos , Interleucina-6/biossíntese , Cinética , Masculino , Inibidor de NF-kappaB alfa , Estresse Oxidativo , RNA Mensageiro/biossíntese , Ativação Transcricional , Células Tumorais Cultivadas , Quinase Induzida por NF-kappaB
15.
Cancer Gene Ther ; 3(3): 186-91, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8725883

RESUMO

Cytokines can stimulate immune effector cells present within the oral mucosa and epidermis to respond to vaccination or to combat cancer. However, intravenous cytokine delivery is often inefficient and frequently accompanied by systemic toxicity. The goal of this study was to evaluate dogs as a large animal model for gene therapy of cancer because they develop spontaneous oral and epidermal tumors. In this report, we demonstrate that particle-mediated gene transfer of beta-galactosidase, luciferase, interleukin-2, interleukin-6, and granulocyte-macrophage colony stimulating factor (GM-CSF) complementary DNA (cDNA) into the oral mucosa and epidermis of healthy dogs resulted in effective, localized, transgenic protein expression. Additionally, the epidermal sites transfected with GM-CSF developed a profound inflammatory reaction characterized by neutrophilic infiltration. Clinical pathology analyses were unremarkable. These results demonstrate that in vivo particle-mediated gene transfer of canine oral mucosa and epidermis with cytokine cDNA can result in production of biologically active transgenic cytokines with minimal toxicity. These findings have applications to cancer immunotherapy using a gene gun approach.


Assuntos
Citocinas/biossíntese , Citocinas/genética , Epiderme/fisiologia , Mucosa Bucal/fisiologia , Transfecção/métodos , Animais , Cães , Expressão Gênica , Técnicas de Transferência de Genes , Genes Reporter , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Interleucina-2/biossíntese , Interleucina-6/biossíntese , Luciferases/biossíntese , Camundongos , Fatores de Tempo , beta-Galactosidase/biossíntese
16.
Clin Exp Metastasis ; 21(5): 381-7, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15672862

RESUMO

Osteoprotegerin (OPG) plays a central role in controlling bone resorption. Exogenous administration of OPG has been shown to be effective in preventing osteolysis and limiting the growth of osteolytic metastasis. The objective of this study was to investigate the effects of OPG on osteoblastic prostate cancer (CaP) metastases in an animal model. LuCaP 23.1 cells were injected intra-tibially and Fc-OPG (6.0 mg/kg) was administered subcutaneously three times a week starting either 24 hours prior to cell injection (prevention regimen) or at 4 weeks post-injection (treatment regimen). Changes in bone mineral density at the tumor site were determined by dual x-ray absorptiometry. Tumor growth was monitored by evaluating serum prostate specific antigen (PSA). Fc-OPG did not inhibit establishment of osteoblastic bone lesions of LuCaP 23.1, but it decreased growth of the tumor cells, as determined by decreases in serum PSA levels of 73.0 +/- 44.3% (P < 0.001) and 78.3 +/- 25.3% (P < 0.001) under the treatment and prevention regimens, respectively, compared to the untreated tumor-bearing animals. Administration of Fc-OPG decreased the proliferative index by 35.0% (P = 0.1838) in the treatment group, and 75.2% (P = 0.0358) in the prevention group. The results of this study suggest a potential role for OPG in the treatment of established osteoblastic CaP bone metastases.


Assuntos
Neoplasias Ósseas/terapia , Glicoproteínas/administração & dosagem , Osteoblastos/metabolismo , Neoplasias da Próstata/terapia , Receptores Citoplasmáticos e Nucleares/administração & dosagem , Tíbia/patologia , Animais , Densidade Óssea , Neoplasias Ósseas/secundário , Injeções Subcutâneas , Metástase Linfática/patologia , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Osteoprotegerina , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/patologia , Receptores do Fator de Necrose Tumoral/administração & dosagem , Tíbia/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Bone ; 28(2): 167-73, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11182374

RESUMO

Alcoholics frequently suffer from moderate to severe bone loss that results in bone fractures. Both decreased bone production and increased bone resorption have been postulated to contribute to ethanol (ETOH)-mediated bone loss. Bone resorption is induced by several proinflammatory cytokines such as interleukin-1 and -6. The expression of these cytokines is induced by the transcription factor NFkappaB, which, in turn, is activated by several kinases. It follows that protein kinase and NFkappaB activation may contribute to ETOH-induced bone loss. Accordingly, we sought to determine if ETOH activates protein tyrosine kinases (PTK) and NFkappaB DNA binding in a human osteoblast-like cell line (HOBIT). Ethanol at 50 and 100 mmol/L (reflective of blood ethanol levels reached in chronic alcoholics) for 24 h did not alter HOBIT cell viability. In contrast, 200 mmol/L ethanol decreased cell viability by 40%. Treatment of HOBIT cells with 100 mmol/L ETOH induced nuclear NFkappaB:DNA complex formation and NFkappaB activity. Incubation of HOBIT cells with ETOH at 50 and 100 mmol/L for 30 min induced a 2.5- and 4.2-fold increase in PTK activity, respectively. Preincubation of HOBIT cells with damnacanthal (DAM), which inhibits p56lck, blocked ETOH-mediated PTK activity; whereas, preincubation with herbimycin A, which inhibits pp60src, did not. DAM inhibited both ethanol-induced NFkappaB activation in HOBIT cells and interleukin-6 expression in primary human osteoblasts. Finally, preincubation with the protein kinase C inhibitor, bisindolylmaleimide I HCl (BIS), diminished ETOH-mediated PTK activity; whereas, preincubation with the protein kinase A inhibitor, H89, did not. These data demonstrate that ETOH induces NFkappaB nuclear translocation through p56lck in HOBIT cells. BIS' inhibition of PTK activation suggests that ETOH activates PTK through a protein kinase C-dependent pathway. These data suggest that ETOH may contribute to bone loss through activation of signal transduction that results in production of an osteoclastogenic cytokine (i.e., interleukin-6) in osteoblasts.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Alcoolismo/metabolismo , Linhagem Celular , Núcleo Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/genética , Osteoblastos/citologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Bone ; 26(1): 55-62, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10617157

RESUMO

To characterize the role of interleukin-6 (IL-6) in estrogen (E2)-depletion bone loss, we utilized a nonhuman primate model of human skeletal physiology. Adult female rhesus monkeys were sham-operated (S; n = 5), ovariectomized (ovx; n = 10), or ovx followed by E2 replacement (ovx + E2; n = 10) and evaluated for the indicated parameters at 0, 3, 6, and 9 months post-ovx. Lumbar spine bone mineral density (BMD) decreased by 3 months and continued to decline through 9 months in the ovx, but not in the ovx + E2 or S groups. Middle and distal radius BMD was decreased at 9 months in the ovx, but not in the ovx + E2 or S groups. The S group had marked fluctuations in bone remodeling parameters, and cytokine levels in S animals were consistent with menstrual cycling, and therefore only those values in the ovx and ovx + E2 groups are reported. Serum osteocalcin and skeletal-specific alkaline phosphatase were elevated in the ovx group compared with the ovx + E2 group. There was no difference in serum or bone marrow plasma IL-6 levels between the ovx and ovx + E2 groups. Similarly, there was no difference in basal or phorbol ester-stimulated IL-6 levels of peripheral blood mononuclear cell or bone marrow cell culture supernatants between groups. There was no difference in serum or bone marrow soluble IL-6 receptor between groups. However, the bone marrow plasma soluble IL-6 receptor levels were transiently increased from baseline at 3 months in the ovx but not in the ovx + E2 group. In summary, there was no bone loss in the ovx + E2 group, although the serum and bone marrow IL-6 levels were similar to those of the ovx group. These data suggest that modulation of IL-6 is not the key mechanism through which estrogen deprivation mediates bone loss in rhesus monkeys.


Assuntos
Doenças Ósseas Metabólicas/fisiopatologia , Interleucina-6/fisiologia , Animais , Biomarcadores , Densidade Óssea , Doenças Ósseas Metabólicas/metabolismo , Remodelação Óssea , Dinoprostona/metabolismo , Feminino , Humanos , Macaca mulatta , Ovariectomia
19.
Bone ; 26(1): 79-85, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10617160

RESUMO

Rodent models suggest that estradiol deficiency promotes bone loss through increasing interleukin-6 (IL-6) activity. However, it is controversial as to whether these findings are applicable to humans. To evaluate estradiol-mediated modulation of IL-6 activity in relation to bone metabolism in humans, we measured serum IL-6, soluble interleukin-6 receptor (sIL-6R), estradiol (E2), progesterone, luteinizing hormone, follicle-stimulating hormone, intact parathyroid hormone (PTH), serum and urine Ca, and bone biochemical markers (serum bone-specific alkaline phosphatase, osteocalcin, and serum and urine deoxypyridinoline [Dpd]) across one menstrual cycle for 211 women. Neither IL-6 nor sIL-6R levels differed between the follicular phase (FP) and the luteal phases (LP). However, IL-6 was negatively correlated with E2 during the FP (p =0.003). Furthermore, IL-6 correlated positively with serum Ca over the entire cycle (p = 0.0091. Serum Ca correlated positively with serum (p = 0.040) and urine (p = 0.006) Dpd. PTH was significantly higher during the FP than in the LP (p = 0.004). PTH was negatively related to E2 (p = 0.002), serum Ca (p < 0.001), and urine Ca (p = 0.036), whereas it was positively correlated with IL-6 (p = 0.027). These data demonstrate that IL-6 and PTH fluctuate with E2, and serum II-6 is associated with PTH levels during the menstrual cycle. However, the role of 11-6 in bone remodeling during the normal menstrual cycle remains to be determined.


Assuntos
Estradiol/sangue , Interleucina-6/sangue , Ciclo Menstrual , Hormônio Paratireóideo/sangue , Receptores de Interleucina-6/sangue , Adulto , Biomarcadores , Remodelação Óssea , Cálcio/metabolismo , Feminino , Humanos , Masculino , Solubilidade
20.
Front Biosci ; 1: d59-71, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9159212

RESUMO

Androgens mediate a number of diverse responses through the androgen receptor, a 110 kD ligand-activated nuclear receptor. Androgen receptor expression, which is found in a variety of tissues, changes throughout development, aging, and malignant transformation. The androgen receptor can be activated by two ligands, testosterone and dihydrotestosterone, which bind to the androgen receptor with different affinities. This difference in binding affinity results in different levels of activation of the androgen receptor by the two ligands. The androgen receptor acts as a transcriptional modifier of a variety of genes by binding to an androgen response element. The ability to confer androgen specific actions by the androgen response element may depend on other cell-specific transcription factors and cis-acting DNA elements in close proximity to it. Testosterone and dihydrotestosterone appear to act upon an identical nuclear receptor. However, in certain instances, they mediate different physiologic responses. For example, dihydrotestosterone, but not testosterone, is capable of mediating full sexual development of the male external genitalia. In some cases, the androgen receptor may induce opposite physiologic responses in similar tissue types depending on their location. For example, in male pattern baldness, activated androgen receptors may suppress the growth of distinct hair follicle populations through initiating stromal-epithelial actions, whereas other hair follicles continue to proliferate. In other cases, altered androgen receptor activity due to its mutation or altered expression may lead to pathology such as recurrence of prostate cancer due to development of androgen independence allowing tumor cell proliferation under androgen deprivation.


Assuntos
Receptores Androgênicos/fisiologia , Androgênios/fisiologia , Epitélio/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/química , Receptores Androgênicos/genética , Elementos de Resposta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA