Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Prev Res (Phila) ; 12(8): 493-506, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31266827

RESUMO

Physical activity and the prevention of weight gain decrease breast cancer incidence and improve survival. Unraveling the biological mechanisms underlying these cancer prevention effects is difficult because activity and dietary restriction are often linked. The goal of this study was to determine whether physical activity (PA), preventing weight gain via energy restriction (ER), or the combination was most effective in delaying tumor growth, reducing metastatic progression, and improving survival in the 4T1.2 mammary tumor model. Furthermore, we determined whether any of these interventions prevented the expansion of protumor immunosuppressive cells and altered the tumor microenvironment (TME). Female BALB/c mice (n = 7-20/group) were randomized to sedentary (SED) or PA wheel cages and fed ad libitum (AL) or 90% of control food intake (ER). After 8 weeks on the interventions, mice were inoculated with 5 × 104 4T1.2luc cells into the 4th mammary fat pad and continued on their respective intervention. PA+ER significantly delayed primary tumor growth (final tumor volume, 0.193 ± 0.042 vs. 0.369 ± 0.049 cm3, P < 0.001), reduced metastatic burden in the lungs (0.72 ± 0.36 vs. 16.27 ± 6.98, P = 0.054) and increased survival (median survival, 68 vs 40 days, P = 0.043) compared with SED+AL mice. PA+ER also reduced the expression level of metastatic and immunosuppressive genes and resulted in favorable changes in immune cell infiltrates in the tumor. These data suggest that both PA and ER are needed to reduce tumor growth, delay metastatic progression, and improve survival, and that this protection is associated with changes in immune-mediated mechanisms.


Assuntos
Restrição Calórica , Neoplasias Mamárias Experimentais/terapia , Condicionamento Físico Animal/fisiologia , Comportamento Sedentário , Microambiente Tumoral/imunologia , Animais , Comportamento Animal , Linhagem Celular Tumoral/transplante , Progressão da Doença , Feminino , Humanos , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/mortalidade , Neoplasias Mamárias Experimentais/patologia , Camundongos , Células Supressoras Mieloides/imunologia
2.
Small ; 14(12): e1702787, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29399951

RESUMO

Bone metastasis occurs at ≈70% frequency in metastatic breast cancer. The mechanisms used by tumors to hijack the skeleton, promote bone metastases, and confer therapeutic resistance are poorly understood. This has led to the development of various bone models to investigate the interactions between cancer cells and host bone marrow cells and related physiological changes. However, it is challenging to perform bone studies due to the difficulty in periodic sampling. Herein, a bone-on-a-chip (BC) is reported for spontaneous growth of a 3D, mineralized, collagenous bone tissue. Mature osteoblastic tissue of up to 85 µm thickness containing heavily mineralized collagen fibers naturally formed in 720 h without the aid of differentiation agents. Moreover, co-culture of metastatic breast cancer cells is examined with osteoblastic tissues. The new bone-on-a-chip design not only increases experimental throughput by miniaturization, but also maximizes the chances of cancer cell interaction with bone matrix of a concentrated surface area and facilitates easy, frequent observation. As a result, unique hallmarks of breast cancer bone colonization, previously confirmed only in vivo, are observed. The spontaneous 3D BC keeps the promise as a physiologically relevant model for the in vitro study of breast cancer bone metastasis.


Assuntos
Neoplasias Ósseas/diagnóstico , Neoplasias da Mama/diagnóstico , Microfluídica/métodos , Neoplasias Ósseas/etiologia , Neoplasias Ósseas/patologia , Neoplasias da Mama/complicações , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Humanos , Metástase Neoplásica/diagnóstico , Metástase Neoplásica/patologia
3.
Cancer Res ; 77(8): 1942-1954, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28202531

RESUMO

Little is known about how megakaryocytes may affect metastasis beyond serving as a source of platelets. In this study, we explored the functional implications of megakaryocyte accumulation in the femurs of mice after injection of metastatic or non-metastatic breast cancer cells in 4T1.2 BALB/cJ and MDA-MB-231 nude mouse models. At bone metastatic sites, but not primary growth sites, tumor growth was associated with increased megakaryopoiesis in both model systems. In the orthotopic BALB/cJ model, extramedullary hematopoiesis occurred in the spleen, resulting in a four-fold increase in megakaryocytes. In support of the hypothesis that reducing megakaryocytes may reduce metastasis, we found that thrombopoietin-deficient mice exhibited a 90% relative decrease in megakaryocytes, yet they developed more aggressive metastasis than wild-type hosts. In human clinical specimens, we observed an increase in megakaryocytes in the bone marrow of 6/8 patients with metastatic breast cancer compared with age- and gender-matched controls. Taken together, our results suggested that an increase in megakaryocytes occurring in response to metastatic cells entering the bone marrow confers some measure of protection against metastasis, challenging present views on the role of megakaryocytes in this setting. Cancer Res; 77(8); 1942-54. ©2017 AACR.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Megacariócitos/patologia , Células 3T3 , Animais , Neoplasias da Mama/sangue , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Mamárias Experimentais/sangue , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Osteoblastos/patologia
4.
Clin Exp Metastasis ; 32(4): 335-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25749879

RESUMO

Breast cancer can reoccur, often as bone metastasis, many years if not decades after the primary tumor has been treated. The factors that stimulate dormant metastases to grow are not known, but bone metastases are often associated with skeletal trauma. We used a dormancy model of MDA-MB-231BRMS1, a metastasis-suppressed human breast cancer cell line, co-cultured with MC3T3-E1 osteoblasts in a long term, three dimensional culture system to test the hypothesis that bone remodeling cytokines could stimulate dormant cells to grow. The cancer cells attached to the matrix produced by MC3T3-E1 osteoblasts but grew slowly or not at all until the addition of bone remodeling cytokines, TNFα and IL-ß. Stimulation of cell proliferation by these cytokines was suppressed with indomethacin, an inhibitor of cyclooxygenase and of prostaglandin production, or a prostaglandin E2 (PGE2) receptor antagonist. Addition of PGE2 directly to the cultures also stimulated cell proliferation. MCF-7, non-metastatic breast cancer cells, remained dormant when co-cultured with normal human osteoblast and fibroblast growth factor. Similar to the MDA-MB-231BRMS1 cells, MCF-7 proliferation increased in response to TNFα and IL-ß. These findings suggest that changes in the bone microenvironment due to inflammatory cytokines associated with bone repair or excess turnover may trigger the occurrence of latent bone metastasis.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Dinoprostona/farmacologia , Interleucina-1beta/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Células 3T3 , Animais , Remodelação Óssea , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Humanos , Indometacina/farmacologia , Interleucina-6/farmacologia , Células MCF-7 , Camundongos , Osteoblastos , Receptores de Prostaglandina E/antagonistas & inibidores , Microambiente Tumoral
5.
J Cell Physiol ; 229(4): 453-62, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24022654

RESUMO

Bone remodeling is a natural process that enables growth and maintenance of the skeleton. It involves the deposition of mineralized matrix by osteoblasts and resorption by osteoclasts. Several cancers that metastasize to bone negatively perturb the remodeling process through a series of interactions with osteoclasts, and osteoblasts. These interactions have been described as the "vicious cycle" of cancer metastasis in bone. Due to the inaccessibility of the skeletal tissue, it is difficult to study this system in vivo. In contrast, standard tissue culture lacks sufficient complexity. We have developed a specialized three-dimensional culture system that permits growth of a non-vascularized, multiple-cell-layer of mineralized osteoblastic tissue from pre-osteoblasts. In this study, the essential properties of bone remodeling were created in vitro by co-culturing the mineralized collagenous osteoblastic tissue with actively resorbing osteoclasts followed by reinfusion with proliferating pre-osteoblasts. Cell-cell and cell-matrix interactions were determined by confocal microscopy as well as by assays for cell specific cytokines and growth factors. Osteoclasts, differentiated in the presence of osteoblasts, led to degradation of the collagen-rich extracellular matrix. Further addition of metastatic breast cancer cells to the co-culture mimicked the vicious cycle; there was a further reduction in osteoblastic tissue thickness, an increase in osteoclastogenesis, chemotaxis of cancer cells to osteoclasts and formation of cancer cells into large colonies. The resulting model system permits detailed study of fundamental osteobiological and osteopathological processes in a manner that will enhance development of therapeutic interventions to skeletal diseases.


Assuntos
Neoplasias Ósseas/metabolismo , Remodelação Óssea/fisiologia , Técnicas de Cultura de Tecidos/métodos , Animais , Comunicação Celular , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Humanos , Camundongos , Osteoblastos/fisiologia , Osteoclastos , Transcriptoma
6.
Int J Breast Cancer ; 2012: 160265, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22315691

RESUMO

It is commonly accepted that cancer cells interact with host cells to create a microenvironment favoring malignant colonization. The complex bone microenvironment produces an ever changing array of cytokines and growth factors. In this study, we examined levels of MCP-1, IL-6, KC, MIP-2, VEGF, MIG, and eotaxin in femurs of athymic nude mice inoculated via intracardiac injection with MDA-MB-231(GFP) human metastatic breast cancer cells, MDA-MB-231BRMS1(GFP), a metastasis suppressed variant, or PBS. Animals were euthanized (day 3, 11, 19, 27 after injection) to examine femoral cytokine levels at various stages of cancer cell colonization. The epiphysis contained significantly more cytokines than the diaphysis except for MIG which was similar throughout the bone. Variation among femurs was evident within all groups. By day 27, MCP-1, MIG, VEGF and eotaxin levels were significantly greater in femurs of cancer cell-inoculated mice. These pro-osteoclastic and angiogenic cytokines may manipulate the bone microenvironment to enhance cancer cell colonization.

7.
J Cell Physiol ; 226(8): 2150-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21520067

RESUMO

Breast cancer cell colonization of osteoblast monolayers grown in standard tissue culture (2D) is compared to colonization of a multi-cell-layer osteoblastic tissue (3D) grown in a specialized bioreactor. Colonization of 3D tissue recapitulates events observed in clinical samples including cancer penetration of tissue, growth of microcolonies, and formation of "Single cell file" commonly observed in end-stage pathological bone tissue. By contrast, adherent cancer cell colonies did not penetrate 2D tissue and did not form cell files. Thus, it appears that 3D tissue is a more biologically (clinically) relevant model than 2D monolayers in which to study cancer cell interactions with osteoblastic tissue. This direct comparison of 2D and 3D formats is implemented using MC3T3-E1 murine osteoblasts and MDA-MB-231 human metastatic breast cancer cells, or the metastasis-suppressed line, MDA-MB-231BRMS1, for comparison. When osteoblasts were co-cultured with metastatic cells, production of osteocalcin (a mineralization marker) decreased and secretion of the pro-inflammatory cytokine IL-6 increased in both 2D and 3D formats. Cancer cell penetration of the 3D tissue coincided with a changed osteoblast morphology from cuboidal to spindle-shaped, and with osteoblasts alignment parallel to the cancer cells. Metastasis-suppressed cells did not penetrate 3D tissue, did not cause a change in osteoblast morphology or align in rows. Moreover, they proliferated much less in the 3D culture than in the 2D culture in a manner similar to their growth in bone. In both systems, the cancer cells proliferated to a greater extent with immature osteoblasts compared to more mature osteoblasts.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias da Mama/fisiopatologia , Técnicas de Cultura de Células/métodos , Osteoblastos/fisiologia , Adenocarcinoma/patologia , Animais , Reatores Biológicos , Neoplasias da Mama/patologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Humanos , Interleucina-6/metabolismo , Camundongos , Metástase Neoplásica , Osteoblastos/patologia , Osteocalcina/biossíntese
8.
Breast Cancer Res ; 12(6): 215, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21176175

RESUMO

Breast cancer frequently metastasizes to the skeleton, interrupting the normal bone remodeling process and causing bone degradation. Osteolytic lesions are the end result of osteoclast activity; however, osteoclast differentiation and activation are mediated by osteoblast production of RANKL (receptor activator for NFκB ligand) and several osteoclastogenic cytokines. Osteoblasts themselves are negatively affected by cancer cells as evidenced by an increase in apoptosis and a decrease in proteins required for new bone formation. Thus, bone loss is due to both increased activation of osteoclasts and suppression of osteoblasts. This review summarizes the current understanding of the osteolytic mechanisms of bone metastases, including a discussion of current therapies.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteólise/metabolismo , Apoptose , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Remodelação Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Neoplasias da Mama/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Osteoblastos/fisiologia , Osteoclastos/patologia , Ligante RANK/metabolismo
9.
Carcinogenesis ; 30(11): 1941-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19759193

RESUMO

Breast cancer frequently metastasizes to the skeleton resulting in bone degradation due to osteoclast activation. Metastases also downregulate differentiation and the bone-rebuilding function of osteoblasts. Moreover, cancer cells trigger osteoblast inflammatory stress responses. Pro-inflammatory mediators such as interleukin (IL)-6, monocyte chemoattractant protein-1 (MCP-1), cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), expressed by osteoblasts (MC3T3-E1) stimulated with human breast cancer cell (MDA-MB-231) conditioned medium, are pivotal to osteoclast activation and metastasis. Given that these genes are regulated by nuclear factor-kappaB (NF-kappaB), a redox-sensitive transcription factor, we hypothesized that selenium (Se) could abrogate the inflammatory response to metastatic breast cancer cells by modulating NF-kappaB. Caffeic acid phenethyl ester and parthenolide inhibited NF-kappaB activation, as seen by gel shift assays and immunoblotting for p65 in nuclear fractions, as well as decreased production of IL-6 and MCP-1. Supplementation of MC3T3-E1 with methylseleninic acid (MSA) (0.5 microM to 4 microM) reduced the activation of NF-kappaB leading to a decrease in IL-6, MCP-1, COX-2 and iNOS in response to MDA-MB-231 conditioned medium. Addition of MSA to osteoblasts for as little as 15 min suppressed activation of NF-kappaB suggesting that short-lived active metabolites might be involved. However, brief exposure to MSA also brought about an increase in selenoprotein glutathione peroxidase 1. In summary, our data indicate that the osteoblast response to metastatic breast cancer cells is regulated by NF-kappaB activation, which can be effectively suppressed by MSA either through short-lived active metabolites and/or selenoproteins. Thus, Se supplementation may prevent the osteoblast inflammatory response or dampen the vicious cycle established when breast cancer cells, osteoblasts and osteoclasts interact.


Assuntos
Neoplasias da Mama/metabolismo , Compostos Organosselênicos/farmacologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Selênio/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quimiocina CCL2/biossíntese , Meios de Cultivo Condicionados/farmacologia , Ciclo-Oxigenase 2/metabolismo , Feminino , Humanos , Inflamação , Interleucina-6/biossíntese , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Osteoblastos/efeitos dos fármacos
10.
J Cell Biochem ; 103(4): 1101-10, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17668452

RESUMO

The calcium phosphate-based skeleton of vertebrates serves as the major reservoir for metabolically available calcium ions. The skeleton is formed by osteoblasts which first secrete a proteinaceous matrix and then provide Ca++ for the calcification process. The two calcium efflux ports found in most cells are the plasma membrane Ca-ATPase (PMCA) and the sodium-calcium exchanger (NCX). In osteoblasts, PMCA and NCX are located on opposing sides of the cell with NCX facing the mineralizing bone surface. Two isoforms of NCX have been identified in osteoblasts NCX1, and NCX3. The purpose of this study was to determine the extent to which each of the two NCX isoforms support delivery of Ca++ into sites of calcification and to discern if one could compensate for the other. SiRNA technology was used to knockdown each isoform separately in MC3T3-E1 osteoblasts. Osteoblasts in which either NCX1 or NCX3 was impaired were tested for Ca++ efflux using the Ca++ specific fluorophore, fluo-4, in a sodium-dependent calcium uptake assay adapted for image analysis. NCX3 was found to serve as a major contributor of Ca++ translocation out of osteoblasts into calcifying bone matrix. NCX1 had little to no involvement.


Assuntos
Calcificação Fisiológica , Cálcio/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Osteoblastos/fisiologia , Trocador de Sódio e Cálcio/fisiologia , Células 3T3 , Animais , Transporte Biológico Ativo , Matriz Óssea/fisiologia , Linhagem Celular , Membrana Celular/fisiologia , Camundongos , ATPases Transportadoras de Cálcio da Membrana Plasmática/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Trocador de Sódio e Cálcio/genética
11.
J Cell Biochem ; 102(2): 463-72, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17372935

RESUMO

This study focused on the differential expression levels of proteins that may exist between bone-derived and marrow-derived vascular endothelial cells (BVEC and MVEC). The vascular cells were isolated from trabecular bone regions and central marrow cavity regions of mouse long bones. Cells were cultured for 1 week to expand the population then separated from non-vascular cells using biotinylated isolectin B4, streptavidin-coated metallic microbeads, and a magnetic column. After an additional week of culture time, RNA was isolated from both cell types and compared using microarray analysis. RT-PCR was used to confirm and relatively quantitate the RNA messages. The bone-derived cells expressed more aldehyde dehydrogenase 3A1 (ALDH3A1), Secreted Modular Calcium-2 (SMOC-2), CCAAT enhancer binding protein (C/EBP-beta), matrix metalloproteinase 13 (MMP-13), and annexin 8 (ANX8) than the marrow-derived cells. Spalpha and matrix GLA-protein (MGP) were produced in greater abundance by the marrow-derived cells. This study reveals that there are profound and unique differences between the vasculature of the metaphysis as compared to that of the central marrow cavity. The unique array of proteins expressed by the bone-derived endothelial cells may support growth of tumors from cancer cells that frequently metastasize and lodge in the trabecular bone regions.


Assuntos
Células da Medula Óssea/citologia , Osso e Ossos/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Animais , Células da Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Feminino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
12.
J Cell Biochem ; 98(5): 1221-9, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16514647

RESUMO

Breast cancer cells frequently metastasize to the ends of long bones, ribs and vertebrae, structures which contain a rich microvasculature that is closely juxtaposed to metabolically active trabecular bone surfaces. This study focuses on the effects of osteoblast secretions on the surface presentation of adhesive proteins on skeletal vascular endothelial cells. Vascular endothelial cells were isolated from trabecular bone regions of the long bones of 7-week-old Swiss Webster mice and also from the central marrow cavity where trabecular bone is absent. Both types of endothelial cells were placed in culture for 7 days, then exposed 24 h to conditioned media from MC3T3-E1 osteoblasts. Conditioned medium (CM) from two different stages of osteoblast development were tested: (1) from immature MC3T3-E1 cells cultured for 5-7 days and (2) from mature MC3T3-E1 cells cultured for 28-30 days. The immature osteoblasts were in a stage of rapid proliferation; the mature osteoblasts formed a matrix that mineralized. Following exposure to the conditioned media, the vascular cells were exposed to anti-P-selectin, anti-E-selectin, anti-ICAM-1, and anti-VCAM-1 to detect the corresponding adhesive proteins on their surfaces. Breast cancer cells are known to bind to these adhesive proteins. Of the four proteins evaluated, E-selectin was consistently found on more cell surfaces (approximately 30%) of bone-derived vascular endothelial cells (BVECs) when exposed to the immature CM whereas vascular endothelial cells from marrow (MVECs) did not show this response to either immature CM or mature CM. These studies suggest that the BVEC blood vessels near immature bone cells express more surface adhesive protein that could enhance entrapment and extravasation of breast cancer cells. Once cancer cells have undergone extravasation into marrow adjacent to bone, they could be readily attracted to nearby bone surfaces.


Assuntos
Selectina E/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Meios de Cultivo Condicionados , Camundongos
13.
J Cell Biochem ; 97(2): 288-302, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16173048

RESUMO

The focus of this study was to gain insight into the role(s) of osteonectin in the preferential metastasis of breast cancer cells to bone. Osteonectin was isolated from conditioned media of several cell lines including breast cancer (MDA-MB-435, MDA-MB-468), osteoblasts (hFOB1.19), non-neoplastic breast epithelial (hTERT-HME1), and vascular endothelial cells isolated from a bone biopsy (HBME-1). Chemical/physical properties of osteonectin from these five sources was analyzed to determine if unique configurations of osteonectin exist and therefore identify a chemotactic isoform. Osteonectin from all sources had a molecular weight of approximately 46 kDa, N-linked glycosylation, and undetectable phosphorylated serines, sialic acids and O-linked oligosaccharides. The cDNA for osteonectin from the breast cancer, osteoblast, and breast epithelial cell lines was identical, while the vascular endothelial cell cDNA contained point mutations that resulted in eight amino acid substitutions. Bone-derived osteonectin was then analyzed to assess its influence on breast cancer cell motility and migration. Although osteonectin increased undirected MDA-MB-231 cell motility, it did not chemoattract the same breast cancer cell line. However, the breast cancer cells did migrate toward the known chemoattractant vitronectin and to bone extracts derived from wild-type and osteonectin-null mice. Migration to vitronectin was enhanced when osteonectin was also present. We concluded that osteonectin was not a chemotactic factor. However, through its anti-adhesive properties, osteonectin induced undirected breast cancer cell motility, and may have enhanced chemoattraction to vitronectin.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Osteonectina/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Quimiotaxia , DNA Complementar/química , Glicosilação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Osteonectina/genética , Osteonectina/metabolismo , Fosforilação , Vitronectina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA