Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Neoplasia ; 21(2): 172-184, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30591422

RESUMO

Macrophages play a dual role in regulating tumor progression. They can either reduce tumor growth by secreting antitumorigenic factors or promote tumor progression by secreting a variety of soluble factors. The purpose of this study was to define the monocyte/macrophage population prevalent in skeletal tumors, explore a mechanism employed in supporting prostate cancer (PCa) skeletal metastasis, and examine a novel therapeutic target. Phagocytic CD68+ cells were found to correlate with Gleason score in human PCa samples, and M2-like macrophages (F4/80+CD206+) were identified in PCa bone resident tumors in mice. Induced M2-like macrophages in vitro were more proficient at phagocytosis (efferocytosis) of apoptotic tumor cells than M1-like macrophages. Moreover, soluble factors released from efferocytic versus nonefferocytic macrophages increased PC-3 prostate cancer cell numbers in vitro. Trabectedin exposure reduced M2-like (F4/80+CD206+) macrophages in vivo. Trabectedin administration after PC-3 cell intracardiac inoculation reduced skeletal metastatic tumor growth. Preventative pretreatment with trabectedin 7 days prior to PC-3 cell injection resulted in reduced M2-like macrophages in the marrow and reduced skeletal tumor size. Together, these findings suggest that M2-like monocytes and macrophages promote PCa skeletal metastasis and that trabectedin represents a candidate therapeutic target.


Assuntos
Neoplasias Ósseas/secundário , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Fagocitose/efeitos dos fármacos , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Trabectedina/farmacologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Medula Óssea , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Fenótipo , Neoplasias da Próstata/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Curr Osteoporos Rep ; 16(5): 584-595, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30155844

RESUMO

PURPOSE OF REVIEW: Osteonecrosis of the jaw (ONJ) is a rare and severe necrotic bone disease reflecting a compromise in the body's osseous healing mechanisms and unique to the craniofacial region. Antiresorptive and antiangiogenic medications have been suggested to be associated with the occurrence of ONJ; yet, the pathophysiology of this disease has not been fully elucidated. This article raises the current theories underlying the pathophysiology of ONJ. RECENT FINDINGS: The proposed mechanisms highlight the unique localization of ONJ. The evidence-based mechanisms of ONJ pathogenesis include disturbed bone remodeling, inflammation or infection, altered immunity, soft tissue toxicity, and angiogenesis inhibition. The role of dental infections and the oral microbiome is central to ONJ, and systemic conditions such as rheumatoid arthritis and diabetes mellitus contribute through their impact on immune resiliency. Current experimental studies on mechanisms of ONJ are summarized. The definitive pathophysiology is as yet unclear. Recent studies are beginning to clarify the relative importance of the proposed mechanisms. A better understanding of osteoimmunology and the relationship of angiogenesis to the development of ONJ is needed along with detailed studies of the impact of drug holidays on the clinical condition of ONJ.


Assuntos
Remodelação Óssea/imunologia , Infecções/imunologia , Inflamação/imunologia , Doenças Maxilomandibulares/imunologia , Osteonecrose/imunologia , Inibidores da Angiogênese/efeitos adversos , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/etiologia , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/imunologia , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/metabolismo , Conservadores da Densidade Óssea/efeitos adversos , Remodelação Óssea/fisiologia , Colágeno/metabolismo , Difosfonatos/efeitos adversos , Humanos , Infecções/metabolismo , Inflamação/metabolismo , Doenças Maxilomandibulares/induzido quimicamente , Doenças Maxilomandibulares/metabolismo , Células Matadoras Naturais/imunologia , Mucosa Bucal/imunologia , Mucosa Bucal/lesões , Mucosa Bucal/metabolismo , Neutrófilos/imunologia , Osteonecrose/induzido quimicamente , Osteonecrose/metabolismo , Linfócitos T/imunologia , Cicatrização
3.
Osteoporos Int ; 28(8): 2321-2333, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28429052

RESUMO

Effects of the chemotherapeutic agent etoposide on the skeleton were determined in mice. Numbers of bone marrow cells were reduced and myeloid cells were increased. Bone volume was significantly decreased with signs of inhibition of bone formation. Etoposide after pre-treatment with zoledronic acid still reduced bone but overall bone volume was higher than with etoposide alone. INTRODUCTION: Chemotherapeutics target rapidly dividing tumor cells yet also impact hematopoietic and immune cells in an off target manner. A wide array of therapies have negative side effects on the skeleton rendering patients osteopenic and prone to fracture. This study focused on the pro-apoptotic chemotherapeutic agent etoposide and its short- and long-term treatment effects in the bone marrow and skeleton. METHODS: Six- to 16-week-old mice were treated with etoposide (20-25 mg/kg) or vehicle control in short-term (daily for 5-9 days) or long-term (3×/week for 17 days or 6 weeks) regimens. Bone marrow cell populations and their phagocytic/efferocytic functions were analyzed by flow cytometry. Blood cell populations were assessed by CBC analysis. Bone volume and area compartments and osteoclast numbers were measured by microCT, histomorphometry, and TRAP staining. Biomarkers of bone formation (P1NP) and resorption (TRAcP5b) were assayed from serum. Gene expression in bone marrow was assessed using qPCR. RESULTS: Flow cytometric analysis of the bone marrow revealed short-term etoposide reduced overall cell numbers and B220+ cells, with increased marrow apoptotic (AnnexinV+PI-) cells, mesenchymal stem-like cells, and CD68+, CD45+, and CD11b+ monocyte/myeloid cells (as a percent of the total marrow). After 6 weeks, the CD68+, Gr1+, CD11b+, and CD45+ cell populations were still relatively increased in etoposide-treated bone marrow. Skeletal phenotyping revealed etoposide decreased bone volume, trabecular thickness, and cortical bone volume. Gene expression in the marrow for the leptin receptor and CXCL12 were reduced with short-term etoposide, and an increased ratio of RANKL/OPG mRNA was observed. In whole bone, Runx2 and osteocalcin gene expressions were reduced, and in serum, P1NP was significantly reduced with etoposide. Treatment with the antiresorptive agent zoledronic acid prior to etoposide increased bone volume and improved the etoposide-induced decrease in skeletal parameters. CONCLUSIONS: These data suggest that etoposide induces apoptosis in the bone marrow and significantly reduces parameters of bone formation with rapid reduction in bone volume. Pre-treatment with an antiresorptive agent results in a preservation of bone mass. Preventive approaches to preserving the skeleton should be considered in human clinical studies.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Etoposídeo/efeitos adversos , Osteoporose/induzido quimicamente , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Células Sanguíneas/efeitos dos fármacos , Conservadores da Densidade Óssea/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Difosfonatos/uso terapêutico , Esquema de Medicação , Etoposídeo/administração & dosagem , Etoposídeo/farmacologia , Feminino , Imidazóis/uso terapêutico , Camundongos Endogâmicos C57BL , Osteoporose/diagnóstico por imagem , Osteoporose/fisiopatologia , Osteoporose/prevenção & controle , Tíbia/diagnóstico por imagem , Tíbia/efeitos dos fármacos , Tíbia/fisiopatologia , Microtomografia por Raio-X/métodos , Ácido Zoledrônico
4.
J Dent Res ; 94(10): 1408-16, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26276370

RESUMO

A complex feedback mechanism between parathyroid hormone (PTH), 1,25(OH)2D3 (1,25D), and fibroblast growth factor 23 (FGF-23) maintains mineral homeostasis, in part by regulating calcium and phosphate absorption/reabsorption. Previously, we showed that 1,25D regulates mineral homeostasis by repressing dentin matrix protein 1 (DMP1) via the vitamin D receptor pathway. Similar to 1,25D, PTH may modulate DMP1, but the underlying mechanism remains unknown. Immortalized murine cementoblasts (OCCM.30), similar to osteoblasts and known to express DMP1, were treated with PTH (1-34). Real-time quantitative polymerase chain reaction (PCR) and Western blot revealed that PTH decreased DMP1 gene transcription (85%) and protein expression (30%), respectively. PTH mediated the downregulation of DMP1 via the cAMP/protein kinase A (PKA) pathway. Immunohistochemistry confirmed the decreased localization of DMP1 in vivo in cellular cementum and alveolar bone of mice treated with a single dose (50 µg/kg) of PTH (1-34). RNA-seq was employed to further identify patterns of gene expression shared by PTH and 1,25D in regulating DMP1, as well as other factors involved in mineral homeostasis. PTH and 1,25D mutually upregulated 36 genes and mutually downregulated 27 genes by ≥2-fold expression (P ≤ 0.05). Many identified genes were linked with the regulation of bone/tooth homeostasis, cell growth and differentiation, calcium signaling, and DMP1 transcription. Validation of RNA-seq results via PCR array confirmed a similar gene expression pattern in response to PTH and 1,25D treatment. Collectively, these results suggest that PTH and 1,25D share complementary effects in maintaining mineral homeostasis by mutual regulation of genes/proteins associated with calcium and phosphate metabolism while also exerting distinct roles on factors modulating mineral metabolism. Furthermore, PTH may modulate phosphate homeostasis by downregulating DMP1 expression via the cAMP/PKA pathway. Targeting genes/proteins mutually governed by PTH and 1,25D may be a viable approach for designing new therapies for preserving mineralized tissue health.


Assuntos
Cemento Dentário/efeitos dos fármacos , Proteínas da Matriz Extracelular/antagonistas & inibidores , Hormônio Paratireóideo/farmacologia , Vitamina D/farmacologia , Animais , Western Blotting , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Cemento Dentário/fisiologia , Regulação para Baixo/efeitos dos fármacos , Proteínas da Matriz Extracelular/fisiologia , Fator de Crescimento de Fibroblastos 23 , Imunofluorescência , Expressão Gênica/efeitos dos fármacos , Camundongos , Hormônio Paratireóideo/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Vitamina D/fisiologia
5.
Oncogene ; 33(19): 2464-77, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23752183

RESUMO

Prostate cancer (PCa)bone metastases are unique in that majority of them induce excessive mineralized bone matrix, through undefined mechanisms, as opposed to most other cancers that induce bone resorption. Parathyroid hormone-related protein (PTHrP) is produced by PCa cells and intermittent PTHrP exposure has bone anabolic effects, suggesting that PTHrP could contribute to the excess bone mineralization. Wnts are bone-productive factors produced by PCa cells, and the Wnt inhibitor Dickkopfs-1 (DKK1) has been shown to promote PCa progression. These findings, in conjunction with the observation that PTHrP expression increases and DKK1 expression decreases as PCa progresses, led to the hypothesis that PTHrP could be a negative regulator of DKK1 expression in PCa cells and, hence, allow the osteoblastic activity of Wnts to be realized. To test this, we first demonstrated that PTHrP downregulated DKK1 mRNA and protein expression. We then found through multiple mutated DKK1 promoter assays that PTHrP, through c-Jun activation, downregulated the DKK1 promoter through a transcription factor (TCF) response element site. Furthermore, chromatin immunoprecipitation (ChIP) and re-ChIP assays revealed that PTHrP mediated this effect through inducing c-Jun to bind to a transcriptional activator complex consisting of ß-catenin, which binds the most proximal DKK1 promoter, the TCF response element. Together, these results demonstrate a novel signaling linkage between PTHrP and Wnt signaling pathways that results in downregulation of a Wnt inhibitor allowing for Wnt activity that could contribute the osteoblastic nature of PCa.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Proteína Relacionada ao Hormônio Paratireóideo/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Via de Sinalização Wnt/efeitos da radiação , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , beta Catenina/genética , beta Catenina/metabolismo
6.
J Dent Res ; 90(8): 1007-12, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21555774

RESUMO

Vitamin D regulates calcium and immune function. While vitamin D deficiency has been associated with periodontitis, little information exists regarding its effect on wound healing and periodontal surgery outcomes. This longitudinal clinical trial assessed outcomes of periodontal surgery and teriparatide administration in vitamin-D-sufficient and -insufficient individuals. Forty individuals with severe chronic periodontitis received periodontal surgery, daily calcium and vitamin D supplements, and self-administered teriparatide or placebo for 6 wks to correspond with osseous healing time. Serum 25(OH)D was evaluated at baseline, 6 wks, and 6 mos post-surgery. Clinical and radiographic outcomes were evaluated over 1 yr. Placebo patients with baseline vitamin D deficiency [serum 25(OH)D, 16-19 ng/mL] had significantly less clinical attachment loss (CAL) gain (-0.43 mm vs. 0.92 mm, p < 0.01) and probing depth (PPD) reduction (0.43 mm vs. 1.83 mm, p < 0.01) than vitamin-D-sufficient individuals. Vitamin D levels had no significant impact on CAL and PPD improvements in teriparatide patients at 1 yr, but infrabony defect resolution was greater in teriparatide-treated vitamin-D-sufficient vs. -deficient individuals (2.05 mm vs. 0.87 mm, p = 0.03). Vitamin D deficiency at the time of periodontal surgery negatively affects treatment outcomes for up to 1 yr. Analysis of these data suggests that vitamin D status may be critical for post-surgical healing. (ClinicalTrials.gov number, CT00277706).


Assuntos
Periodontite Crônica/metabolismo , Periodontite Crônica/cirurgia , Deficiência de Vitamina D/metabolismo , Vitamina D/farmacologia , Vitaminas/farmacologia , Cicatrização/efeitos dos fármacos , 24,25-Di-Hidroxivitamina D 3/sangue , Adulto , Idoso , Conservadores da Densidade Óssea/farmacologia , Feminino , Humanos , Modelos Lineares , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Teriparatida/farmacologia , Resultado do Tratamento , Vitamina D/metabolismo , Vitaminas/metabolismo
7.
Oral Dis ; 14(8): 713-26, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19193201

RESUMO

Parathyroid hormone-related protein (PTHrP) is an integral mediator of physiologic and pathologic processes and has demonstrated actions in the periodontium. PTHrP functions via AP-1, and specifically through JunB. This study identified JunB-dependent downstream mediators of PTHrP using OCCM cementoblastic transfectants with JunB over- or reduced expression. Over-expressing cells showed an increase in proliferation, while the opposite was seen in siRNA transfected cells. Microarray analysis of over-expressing cells revealed more than 1000 regulated genes. Three genes were investigated in more detail. The PTH/PTHrP receptor (PTHR1) and ephrin B1 (EfnB1) were down-regulated, and vascular cell adhesion molecule-1 (VCAM-1) was up-regulated with JunB over-expression. JunB siRNA transfectants had increased PTHR1, but reduced ephrin B1 and unaltered VCAM-1 in vitro. To validate these targets, parental OCCM cells and primary osteoblasts were treated with PTHrP, resulting in reduced PTHR1 and ephrin B1, and increased VCAM-1. Cell transfectants were implanted subcutaneously in vivo, and microarray analysis and RT-PCR performed. Over-expression of JunB down-regulated PTHR1 and ephrin B1, and increased VCAM-1. JunB siRNA transfectant implants had increased PTHR1 and ephrin B1, but no altered VCAM-1. These data highlight new gene targets for PTHrP and indicate JunB is a critical mediator of PTHrP actions.


Assuntos
Efrina-B1/genética , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteínas Proto-Oncogênicas c-jun/genética , Molécula 1 de Adesão de Célula Vascular/genética , Animais , Apoptose/genética , Contagem de Células , Linhagem Celular , Proliferação de Células , Cemento Dentário/patologia , Regulação para Baixo/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Nus , Osteoblastos/patologia , Osteoblastos/transplante , Análise Serial de Proteínas , RNA Interferente Pequeno/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tela Subcutânea/patologia , Fator de Transcrição AP-1/genética , Transfecção , Regulação para Cima/genética
8.
Bone ; 38(4): 497-508, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16337237

RESUMO

Stromal derived factor-1 (SDF-1 or CXCL12) controls many aspects of stem cell function including trafficking and proliferation. Previously, it was demonstrated that DNA-damaging agents such as irradiation, cyclophosphamide or 5-fluorouracil increase the expression of SDF-1 by osteoblasts in murine marrow. Here, the production of SDF-1 by osteoblasts in vitro in response to cytokines known to be particularly important in bone physiology was examined using primary human osteoblasts (HOBs), mixed marrow stromal cells (BMSCs), and by, mouse, rat and human osteoblast-like cell lines. From these studies, it was determined that the expression of SDF-1 is an early feature of osteoblastic induction that may be modulated by IL-1beta, PDGF-BB, VEGF, TNF-alpha and PTH. Each of these factors increased SDF-1 synthesis, while TGF-beta1 decreased SDF-1 secretion. Of note, the biodistribution of SDF-1 in culture was equally distributed between the medium and detergent-soluble and -insoluble fractions of the cultures. Immunohistochemistry of developing bones demonstrated that SDF-1 was also a feature of early bone development first beginning in the perichondrium and moving into the marrow cavity of the developing bone analogue. As SDF-1 expression increases in response to PTH in vitro, animals were treated with an anabolic regime of PTH for 21 days. Under these conditions, significant increases in SDF-1 mRNA expression were observed near the growth plate and epiphysis regions of the long bones. Yet, in serum, immunodetectable SDF-1 levels were significantly reduced (24%) in the PTH-treated animals (Vehicle: 408 +/- 25 vs. PTH 308 +/- 20 SDF-1 pg/ml). Together, these data suggest a possible mechanism for localizing stem cells into a developing marrow where increased expression of SDF-1 in the local marrow environment along with decreased SDF-1 in the serum may create a homing gradient.


Assuntos
Quimiocinas CXC/biossíntese , Células-Tronco Hematopoéticas/citologia , Osteoblastos/metabolismo , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12 , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Hibridização In Situ , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Calcif Tissue Int ; 75(1): 60-70, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15037970

RESUMO

The interaction between estrogens and androgens, with their protective effects in bone, and parathyroid hormone (PTH), a calcitropic peptide hormone, is complex but may be better understood with murine models. The purpose of this study was to characterize skeletal phenotypes of mice deficient in estrogen receptor alpha (ERalpha), androgen receptor (AR, mutant tfm), or both, and determine if ERalpha and AR alter osteoblast differentiation and/or PTH response in vitro. Loss of ERalpha resulted in increased long bone length in females, but reduced length in males, suggesting loss of ERalpha reversed sex steroid-dependent skeletal dimorphism. The AR deficient tfm mice (genetically male but phenotypically female) had the longest bones and, similar to males, lengths were reduced with loss of ERalpha. Loss of AR and/or ERalpha resulted in a reduction in femoral bone mineral density (BMD) compared to male wildtype (WT) mice, suggesting tfm mice follow the female sex for BMD. In males or tfm mice, but not females, loss of AR and/or ERalpha caused a reduction in cortical width of the tibia compared to male WT mice. Reduced trabecular bone was found in tibiae of female and tfm mice versus male littermates, suggesting that tfm mice follow the female sex for trabecular bone but loss of ERalpha did not alter trabecular bone levels. Primary calvarial osteoblasts of male WT mice were less responsive to PTH stimulation of cAMP than all other genotypes, suggesting the female chromosomal sex and/ or loss of ERalpha or AR results in increased sensitivity to PTH. In conclusion, tfm mice follow the male pattern of long bone development, but imitate females in bone density and trabecular bone. Loss of ERalpha and/or AR results in increased osteoblast sensitivity to PTH and may explain actions of PTH noted in hypogonadal humans.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Fêmur/metabolismo , Osteoblastos/enzimologia , Receptores Androgênicos/metabolismo , Tíbia/metabolismo , Animais , Densidade Óssea , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Feminino , Fêmur/diagnóstico por imagem , Fêmur/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Modelos Animais , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Hormônio Paratireóideo/farmacologia , Fenótipo , Radiografia , Receptores Androgênicos/deficiência , Receptores Androgênicos/genética , Caracteres Sexuais , Crânio/citologia , Tíbia/diagnóstico por imagem , Tíbia/patologia
10.
Connect Tissue Res ; 44 Suppl 1: 250-63, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12952206

RESUMO

Estrogen has protective effects on the skeleton via its inhibition of bone resorption. Mechanisms for these effects and the selectivity to the estrogen receptor alpha (ER alpha) or ER beta are unclear. The purpose of our study was to determine the impact of the ER alpha on skeletal metabolism using murine models with targeted disruption of the ER alpha and beta. Mice generated by homologous recombination and Cre/loxP technology yielding a deletion of the ER alpha exon 3 were evaluated and also crossed with mice with a disruption of the exon 3 of the ER beta to result in double ER alpha and ER beta knockout mice. Skeletal analysis of long bone length and width, radiographs, dual X-ray absorptiometry, bone histomorphometry, micro computerized tomography, biomechanical analysis, serum biochemistry, and osteoblast differentiation were evaluated. Male ER alpha knockout mice had the most dramatic phenotype consisting of reduced bone mineral density (BMD), and bone mineral content (BMC) of femurs at 10 and 16 weeks and 8-9 months of age. Female ER alpha knockout mice also had reduced density of long bones but to a lesser degree than male mice. The reduction of trabecular and cortical bone in male ER alpha knockout mice was statistically significant. Male double ER alpha and ER beta knockouts had similar reductions in bone density versus the single ER alpha knockout mice suggesting that the ER alpha is more protective than the ER beta in bone. In vitro analysis revealed no differences in osteoblast differentiation or mineralized nodule formation among cells from ER alpha genotypes. These data suggest that estrogens are important in skeletal metabolism in males; the ER alpha plays an important role in estrogen protective effects; osteoblast differentiation is not altered with loss of the ER alpha; and compensatory mechanisms are present in the absence of the ER alpha and/or another receptor for estrogen exists that mediates further effects of estrogen on the skeleton.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Osso e Ossos/metabolismo , Receptores de Estrogênio/metabolismo , Absorciometria de Fóton , Animais , Densidade Óssea , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/patologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Calcificação Fisiológica/fisiologia , Modelos Animais de Doenças , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Fêmur/diagnóstico por imagem , Fêmur/metabolismo , Fêmur/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Osteoblastos/metabolismo , Osteoblastos/patologia , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética , Fatores Sexuais , Tíbia/diagnóstico por imagem , Tíbia/metabolismo , Tíbia/patologia
11.
Domest Anim Endocrinol ; 22(3): 169-77, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11934525

RESUMO

A canine genomic library in Lambda FIX II vector was screened with a 281-base pair canine PTHrP cDNA to the prepro- and coding regions. Two genomic clones were isolated and mapped to the 3'-end of the PTHrP gene by polymerase chain reaction (PCR) amplification of exons in this region. One clone (3.5 kb) was amplified by PCR, partially sequenced, and compared to the human PTHrP gene. Regions were identified with a high degree of homology to exons 6, 7, and 8 of the human PTHrP gene. A polyadenylation site was present 3' to the exon 8-like region. Reverse transcriptase-polymerase chain reaction (RT-PCR) demonstrated that exon 7 of the PTHrP gene was transcribed in two canine carcinomas (SCC 2/88 cells and CAC-8 tumor line) which produce PTHrP. This confirmed that the 3'-region of the canine PTHrP gene is alternately spliced with splicing of exon 6 to exons 7 or 9. Transcription of exon 8 was not demonstrated by RT-PCR and suggests that the exon 8-like region of the dog PTHrP gene is not utilized. The exon 8-like region contained an early stop codon that was not present in exon 8 of the human PTHrP gene.


Assuntos
Processamento Alternativo , Clonagem Molecular , Doenças do Cão/genética , Neoplasias/veterinária , Proteínas/genética , Análise de Sequência de DNA , Adenocarcinoma/genética , Adenocarcinoma/veterinária , Sequência de Aminoácidos , Animais , Glândulas Apócrinas , Sequência de Bases , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/veterinária , Cães , Biblioteca Gênica , Dados de Sequência Molecular , Neoplasias/genética , Proteína Relacionada ao Hormônio Paratireóideo , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Neoplásico/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias das Glândulas Sudoríparas/genética , Neoplasias das Glândulas Sudoríparas/veterinária , Células Tumorais Cultivadas
12.
Curr Opin Rheumatol ; 13(4): 316-25, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11555736

RESUMO

The approach of gene-targeted animal models is likely the most important experimental tool contributing to recent advances in skeletal biology. Modifying the expression of a gene in vivo, and the analysis of the consequences of the mutation, are central to the understanding of gene function during development and physiology, and therefore to our understanding of the gene's role in disease states. Researchers had been limited to animal models primarily involving pharmaceutical manipulations and spontaneous mutations. With the advent of gene targeting, however, animal models that impact our understanding of metabolic bone disease have evolved dramatically. Interestingly, some genes that were expected to yield dramatic phenotypes in bone, such as estrogen receptor-alpha or osteopontin, proved to have subtle phenotypes, whereas other genes, such as interleukin-5 or osteoprotegerin, were initially identified as having a role in bone metabolism via the analysis of their phenotype after gene ablation or overexpression. Particularly important has been the advance in knowledge of osteoblast and osteoclast independent and dependent roles via the selective targeting of genes and the consequent disruption of bone formation, bone resorption, or both. Our understanding of interactions of the skeletal system with other systems, ie, the vascular system and homeostatic controls of adipogenesis, has evolved via animal models such as the matrix gla protein, knock-out, and the targeted overexpression of Delta FosB. Challenging transgenic models such as the osteopontin-deficient mice with mediators of bone remodeling like parathyroid hormone and mechanical stimuli and extending phenotype characterization to mechanistic in vitro studies of primary bone cells is providing additional insight into the mechanisms involved in pathologic states and their potentials for therapeutic strategies. This review segregates characterization of transgenic models based on the category of gene altered, eg, reproductive hormones, calcitropic hormones, growth factors and cytokines, signaling molecules, extracellular matrix molecules and "other" genes. Models are also segregated based on phenotypes that are primarily osteoclastic, osteoblastic or mixed. As the technical ability to alter gene expression negatively or positively and in a tissue-specific and temporal manner continues to evolve, there are endless possibilities for generating genetically altered animal models with which to gain insight into metabolic bone diseases.


Assuntos
Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/fisiopatologia , Modelos Animais de Doenças , Camundongos Transgênicos , Animais , Camundongos
13.
Endocrinology ; 142(5): 1975-81, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316763

RESUMO

PTH and PTH-related protein (PTHrP) are key mediators of skeletal development and homeostasis through their activation of the PTH-1 receptor. Previous studies have found that several AP-1 family members are regulated by PTH, such as c-fos, fra-1, and c-jun. There are numerous genes in the bone microenvironment that contain AP-1 sites, and different Fos family members are reported to have opposing transcriptional activities at AP-1 sites. The purpose of this study was to identify the effects of PTH on expression of the AP-1 protein complex member, fra-2, to extend our understanding of transcriptional regulators of PTH action. PTH induction of fra-2 messenger RNA (mRNA) levels in MC3T3-E1 preosteoblastic cells was maximal with 0.1 microM PTH (1-34). The expression in vitro was greatest 1 h after treatment and was present with N-terminal PTH but not PTH (7-34) or (53-84). Cycloheximide treatment induced fra-2 expression, and actinomycin D inhibited basal and PTHrP-induced expression. AP-1 protein in nuclear extracts of MC3T3-E1 cells was increased with PTH treatment at 3 h and consisted of high levels of Fra-2 protein, as evidenced by a supershift in an electrophoretic mobility shift assay and Western blot analysis. Up-regulation of steady-state fra-2 mRNA was also noted in vivo, where injection of PTH (1-34) (20 microgram) resulted in a more-than-7-fold maximal increase in fra-2 mRNA expression in the calvaria of mice, after 1 h of treatment. These data add to the transcriptional mediators induced by PTH and suggest that the interplay of AP-1 family members will provide insight into regulatory pathways of PTH and PTHrP for their anabolic and catabolic actions in bone.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fatores de Transcrição/genética , Animais , Células Cultivadas , Cicloeximida/farmacologia , DNA/metabolismo , Antígeno 2 Relacionado a Fos , Genes fos , Camundongos , Osteoblastos/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/farmacologia , RNA Mensageiro/análise , Fator de Transcrição AP-1/metabolismo
14.
Cancer Metastasis Rev ; 20(3-4): 333-49, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12085970

RESUMO

The majority of men with progressive prostate cancer develop metastases with the skeleton being the most prevalent metastatic site. Unlike many other tumors that metastasize to bone and form osteolytic lesions, prostate carcinomas form osteoblastic lesions. However, histological evaluation of these lesions reveals the presence of underlying osteoclastic activity. These lesions are painful, resulting in diminished quality of life of the patient. There is emerging evidence that prostate carcinomas establish and thrive in the skeleton due to cross-talk between the bone microenvironment and tumor cells. Bone provides chemotactic factors, adhesion factors, and growth factors that allow the prostate carcinoma cells to target and proliferate in the skeleton. The prostate carcinoma cells reciprocate through production of osteoblastic and osteolytic factors that modulate bone remodeling. The prostate carcinoma-induced osteolysis promotes release of the many growth factors within the bone extracellular matrix thus further enhancing the progression of the metastases. This review focuses on the interaction between the bone and the prostate carcinoma cells that allow for development and progression of prostate carcinoma skeletal metastases.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Próstata/patologia , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias Ósseas/metabolismo , Cálcio/metabolismo , Quimiocinas CXC/metabolismo , Humanos , Masculino , Metaloproteinases da Matriz/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Hormônios Peptídicos/metabolismo , Neoplasias da Próstata/metabolismo , Células Estromais/metabolismo
15.
Endocrinology ; 141(12): 4671-80, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11108282

RESUMO

PTH-related protein (PTHrP) acts as a paracrine and/or autocrine regulator of cell proliferation, apoptosis, and differentiation and is implicated in tooth development. The current studies employed cementoblasts to determine the role(s) and mechanisms of PTHrP in regulating cementum formation. Results demonstrated that PTHrP repressed gene expression and protein synthesis of bone sialoprotein (BSP) and abolished cementoblast-mediated biomineralization in vitro. The BSP gene inhibition required protein synthesis. The PTHrP analog (1-31) and other activators of the PKA pathway (3-isobutyl-1-methylxathine (IBMX), forskolin (FSK) and Sp-Adenosine-3', 5'-cyclic monophosphorothioate (Sp-cAMPss) also down-regulated BSP gene expression and blocked cementoblast-mediated biomineralization. In contrast, the PTHrP analog (7-34), a PTHrP antagonist, and the activators of the PKC pathway [phorbol 12-myristate 13-acetate (PMA) and phorbol 12, 13-dibutyrate (PDBu)] promoted BSP gene expression. In addition, the PKA pathway inhibitor (9-(2-tetrahydrofuryl) adenine (THFA) partially, but significantly reversed the PTHrP-mediated down-regulation of BSP gene expression. Furthermore, THFA alone significantly increased BSP messenger RNA (mRNA) expression in cementoblasts. In contrast, the inhibitor of the PKC pathway (GF109203X) did not reverse the PTHrP inhibitory effect on BSP gene expression. Furthermore, GF109203X alone dramatically reduced the BSP transcript levels. These data indicate that the cAMP/PKA pathway mediates the PTHrP-mediated down-regulation of BSP mRNA expression in cementoblasts; and furthermore, this pathway may, through an intrinsic inhibition mechanism, regulate the basal level of BSP mRNA expression. In contrast, the activation of PKC promotes BSP gene expression. These data provide new insights into the molecular mechanisms involved in PTHrP regulation of cementogenesis.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/análogos & derivados , Cemento Dentário/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas/farmacologia , Sialoglicoproteínas/genética , 1-Metil-3-Isobutilxantina/farmacologia , Linhagem Celular , Colforsina/farmacologia , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Sialoproteína de Ligação à Integrina , Cinética , Proteína Relacionada ao Hormônio Paratireóideo , Dibutirato de 12,13-Forbol/farmacologia , Proteínas/administração & dosagem , RNA Mensageiro/análise , Sistemas do Segundo Mensageiro , Acetato de Tetradecanoilforbol/farmacologia , Tionucleotídeos/farmacologia
16.
J Bone Miner Res ; 15(11): 2140-53, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11092395

RESUMO

Parathyroid hormone-related protein (PTHrP) has been implicated in regulating tooth eruption and/or development. Formation of cementum, a mineralized tissue covering the tooth root surface, is a critical biological event for tooth root development. To test the hypothesis that PTHrP targets cementoblasts (CMs) and acts to regulate cementogenesis, CM cell lines were established and their responsiveness to PTHrP stimulation was determined, in vitro. First, subclones were derived from two immortalized murine cell populations that contained CMs; SV-CM/periodontal ligament (PDL) cells were obtained from the root surface of first mandibular molars of CD-1 mice and immortalized with SV40 T-antigen (TAg), and OC-CM cell population was established from OC-TAg transgenic mice in which their cells harbor an osteocalcin (OC and/or OCN) promoter-driving immortal gene SV40 TAg. Based on our previous in situ studies, CM subclones were identified as cells expressing bone sialoprotein (BSP) and OCN transcripts, while PDL cell lines were designated as cells lacking BSP and OCN messenger RNA (mRNA). CMs exhibited a cuboidal appearance and promoted biomineralization, both in vitro and in vivo. In contrast, PDL cells (PDL subclones) displayed a spindle-shaped morphology and lacked the ability to promote mineralized nodule formation, both in vitro and in vivo. Next, using these subclones, the effect of PTHrP on cementogenesis was studied. CMs, not PDL cells, expressed PTH/PTHrP receptor mRNA and exhibited PTHrP-mediated elevation in cyclic adenosine monophosphate (cAMP) levels and c-fos gene induction. PTHrP stimulation repressed mRNA expression of BSP and OCN in CMs and blocked CM-mediated mineralization, in vitro. Collectively, these data suggest that CMs possess PTH/PTHrP receptors and, thus, are direct targets for PTHrP action during cementogenesis and that PTHrP may serve as an important regulator of cementogenesis.


Assuntos
Cemento Dentário/fisiologia , Matriz Extracelular/genética , Regulação da Expressão Gênica , Proteínas/metabolismo , Animais , Transplante de Células , Células Cultivadas , Células Clonais , Colágeno/genética , Colágeno/metabolismo , AMP Cíclico/metabolismo , Cemento Dentário/citologia , Cemento Dentário/transplante , Matriz Extracelular/metabolismo , Genes fos , Sialoproteína de Ligação à Integrina , Camundongos , Camundongos Endogâmicos , Camundongos SCID , Minerais/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Proteínas/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Ativação Transcricional
17.
Arch Oral Biol ; 45(4): 293-303, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10708669

RESUMO

Cementum is an essential component of the periodontium, but the mechanisms involved in regulating the activity of this tissue are poorly understood. As one approach to better defining the cellular and molecular properties of cementum and the associated ligament, immortalized murine cell populations expressing gene markers associated with both cementoblasts (CM) and periodontal ligament cells (PDL), termed CM/PDL cells, were established. To further characterize these cells, their responsiveness to parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP) was examined. CM/PDL cells were tested for the presence of steady state PTH-1 receptor mRNA using Northern blot analysis. In addition, the ability of PTH and PTHrP to stimulate cAMP production and c-fos mRNA expression in CM/PDL cells was determined, using a cAMP-binding assay and northern blot hybridization, respectively. Rat osteosarcoma cells (ROS 17/2.8) were used as a positive control and human periodontal ligament cells as a negative control. Northern blot analysis demonstrated that cells within the CM/PDL cell population expressed PTH-1 receptor mRNA. Both PTH (1-34) and PTHrP (1-34) increased cAMP and c-fos mRNA in CM/PDL cells. Furthermore, PTHrP treatment for either 24 or 48 h downregulated expression of transcripts for bone sialoprotein, osteocalcin and PTH-1 receptor by CM/PDL cells and abolished CM/PDL cell-mediated mineralization in vitro. These results indicate that cells within the CM/PDL population are targets for PTH and PTHrP action and that PTHrP may play an important part in regulating the biomineralization of cementum.


Assuntos
Cemento Dentário/efeitos dos fármacos , Proteínas de Neoplasias/farmacologia , Hormônio Paratireóideo/farmacologia , Ligamento Periodontal/efeitos dos fármacos , Proteínas/farmacologia , Animais , Northern Blotting , Calcificação Fisiológica/efeitos dos fármacos , Linhagem Celular , AMP Cíclico/biossíntese , Regulação para Baixo , Marcadores Genéticos , Humanos , Hibridização In Situ , Sialoproteína de Ligação à Integrina , Camundongos , Camundongos Endogâmicos , Osteocalcina/efeitos dos fármacos , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Proteína Relacionada ao Hormônio Paratireóideo , Ligamento Periodontal/citologia , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , RNA Mensageiro/análise , Ratos , Receptores de Hormônios Paratireóideos/efeitos dos fármacos , Receptores de Hormônios Paratireóideos/genética , Sialoglicoproteínas/efeitos dos fármacos , Células Tumorais Cultivadas
18.
Cancer Res ; 59(23): 6015-22, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10606251

RESUMO

Parathyroid hormone-related protein (PTHrP) is produced by prostate carcinoma cells and tumors, but little is known of its role in prostate carcinogenesis. The goal of this study was to evaluate PTHrP expression in the regulation of prostate carcinoma growth using human and animal models. PTHrP expression was assessed in prostate cancer cell lines in vitro. Seven of nine cell lines produced PTHrP, and increased expression was seen during cell proliferation. The MatLyLu rat prostate carcinoma model was used to determine the effects of PTHrP overexpression on prostate tumor growth. PTHrP overexpression did not alter proliferation of the cells in vitro. However, when PTHrP-overexpressing cells were injected into rat hind limbs, primary tumor growth and tumor size were significantly enhanced as compared with control cells. To evaluate PTHrP in human prostate carcinoma patients, immunohistochemistry was performed on metastatic bone lesions. Immunolocalization of PTHrP protein was found in the cytoplasm and nucleus of cancer cells in the bone microenvironment. Because nuclear localization of PTHrP has been associated with an inhibition of apoptosis, the ability of full-length PTHrP to protect prostate cancer cells from apoptotic stimuli was examined. Cells transfected with full-length PTHrP showed significantly increased cell survival after exposure to apoptotic agents as compared with cells producing no PTHrP (plasmid control) or cells transfected with PTHrP lacking its nuclear localization signal. To determine the mechanism of action of PTHrP in prostate cancer cells, the parathyroid hormone/PTHrP receptor status of the cells was determined. These cell lines did not demonstrate parathyroid hormone/PTHrP receptor-mediated binding of iodinated PTHrP or steady-state receptor message by Northern blot analysis, but they did have a detectable receptor message by reverse transcription-PCR analysis. In summary, PTHrP is expressed in many prostate cancer cell lines in vitro and in metastatic bone lesions in vivo. PTHrP expression positively influences primary tumor size in vivo and protects cells from apoptotic stimuli. These data suggest that PTHrP plays an important role in the promotion of prostate tumor establishment and/or progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Hormônio Paratireóideo/fisiologia , Neoplasias da Próstata/patologia , Proteínas/fisiologia , Animais , Apoptose , Divisão Celular , Humanos , Cinética , Masculino , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/genética , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
19.
Endocrinology ; 140(7): 3154-62, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10385409

RESUMO

PTH has anabolic and catabolic effects in bone through activation of the PTH-1 (PTH/PTHrP) receptor and the cAMP/protein kinase A pathway. The effects of agents that regulate cAMP in nontransformed osteoblasts in relation to cell differentiation have not been described. The purpose of this study was to determine the effects of PTH fragments with differing cAMP-stimulating activity, and nonPTH cAMP regulators on PTH-1 receptor expression and activity, and osteoblast differentiation in vitro using MC3T3-E1 and primary rat calvarial cells. PTH (1-34), but not PTH (53-84), (7-34), or PTHrP (107-139) treatment (24 h) resulted in down-regulation of steady-state messenger RNA for the PTH-1 receptor. Forskolin (a stimulator of cAMP accumulation) also down regulated the PTH-1 receptor, whereas 9-(tetrahydro-2-furyl) adenine (THFA) (an inhibitor of adenylyl cyclase) had no effect. Similarly, PTH (1-34) treatment for 48 h abolished PTHrP binding to cell surface receptors; however, neither the PTH analogs nor the cAMP regulating agents altered PTH binding or numbers of binding sites on osteoblastic cells. Basal levels of cAMP were reduced in cultured cells treated for 6 days with PTH (7-34) or THFA compared with controls. In contrast, PTH-stimulated cAMP levels were significantly increased in cultures treated with PTH (7-34) and THFA for 6 days during osteoblast differentiation and were decreased in cultures treated with PTH (1-34) and forskolin compared with controls. To evaluate effects of the cAMP pathway on osteoblast differentiation, cultures were treated continuously with PTH analogs and cAMP regulators during an 18-day differentiation regime, total RNA was isolated at multiple time points, and Northern blot analysis for osteocalcin (OCN) was performed. THFA and PTH (7-34)-treated cultures had increased OCN expression; whereas, PTH (1-34) and forskolin reduced OCN expression. Interestingly, PTH (7-34) and THFA-treated cultures had increased mineralized nodule formation, in contrast to PTH (1-34) and forskolin treatment, which reduced nodule formation. Similarly, calcium accumulation in cultures was significantly increased in the PTH (7-34) and THFA-treated cultures and reduced in the PTH (1-34) and forskolin-treated cultures. These data demonstrate that agents that increase cAMP down regulate PTH-1 receptor messenger RNA and inhibit osteoblast differentiation in vitro. Agents that reduce or block adenylyl cyclase or cAMP activity do not alter PTH-1 receptor expression or binding, but have striking effects on promoting osteoblast differentiation. We conclude that many effects of PTH on osteoblasts may be mimicked or antagonized by agents that alter cAMP activity and bypass the PTH-1 receptor.


Assuntos
AMP Cíclico/fisiologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Receptores de Hormônios Paratireóideos/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Colforsina/farmacologia , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/farmacologia , Proteínas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Receptores de Hormônios Paratireóideos/genética
20.
Prostate ; 39(3): 187-97, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10334108

RESUMO

BACKGROUND: Prostate cancer frequently metastasizes to bone, where it induces osteoblastic lesions. Parathyroid hormone-related protein (PTHrP), a product of normal and neoplastic prostate cells, may promote growth and bone metastasis of certain types of cancer. In this study, we investigated the: 1) pathogenesis and morphology of bone metastases in the MATLyLu rat prostate adenocarcinoma model, and 2) effect of PTHrP overexpression on tumor growth and incidence of bone metastasis. METHODS: MATLyLu cells were stably transfected with a PTHrP expression vector or control plasmid. PTHrP expression was determined in vitro by immunoradiometric assay and Northern blot analysis. MATLyLu cells were injected into the left ventricle of Copenhagen rats to induce bone metastases. Histology and radiography were used to quantify the size and number of bone metastases. Serum alkaline phosphatase isoenzyme concentrations and histomorphometric analysis were used to evaluate bone formation and resorption. RESULTS: All rats developed osteolytic metastases in long bones and vertebrae. There was no evidence of increased intramedullary bone formation. PTHrP overexpression by MATLyLu cells was not associated with any difference in the incidence of bone metastasis, size of metastatic foci or tumor-cell proliferation. CONCLUSIONS: The MATLyLu intracardiac injection model of prostate carcinoma is an aggressive tumor model with a high incidence of osteolytic skeletal metastases, and is not altered by increased PTHrP production by neoplastic prostate epithelial cells.


Assuntos
Adenocarcinoma/secundário , Neoplasias Ósseas/secundário , Neoplasias da Próstata/patologia , Biossíntese de Proteínas , Adenocarcinoma/metabolismo , Fosfatase Alcalina/sangue , Animais , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/epidemiologia , Neoplasias Ósseas/metabolismo , Cálcio/sangue , Divisão Celular , Modelos Animais de Doenças , Incidência , Isoenzimas/sangue , Masculino , Metástase Neoplásica , Transplante de Neoplasias , Proteína Relacionada ao Hormônio Paratireóideo , Neoplasias da Próstata/metabolismo , Proteínas/genética , RNA Mensageiro/biossíntese , Radiografia , Ratos , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA