Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Curr Oral Health Rep ; 5(1): 7-12, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29910997

RESUMO

PURPOSE OF REVIEW: Dental care is an essential component in the comprehensive treatment for the cancer patient. As such, a review of the literature was completed to determine the relationships between periodontal and dental care in the cancer patient and provide strategic suggestions. RECENT FINDINGS: Periodontal treatment must be personalized depending on the patient's current oral health status, systemic status, and progress in treatment. Oral mucositis, periodontal status, and osteonecrosis of the jaw (ONJ) remain periodontal concerns in the cancer patient. Contributing factors of ONJ include root amputation (OR= 6.64), extraction of a single tooth (OR=3.7), severe tooth mobility (OR = 3.60), and unclosed wound (OR = 2.51). SUMMARY: Preventive maintenance, oral hygiene instruction, use of fluoride and chlorhexidine are all important therapeutic strategies. If extractions are required in patients who have received bone modifying drug infusions, flap management and primary wound closure is needed to reduce the risk of complications.

2.
J Cell Biochem ; 118(8): 1956-1964, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27982511

RESUMO

Metastatic cancer is the leading cause of all cancer related deaths. Prostate cancer (PCa) metastasizes preferentially to the bone marrow, specifically within the endosteal niche. Endosteal cells secrete homing molecules that may recruit PCa cells to the bone marrow. Once there, the biochemical signature of this niche regulates PCa fate including cellular dormancy or cell cycle arrest, reactivation and resistance to chemotherapeutics. Growth factors, interleukins, adhesion molecules, as well as extra-cellular matrix proteins can collectively change the phenotype of PCa cells. Understanding the biochemical signature of endosteal niche parasitism by PCa is imperative for the establishment of new and innovative therapeutic strategies. This review seeks to summarize these important niche signatures and the potential therapeutic approaches to target metastatic PCa within the bone marrow hematopoietic stem cell (HSC) niche. J. Cell. Biochem. 118: 1956-1964, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Neoplasias da Medula Óssea/genética , Medula Óssea/patologia , Regulação Neoplásica da Expressão Gênica , Células-Tronco Hematopoéticas/patologia , Periósteo/patologia , Neoplasias da Próstata/genética , Nicho de Células-Tronco/genética , Medula Óssea/imunologia , Neoplasias da Medula Óssea/imunologia , Neoplasias da Medula Óssea/secundário , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Movimento Celular , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/imunologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interleucinas/genética , Interleucinas/imunologia , Masculino , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/patologia , Periósteo/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Transdução de Sinais , Nicho de Células-Tronco/imunologia
3.
J Bone Oncol ; 5(3): 117-120, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27761370

RESUMO

Approximately 80% of prostate cancers exhibit some degree of bone metastasis. The role of the bone marrow and the hematopoietic stem cell (HSC) niche in attracting metastatic cells and maintaining dormancy of disseminated tumor cells (DTCs) is an increasingly important topic towards the development of novel prostate cancer therapies. This paper reviews aspects of the HSC niche that lead to prostate cancer cell homing and dormancy in the bone marrow. This review also discusses the role of DTCs in the niche environment and discusses the role of erythropoietin in targeting DTCs within the HSC niche.

4.
Artigo em Inglês | MEDLINE | ID: mdl-26097863

RESUMO

Prostate cancer (PCa) is one of the most common neoplasms that metastasize to bone. The aim of this study was to determine if osteoclasts play a role in the seeding of disseminated tumor cells to the bone marrow by mobilizing hematopoietic stem cells (HSCs) out of their marrow niche. Human PC-3Luc cells were introduced into male SCID mice by intracardiac (i.c.) injection after mice were treated with the antiresorptive agent Zoledronic Acid (bisphosphonate (BP)) and/or AMD3100, which mobilizes HSCs out of the marrow. Short term homing of PC-3 was assessed at 24 hours by QPCR for human Alu and luciferase and HSC number was determined by FACS. Mice also received pre and/or post treatments of BP by intraperiteneal (i.p.) injections, in addition to PC-3 luc by intratibial (i.t.) injections. TRAP assays were used to determine the osteoclast (OC) number in both studies. AMD3100 enhanced the release of HSCs from the bone marrow, while BP increased the retention of HSCs. PCa entry into bone was facilitated in AMD3100, BP, and AMD3100+BP treatments. Before PCa injection, the number of TRAP+ OC was increased in mice treated with AMD3100, while treatment with BP resulted in relatively lower TRAP+ OCs. TRAP+ OCs were not detected in the AMD3100 + BP treatment. After PCa injection, however, the number of TRAP+ OCs was dramatically increased, but did not differ significantly amongst the treatment groups. The pre and post BP treatments in the Nude mice decreased the size of PCa lesions in the tibia compared to the control. The results indicate that OC activation is not necessary for PCa metastasis to bone at the earliest stages. These findings are critical in proving that OCs' contribution to metastasis occur during the growth phase of the tumor rather than at the initiation phase.

5.
J Dent Res ; 94(3): 455-63, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25586589

RESUMO

Recent studies have demonstrated that erythropoietin (EPO) has extensive nonhematopoietic biological functions. However, little is known about how EPO regulates bone formation, although several studies suggested that EPO can affect bone homeostasis. In this study, we investigated the effects of EPO on the communication between osteoclasts and osteoblasts through the ephrinB2/EphB4 signaling pathway. We found that EPO slightly promotes osteoblastic differentiation with the increased expression of EphB4 in ST2 cells. However, EPO increased the expression of Nfatc1 and ephrinB2 but decreased the expression of Mmp9 in RAW264.7 cells, resulting in an increase of ephrinB2-expressing osteoclasts and a decrease in resorption activity. The stimulation of ephrinB2/EphB4 signaling via ephrinB2-Fc significantly promoted EPO-mediated osteoblastic differentiation in ST2 cells. EphB4 knockdown through EphB4 shRNA inhibited EPO-mediated osteoblastic phenotypes. Furthermore, in vivo assays clearly demonstrated that EPO efficiently induces new bone formation in the alveolar bone regeneration model. Taken together, these results suggest that ephrinB2/EphB4 signaling may play an important role in EPO-mediated bone formation.


Assuntos
Efrina-B2/efeitos dos fármacos , Eritropoetina/farmacologia , Osteogênese/efeitos dos fármacos , Receptor EphB4/efeitos dos fármacos , Processo Alveolar/efeitos dos fármacos , Animais , Regeneração Óssea/efeitos dos fármacos , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Eritropoetina/uso terapêutico , Técnicas de Silenciamento de Genes , Inativação Gênica , Incisivo/cirurgia , Macrófagos/efeitos dos fármacos , Masculino , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Fatores de Transcrição NFATC/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Extração Dentária , Alvéolo Dental/efeitos dos fármacos
6.
Growth Factors ; 30(1): 22-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22117584

RESUMO

It has been well established that blood and bone share a unique, regulatory relationship with one another, though the specifics of this relationship still remain unanswered. Erythropoietin (Epo) is known primarily for its role as a hematopoietic hormone. However, after the discovery of Epo receptor outside the hematopoietic tissues, Epo has been avidly studied for its possible nonhematopoietic effects. It has been proposed that Epo interacts with bone both directly, by activating bone marrow stromal cells, and indirectly, through signaling pathways on hematopoietic stem cells. Yet, the role of Epo in regulating skeletal maintenance and regeneration remains controversial. Here, we review the current state of knowledge pertaining to the effects of Epo on the skeleton.


Assuntos
Eritropoetina/metabolismo , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Eritropoetina/farmacologia , Feminino , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese/efeitos dos fármacos , Coelhos
7.
Endocrinology ; 152(12): 4525-36, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22045660

RESUMO

PTH stimulates bone formation and increases hematopoietic stem cells through mechanisms as yet uncertain. The purpose of this study was to identify mechanisms by which PTH links actions on cells of hematopoietic origin with osteoblast-mediated bone formation. C57B6 mice (10 d) were nonlethally irradiated and then administered PTH for 5-20 d. Irradiation reduced bone marrow cellularity with retention of cells lining trabeculae. PTH anabolic activity was greater in irradiated vs. nonirradiated mice, which could not be accounted for by altered osteoblasts directly or osteoclasts but instead via an altered bone marrow microenvironment. Irradiation increased fibroblast growth factor 2, TGFß, and IL-6 mRNA levels in the bone marrow in vivo. Irradiation decreased B220 cell numbers, whereas the percent of Lin(-)Sca-1(+)c-kit(+) (LSK), CD11b(+), CD68(+), CD41(+), Lin(-)CD29(+)Sca-1(+) cells, and proliferating CD45(-)Nestin(+) cells was increased. Megakaryocyte numbers were reduced with irradiation and located more closely to trabecular surfaces with irradiation and PTH. Bone marrow TGFß was increased in irradiated PTH-treated mice, and inhibition of TGFß blocked the PTH augmentation of bone in irradiated mice. In conclusion, irradiation created a permissive environment for anabolic actions of PTH that was TGFß dependent but osteoclast independent and suggests that a nonosteoclast source of TGFß drives mesenchymal stem cell recruitment to support PTH anabolic actions.


Assuntos
Medula Óssea/efeitos da radiação , Microambiente Celular/efeitos da radiação , Hormônio Paratireóideo/metabolismo , Animais , Contagem de Células , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos , Hormônio Paratireóideo/administração & dosagem , RNA Mensageiro/efeitos da radiação , Fator de Crescimento Transformador beta/fisiologia , Irradiação Corporal Total
8.
Leukemia ; 22(5): 941-50, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18305549

RESUMO

In post-fetal life, hematopoiesis occurs in unique microenvironments or 'niches' in the marrow. Niches facilitate the maintenance of hematopoietic stem cells (HSCs) as unipotent, while supporting lineage commitment of the expanding blood populations. As the physical locale that regulates HSC function, the niche function is vitally important to the survival of the organism. This places considerable selective pressure on HSCs, as only those that are able to engage the niche in the appropriate context are likely to be maintained as stem cells. Since niches are central regulators of stem cell function, it is not surprising that molecular parasites like neoplasms are likely to seek out opportunities to harvest resources from the niche environment. As such, the niche may unwittingly participate in tumorigenesis as a leukemic or neoplastic niche. The niche may also promote metastasis or chemo-resistance of hematogenous neoplasms or solid tumors. This review focuses on what is known about the physical structures of the niche, how the niche participates in hematopoiesis and neoplastic growth and what molecules are involved. Further understanding of the interactions between stem cells and the niche may be useful for developing therapeutic strategies.


Assuntos
Células da Medula Óssea , Medula Óssea/fisiologia , Animais , Neoplasias da Medula Óssea , Citocinas/fisiologia , Hematopoese , Células-Tronco Hematopoéticas , Células-Tronco Mesenquimais , Metástase Neoplásica
9.
J Periodontal Res ; 42(1): 45-52, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17214639

RESUMO

BACKGROUND AND OBJECTIVE: Lead is known to have significant effects on bone metabolism and the immune system. This study tested the hypothesis that lead exposure affects periodontitis in adults. MATERIAL AND METHODS: This study used the data from the Third National Health and Nutrition Examination Survey (NHANES III, 1988-94). It analyzed data from 2500 men and 2399 women, 20-56 yr old, who received complete periodontal examination. Periodontitis was defined as the presence of > 20% of mesial sites with >or= 4 mm of attachment loss. Lead exposure was grouped into three categories: < 3; 3-7; and > 7 microg/dL. Covariates were cotinine levels, poverty ratio, race/ethnicity, education, bone mineral density, diabetes, calcium intake, dental visit, and menopause (for women). All analyses were performed separately for men and women and considering the effect design. Univariate, bivariate, and stratified analysis was followed by multivariable analysis by estimating prevalence ratios through poisson regression. RESULTS: After adjustment for confounders, the prevalence ratios, comparing those with a lead blood level of > 7 microg/dL to those with a lead blood level of < 3 microg/dL was 1.70 (95% confidence interval (CI): 1.02, 2.85) for men and 3.80 (95% CI: 1.66, 8.73) for women. CONCLUSION: The lead blood level was positively and statistically associated with periodontitis for both men and women. Considering the public health importance of periodontitis and lead exposure, further studies are necessary to confirm this association.


Assuntos
Exposição Ambiental/estatística & dados numéricos , Chumbo/sangue , Periodontite/epidemiologia , Adulto , Densidade Óssea , Cálcio da Dieta/administração & dosagem , Fatores de Confusão Epidemiológicos , Cotinina/sangue , Assistência Odontológica/estatística & dados numéricos , Diabetes Mellitus/epidemiologia , Escolaridade , Etnicidade/estatística & dados numéricos , Feminino , Humanos , Masculino , Menopausa , Pessoa de Meia-Idade , Perda da Inserção Periodontal/epidemiologia , Pobreza/estatística & dados numéricos , Fatores Sexuais , Estados Unidos/epidemiologia
10.
BMC Cancer ; 6: 195, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16859559

RESUMO

BACKGROUND: The chemokine stromal derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 have been demonstrated to be crucial for the homing of stem cells and prostate cancers to the marrow. While screening prostate cancers for CXCL12-responsive adhesion molecules, we identified CD164 (MGC-24) as a potential regulator of homing. CD164 is known to function as a receptor that regulates stem cell localization to the bone marrow. RESULTS: Using prostate cancer cell lines, it was demonstrated that CXCL12 induced both the expression of CD164 mRNA and protein. Functional studies demonstrated that blocking CD164 on prostate cancer cell lines reduced the ability of these cells to adhere to human bone marrow endothelial cells, and invade into extracellular matrices. Human tissue microarrays stained for CD164 demonstrated a positive correlation with prostate-specific antigen levels, while its expression was negatively correlated with the expression of androgen receptor. CONCLUSION: Our findings suggest that CD164 may participate in the localization of prostate cancer cells to the marrow and is further evidence that tumor metastasis and hematopoietic stem cell trafficking may involve similar processes.


Assuntos
Neoplasias da Medula Óssea/secundário , Endolina/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias da Próstata/patologia , Neoplasias da Medula Óssea/fisiopatologia , Adesão Celular , Quimiocina CXCL12 , Quimiocinas CXC/fisiologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Antígeno Prostático Específico , Células Tumorais Cultivadas
11.
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
12.
Cytokine ; 32(3-4): 155-62, 2005 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16256361

RESUMO

Osteoblasts constitute part of the stromal cell support system in marrow for hematopoiesis, however little is known as to how they interact with hematopoietic stem cells (HSCs). In vitro studies have demonstrated that the survival of HSCs in co-culture with osteoblasts requires intimate cell-to-cell contact. This suggests that the osteoblast-derived factor(s) that supports stem cell activities are produced in very small quantities, are rapidly turned over, may be membrane-anchored and/or require the engagement of cell-cell adhesion molecules that are yet to be determined. In the present report we found that the survival of hematopoietic progenitor cells on osteoblasts is dependent upon the engagement of VLA-4 (alpha4beta1) and VLA-5 (alpha5beta1) receptors using function blocking antibodies. Cell-to-cell contact is required to support progenitor activity, but can be replaced if receptor-ligand engagement of the VLA-4 and LFA-1 complexes is provided through the use of recombinant ligands (fibronectin, ICAM-1, VCAM-1). Moreover, once these receptors were engaged, conditioned medium derived from HSCs grown on osteoblast ligands supported significantly greater hematopoietic progenitors in vitro than did osteoblast-conditioned or HSC-conditioned medium alone. While the molecules present in the co-cultured medium remain to be identified, the data suggest that hematopoietic cells cooperate with osteoblasts to assemble the various marrow microenvironments by directing the synthesis of osteoblast-derived cytokines to improve HSC survival.


Assuntos
Comunicação Celular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Osteoblastos/fisiologia , Adulto , Antígenos CD34/metabolismo , Adesão Celular/fisiologia , Moléculas de Adesão Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Citocinas/fisiologia , Células-Tronco Hematopoéticas/citologia , Humanos , Osteoblastos/citologia
13.
Braz. j. med. biol. res ; 38(10): 1449-1454, Oct. 2005.
Artigo em Inglês | LILACS | ID: lil-409271

RESUMO

Bone marrow stromal cells are critical regulators of hematopoiesis. Osteoblasts are part of the stromal cell support system in bone marrow and may be derived from a common precursor. Several studies suggested that osteoblasts regulate hematopoiesis, yet the entire mechanism is not understood. It is clear, however, that both hematopoietic precursors and osteoblasts interact for the production of osteoclasts and the activation of resorption. We observed that hematopoietic stem cells (HSCs) regulate osteoblastic secretion of various growth factors, and that osteoblasts express some soluble factors exclusively in the presence of HSCs. Osteoblasts and hematopoietic cells are closely associated with each other in the bone marrow, suggesting a reciprocal relationship between them to develop the HSC niche. One critical component regulating the niche is stromal-derived factor-1 (SDF-1) and its receptor CXCR4 which regulates stem cell homing and, as we have recently demonstrated, plays a crucial role in facilitating those tumors which metastasize to bone. Osteoblasts produce abundant amounts of SDF-1 and therefore osteoblasts play an important role in metastasis. These findings are discussed in the context of the role of osteoblasts in marrow function in health and disease.


Assuntos
Animais , Humanos , Neoplasias Ósseas/secundário , Quimiocinas CXC/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Osteoblastos/fisiologia , /metabolismo , Neoplasias Ósseas/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Células Estromais/metabolismo
14.
Braz J Med Biol Res ; 38(10): 1449-54, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16172737

RESUMO

Bone marrow stromal cells are critical regulators of hematopoiesis. Osteoblasts are part of the stromal cell support system in bone marrow and may be derived from a common precursor. Several studies suggested that osteoblasts regulate hematopoiesis, yet the entire mechanism is not understood. It is clear, however, that both hematopoietic precursors and osteoblasts interact for the production of osteoclasts and the activation of resorption. We observed that hematopoietic stem cells (HSCs) regulate osteoblastic secretion of various growth factors, and that osteoblasts express some soluble factors exclusively in the presence of HSCs. Osteoblasts and hematopoietic cells are closely associated with each other in the bone marrow, suggesting a reciprocal relationship between them to develop the HSC niche. One critical component regulating the niche is stromal-derived factor-1 (SDF-1) and its receptor CXCR4 which regulates stem cell homing and, as we have recently demonstrated, plays a crucial role in facilitating those tumors which metastasize to bone. Osteoblasts produce abundant amounts of SDF-1 and therefore osteoblasts play an important role in metastasis. These findings are discussed in the context of the role of osteoblasts in marrow function in health and disease.


Assuntos
Neoplasias Ósseas/secundário , Quimiocinas CXC/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Osteoblastos/fisiologia , Receptores CXCR4/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Quimiocina CXCL12 , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , Células Estromais/metabolismo
15.
Br J Haematol ; 124(4): 534-46, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14984505

RESUMO

Haematopoietic progenitor cells proliferate and develop predominantly when they adhere to bone marrow stromal cells such as osteoblasts. Therefore, changes in adhesion may be a common mechanism by which stem cells survive, mature and properly traffic between the bone marrow and the circulation. To characterize these adhesion molecules, we reduced the bone marrow cavity to a simple adhesion assay between KG1a (a CD34+ haematopoietic cell line) and osteosarcoma monolayers (MG-63 or SaOS-2). The data demonstrated that adhesion was mediated by cell-to-cell rather than cell-to-matrix contact, was sensitive to trypsin, calcium chelators and glycosylation inhibitors. Selective pretreatment attributed the constitutive binding to N-linked glycans on KG1a. When carboprocessing was inhibited later at the high mannose intermediate (via deoxymannojirimycin), adhesion was retained. Surprisingly, binding of KG1a to SaOS-2 increased past constitutive levels as doses of tunicamycin or deoxymannojirimycin dropped. Selective pretreatment suggested that this 'inducible' binding resides with molecule(s) on SaOS-2. If the terminal sialic acid was digested (via neuraminidase), this induced response was duplicated. These data, verified in primary cells, suggest that the initial tethering between blood and bone cells in this model is probably due to heavily glycosylated, rapidly processed protein(s) on both cell types.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Osteoblastos/fisiologia , Polissacarídeos/fisiologia , Antígenos CD34/análise , Adesão Celular/fisiologia , Moléculas de Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Glicosilação , Humanos , Manose/fisiologia , Ácido N-Acetilneuramínico/fisiologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
16.
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
17.
J Clin Invest ; 106(11): 1331-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11104786

RESUMO

The chemokine stromal-derived factor-1 (SDF-1) controls many aspects of stem cell function. Details of its regulation and sites of production are currently unknown. We report that in the bone marrow, SDF-1 is produced mainly by immature osteoblasts and endothelial cells. Conditioning with DNA-damaging agents (ionizing irradiation, cyclophosphamide, and 5-fluorouracil) caused an increase in SDF-1 expression and in CXCR4-dependent homing and repopulation by human stem cells transplanted into NOD/SCID mice. Our findings suggest that immature osteoblasts and endothelial cells control stem cell homing, retention, and repopulation by secreting SDF-1, which also participates in host defense responses to DNA damage.


Assuntos
Quimiocinas CXC/genética , Dano ao DNA , Células-Tronco/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem Celular , Células Cultivadas , Quimiocina CXCL12 , Ciclofosfamida/farmacologia , Relação Dose-Resposta à Radiação , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Citometria de Fluxo , Fluoruracila/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/citologia , Células Tumorais Cultivadas
18.
Exp Hematol ; 28(7): 815-25, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10907643

RESUMO

OBJECTIVE: Much remains to be learned about the intimate relationship between bone marrow and its surrounding tissue: the bone. We hypothesized that bone marrow accessory cell populations might regulate the development of human bone precursor cells. MATERIALS AND METHODS: We used immunologic phenotyping, and isolation methods to fractionate subpopulations of nonadherent, low-density (NALD) human bone marrow cells. These cells were examined for their ability to support the serum-free survival, proliferation, and expression of bone proteins by highly purified populations of human bone precursor cells. Quantitative assessment of the accessory cell populations as well as human bone precursor cells phenotype was performed using multiparameter flow cytometry. Bone protein expression was evaluated by immunocytochemistry, Western analysis, and enzymatic analysis (for alkaline phosphatase activity). RESULTS: Human bone marrow contains a cell population that stimulates the development of purified bone precursor cells. Feeder-layer studies demonstrate that these osteopoietic accessory cells (OACs) do not require cell-cell interaction to promote bone precursor cell development but, rather, produce soluble molecules responsible for their effects. Flow cytometric analyses reveal that bone marrow derived B cells, T cells, macrophages, natural killer cells, and endothelial cells do not produce this stimulatory factor. The (growth) factor cannot be replaced by addition of exogenous cytokines. The isolation of human transforming growth factor beta receptor type II (TGF-betaRII)-positive cells increases OAC-specific activity in bone cell ex vivo expansion cultures. Moreover, isolation of OAC bone marrow cells characterized by high TGF-betaRII expression, relatively low cellular complexity, and small size yields a population that is highly enriched for OACs. CONCLUSION: We conclude that human bone marrow contains a population of OACs that are an obligate requirement for the early phases of bone cell development ex vivo.


Assuntos
Células Apresentadoras de Antígenos/fisiologia , Células da Medula Óssea/fisiologia , Osteoblastos/fisiologia , Adesão Celular , Separação Celular , Células Cultivadas , Citocinas/biossíntese , Citometria de Fluxo , Humanos , Imunofenotipagem , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
J Cell Biochem ; 78(4): 607-16, 2000 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-10861858

RESUMO

Tumor cells frequently have pronounced effects on the skeleton including bone destruction, bone pain, hypercalcemia, and depletion of bone marrow cells. Despite the serious sequelae associated with skeletal metastasis, the mechanisms by which tumor cells alter bone homeostasis remain largely unknown. In this study, we tested the hypothesis that the disruption of bone homeostasis by tumor cells is due in part to the ability of tumor cells to upregulate osteopontin (OPN) mRNA in osteoblasts. Conditioned media were collected from tumor cells that elicit either osteolytic (MCF-7, PC-3) or osteoblastic responses (LNCaP) in animal models and their effects on OPN gene expression were compared using an osteoblast precursor cell line, MC3T3-E1 cells. Secretory products from osteolytic but not osteoblastic tumor cell lines were demonstrated to upregulate OPN in osteoblasts while inhibiting osteoblast proliferation and differentiation. Signal transduction studies revealed that regulation of OPN was dependent on both protein kinase C (PKC) and the mitogen-activated protein (MAP) kinase cascade. These results suggest that the upregulation of OPN may play a key role in the development of osteolytic lesions. Furthermore, these results suggest that drugs that prevent activation of the MAP kinase pathway may be efficacious in the treatment of osteolytic metastases.


Assuntos
Osteoblastos/metabolismo , Sialoglicoproteínas/metabolismo , Regulação para Cima , Animais , Western Blotting , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias da Mama/metabolismo , Diferenciação Celular , Meios de Cultivo Condicionados/farmacologia , DNA/biossíntese , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Osteopontina , Neoplasias da Próstata/metabolismo , Biossíntese de Proteínas , Proteína Quinase C/metabolismo , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas
20.
Br J Haematol ; 108(2): 275-83, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10691855

RESUMO

Recent investigations have demonstrated that macrophage inhibitory protein 1alpha (MIP-1alpha) plays a critical role in haematopoiesis. In part, MIP-1alpha limits the differentiation of early haematopoietic cells, thereby ensuring that sufficient quantities of blood precursors are available to meet haematopoietic demands. MIP-1alpha is produced by cells of the marrow microenvironment (marrow stromal cells) in response to a variety of stimuli, including interleukin 1beta (IL-1beta) and tumour necrosis factor alpha (TNF-alpha). Our recent investigations demonstrated that normal human osteoblast-like cells (HOBs) maintain the early phenotype of haematopoietic precursors, like other members of the bone marrow stroma. Although the precise molecular mechanisms for these observations have not been determined, the production of MIP-1alpha remains one such possibility. In the present study, we investigated whether cells of the osteoblast lineage under basal, IL-1beta and/or TNF-alpha stimulation produce MIP-1alpha. We observed that IL-1beta and TNF-alpha stimulated HOBs and human osteosarcoma cells to rapidly express MIP-1alpha mRNA and to secrete large quantities of the protein. MIP-1alpha mRNA and protein was not, however, detected under basal conditions. Perhaps more importantly, enriched human CD34+ bone marrow cells in co-culture may be capable of stimulating the expression of MIP-1alpha mRNA by HOBs in vitro. These findings suggest that human osteoblast-like cells may produce MIP-1alpha in vivo to support haematopoiesis at sites where osteoblasts and haematopoietic cells are closely associated.


Assuntos
Neoplasias Ósseas/metabolismo , Interleucina-1/farmacologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Osteoblastos/metabolismo , Osteossarcoma/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Quimiocina CCL3 , Quimiocina CCL4 , Ensaio de Imunoadsorção Enzimática , Humanos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA