Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 102021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34378533

RESUMO

Bone marrow adipocytes accumulate with age and in diverse disease states. However, their origins and adaptations in these conditions remain unclear, impairing our understanding of their context-specific endocrine functions and relationship with surrounding tissues. In this study, by analyzing bone and adipose tissues in the lipodystrophic 'fat-free' mouse, we define a novel, secondary adipogenesis pathway that relies on the recruitment of adiponectin-negative stromal progenitors. This pathway is unique to the bone marrow and is activated with age and in states of metabolic stress in the fat-free mouse model, resulting in the expansion of bone marrow adipocytes specialized for lipid storage with compromised lipid mobilization and cytokine expression within regions traditionally devoted to hematopoiesis. This finding further distinguishes bone marrow from peripheral adipocytes and contributes to our understanding of bone marrow adipocyte origins, adaptations, and relationships with surrounding tissues with age and disease.


Assuntos
Adipócitos/fisiologia , Adipogenia/fisiologia , Medula Óssea/fisiologia , Hematopoese/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiologia , Fatores Etários , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Diferenciação Celular , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/metabolismo , Osteoblastos/fisiologia
2.
Sci Rep ; 9(1): 17427, 2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31758074

RESUMO

Adipocytes within the skeleton are collectively termed bone marrow adipose tissue (BMAT). BMAT contributes to peripheral and local metabolism, however, its capacity for cell-autonomous expression of uncoupling protein 1 (UCP1), a biomarker of beige and brown adipogenesis, remains unclear. To overcome this, Ucp1-Cre was used to drive diphtheria toxin expression in cells expressing UCP1 (Ucp1Cre+/DTA+). Despite loss of brown adipose tissue, BMAT volume was not reduced in Ucp1Cre+/DTA+ mice. Comparably, in mTmG reporter mice (Ucp1Cre+/mTmG+), Ucp1-Cre expression was absent from BMAT in young (3-weeks) and mature (16-weeks) male and female mice. Further, ß3-agonist stimulation failed to induce Ucp1-Cre expression in BMAT. This demonstrates that BMAT adipocytes are not UCP1-expressing beige/brown adipocytes. Thus, to identify novel and emerging roles for BMAT adipocytes in skeletal and whole-body homeostasis, we performed gene enrichment analysis of microarray data from adipose tissues of adult rabbits. Pathway analysis revealed genetic evidence for differences in BMAT including insulin resistance, decreased fatty acid metabolism, and enhanced contributions to local processes including bone mineral density through candidate genes such as osteopontin. In sum, this supports a paradigm by which BMAT adipocytes are a unique subpopulation that is specialized to support cells within the skeletal and hematopoietic niche.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Medula Óssea/metabolismo , Expressão Gênica , Proteína Desacopladora 1/genética , Tecido Adiposo/patologia , Adrenérgicos/farmacologia , Animais , Medula Óssea/patologia , Linhagem da Célula/genética , Feminino , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Esqueleto/metabolismo
3.
PLoS Genet ; 15(6): e1008244, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31233501

RESUMO

Berardinelli-Seip congenital generalized lipodystrophy is associated with increased bone mass suggesting that fat tissue regulates the skeleton. Because there is little mechanistic information regarding this issue, we generated "fat-free" (FF) mice completely lacking visible visceral, subcutaneous and brown fat. Due to robust osteoblastic activity, trabecular and cortical bone volume is markedly enhanced in these animals. FF mice, like Berardinelli-Seip patients, are diabetic but normalization of glucose tolerance and significant reduction in circulating insulin fails to alter their skeletal phenotype. Importantly, the skeletal phenotype of FF mice is completely rescued by transplantation of adipocyte precursors or white or brown fat depots, indicating that adipocyte derived products regulate bone mass. Confirming such is the case, transplantation of fat derived from adiponectin and leptin double knockout mice, unlike that obtained from their WT counterparts, fails to normalize FF bone. These observations suggest a paucity of leptin and adiponectin may contribute to the increased bone mass of Berardinelli-Seip patients.


Assuntos
Adiponectina/genética , Leptina/genética , Lipodistrofia Generalizada Congênita/genética , Osteosclerose/genética , Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Densidade Óssea/genética , Modelos Animais de Doenças , Feminino , Glucose/genética , Glucose/metabolismo , Humanos , Insulina/genética , Gordura Intra-Abdominal/metabolismo , Lipodistrofia Generalizada Congênita/complicações , Lipodistrofia Generalizada Congênita/patologia , Camundongos , Camundongos Knockout , Osteosclerose/etiologia , Osteosclerose/metabolismo , Osteosclerose/patologia , Esqueleto/metabolismo , Esqueleto/patologia , Gordura Subcutânea/metabolismo
4.
Stem Cell Reports ; 13(1): 48-60, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31204302

RESUMO

Mesenchymal stromal cells are key components of hematopoietic niches in the bone marrow. Here we abrogated transforming growth factor ß (TGF-ß) signaling in mesenchymal stem/progenitor cells (MSPCs) by deleting Tgfbr2 in mesenchymal cells using a doxycycline-repressible Sp7 (osterix)-Cre transgene. We show that loss of TGF-ß signaling during fetal development results in a marked expansion of CXCL12-abundant reticular (CAR) cells and adipocytes in the bone marrow, while osteoblasts are significantly reduced. These stromal alterations are associated with significant defects in hematopoiesis, including a shift from lymphopoiesis to myelopoiesis. However, hematopoietic stem cell function is preserved. Interestingly, TGF-ß signaling is dispensable for the maintenance of mesenchymal cells in the bone marrow after birth under steady-state conditions. Collectively, these data show that TGF-ß plays an essential role in the lineage specification of fetal but not definitive MSPCs and is required for the establishment of normal hematopoietic niches in fetal and perinatal bone marrow.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Linhagem Celular , Deleção de Genes , Hematopoese , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/genética
5.
Bone ; 118: 32-41, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29360620

RESUMO

Bone marrow adipose tissue (BMAT) is preserved or increased in states of caloric restriction. Similarly, we found that BMAT in the tail vertebrae, but not the red marrow in the tibia, resists loss of neutral lipid with acute, 48-hour fasting in rats. The mechanisms underlying this phenomenon and its seemingly distinct regulation from peripheral white adipose tissue (WAT) remain unknown. To test the role of ß-adrenergic stimulation, a major regulator of adipose tissue lipolysis, we examined the responses of BMAT to ß-adrenergic agonists. Relative to inguinal WAT, BMAT had reduced phosphorylation of hormone sensitive lipase (HSL) after treatment with pan-ß-adrenergic agonist isoproterenol. Phosphorylation of HSL in response to ß3-adrenergic agonist CL316,243 was decreased by an additional ~90% (distal tibia BMAT) or could not be detected (tail vertebrae). Ex vivo, adrenergic stimulation of lipolysis in purified BMAT adipocytes was also substantially less than iWAT adipocytes and had site-specific properties. Specifically, regulated bone marrow adipocytes (rBMAs) from proximal tibia and femur underwent lipolysis in response to both CL316,243 and forskolin, while constitutive BMAs from the tail responded only to forskolin. This occurred independently of changes in gene expression of ß-adrenergic receptors, which were similar between adipocytes from iWAT and BMAT, and could not be explained by defective coupling of ß-adrenergic receptors to lipolytic machinery through caveolin 1. Specifically, we found that whereas caveolin 1 was necessary to mediate maximal stimulation of lipolysis in iWAT, overexpression of caveolin 1 was insufficient to rescue impaired BMAT signaling. Lastly, we tested the ability of BMAT to respond to 72-hour treatment with CL316,243 in vivo. This was sufficient to cause beiging of iWAT adipocytes and a decrease in iWAT adipocyte cell size. By contrast, adipocyte size in the tail BMAT and distal tibia remained unchanged. However, within the distal femur, we identified a subpopulation of BMAT adipocytes that underwent lipid droplet remodeling. This response was more pronounced in females than in males and resembled lipolysis-induced lipid partitioning rather than traditional beiging. In summary, BMAT has the capacity to respond to ß-adrenergic stimuli, however, its responses are muted and BMAT generally resists lipid hydrolysis and remodeling relative to iWAT. This resistance is more pronounced in distal regions of the skeleton where the BMAT adipocytes are larger with little intervening hematopoiesis, suggesting that there may be a role for both cell-autonomous and microenvironmental determinants. Resistance to ß-adrenergic stimuli further separates BMAT from known regulators of energy partitioning and contributes to our understanding of why BMAT is preserved in states of fasting and caloric restriction.


Assuntos
Adipócitos/citologia , Agonistas Adrenérgicos beta/farmacologia , Células da Medula Óssea/citologia , Lipólise , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Caveolina 1/metabolismo , Tamanho Celular/efeitos dos fármacos , Jejum , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Lipólise/efeitos dos fármacos , Masculino , Camundongos Knockout , Camundongos Transgênicos , Perilipina-1/metabolismo , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Coluna Vertebral/citologia , Esterol Esterase/metabolismo , Cauda , Tíbia/citologia
6.
Bone ; 118: 89-98, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29366839

RESUMO

Unlike white and brown adipose tissues, the bone marrow adipocyte (BMA) exists in a microenvironment containing unique populations of hematopoietic and skeletal cells. To study this microenvironment at the sub-cellular level, we performed a three-dimensional analysis of the ultrastructure of the BMA niche with focused ion beam scanning electron microscopy (FIB-SEM). This revealed that BMAs display hallmarks of metabolically active cells including polarized lipid deposits, a dense mitochondrial network, and areas of endoplasmic reticulum. The distinct orientations of the triacylglycerol droplets suggest that fatty acids are taken up and/or released in three key areas - at the endothelial interface, into the hematopoietic milieu, and at the bone surface. Near the sinusoidal vasculature, endothelial cells send finger-like projections into the surface of the BMA which terminate near regions of lipid within the BMA cytoplasm. In some regions, perivascular cells encase the BMA with their flattened cellular projections, limiting contacts with other cells in the niche. In the hematopoietic milieu, BMAT adipocytes of the proximal tibia interact extensively with maturing cells of the myeloid/granulocyte lineage. Associations with erythroblast islands are also prominent. At the bone surface, the BMA extends organelle and lipid-rich cytoplasmic regions toward areas of active osteoblasts. This suggests that the BMA may serve to partition nutrient utilization between diverse cellular compartments, serving as an energy-rich hub of the stromal-reticular network. Lastly, though immuno-EM, we've identified a subset of bone marrow adipocytes that are innervated by the sympathetic nervous system, providing an additional mechanism for regulation of the BMA. In summary, this work reveals that the bone marrow adipocyte is a dynamic cell with substantial capacity for interactions with the diverse components of its surrounding microenvironment. These local interactions likely contribute to its unique regulation relative to peripheral adipose tissues.


Assuntos
Adipócitos/ultraestrutura , Medula Óssea/ultraestrutura , Imageamento Tridimensional , Microscopia Eletrônica , Nicho de Células-Tronco , Adipócitos/citologia , Animais , Comunicação Celular , Células Endoteliais/citologia , Células Endoteliais/ultraestrutura , Eritrócitos/citologia , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos Endogâmicos C57BL
7.
Matrix Biol ; 71-72: 100-111, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29524629

RESUMO

Microfibril-associated glycoproteins 1 and 2 (MAGP-1, MAGP-2) are protein components of extracellular matrix microfibrils. These proteins interact with fibrillin, the core component of microfibrils, and impart unique biological properties that influence microfibril function in vertebrates. MAGPs bind active forms of TGFß and BMPs and are capable of modulating Notch signaling. Mutations in MAGP-1 or MAGP-2 have been linked to thoracic aneurysms and metabolic disease in humans. MAGP-2 has also been shown to be an important biomarker in several human cancers. Mice lacking MAGP-1 or MAGP-2 have defects in multiple organ systems, which reflects the widespread distribution of microfibrils in vertebrate tissues. This review summarizes our current understanding of the function of the MAGPs and their relationship to human disease.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Biomarcadores/metabolismo , Matriz Extracelular/metabolismo , Humanos , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Camundongos , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Fatores de Processamento de RNA , Receptores Notch/metabolismo , Transdução de Sinais
8.
Calcif Tissue Int ; 100(5): 461-475, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27364342

RESUMO

Adipocytes of the marrow adipose tissue (MAT) are distributed throughout the skeleton, are embedded in extracellular matrix, and are surrounded by cells of the hematopoietic and osteogenic lineages. MAT is a persistent component of the skeletal microenvironment and has the potential to impact local processes including bone accrual and hematopoietic function. In this review, we discuss the initial evolution of MAT in vertebrate lineages while emphasizing comparisons to the development of peripheral adipose, hematopoietic, and skeletal tissues. We then apply these evolutionary clues to define putative functions of MAT. Lastly, we explore the regulation of MAT by two major components of its microenvironment, the extracellular matrix and the nerves embedded within. The extracellular matrix and nerves contribute to both rapid and continuous modification of the MAT niche and may help to explain evolutionary conserved mechanisms underlying the coordinated regulation of blood, bone, and MAT within the skeleton.


Assuntos
Tecido Adiposo , Medula Óssea , Matriz Extracelular , Animais , Humanos
9.
J Clin Invest ; 122(10): 3579-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996695

RESUMO

The adenosine diphosphate (ADP) receptor P2RY12 (purinergic receptor P2Y, G protein coupled, 12) plays a critical role in platelet aggregation, and P2RY12 inhibitors are used clinically to prevent cardiac and cerebral thrombotic events. Extracellular ADP has also been shown to increase osteoclast (OC) activity, but the role of P2RY12 in OC biology is unknown. Here, we examined the role of mouse P2RY12 in OC function. Mice lacking P2ry12 had decreased OC activity and were partially protected from age-associated bone loss. P2ry12-/- OCs exhibited intact differentiation markers, but diminished resorptive function. Extracellular ADP enhanced OC adhesion and resorptive activity of WT, but not P2ry12-/-, OCs. In platelets, ADP stimulation of P2RY12 resulted in GTPase Ras-related protein (RAP1) activation and subsequent αIIbß3 integrin activation. Likewise, we found that ADP stimulation induced RAP1 activation in WT and integrin ß3 gene knockout (Itgb3-/-) OCs, but its effects were substantially blunted in P2ry12-/- OCs. In vivo, P2ry12-/- mice were partially protected from pathologic bone loss associated with serum transfer arthritis, tumor growth in bone, and ovariectomy-induced osteoporosis: all conditions associated with increased extracellular ADP. Finally, mice treated with the clinical inhibitor of P2RY12, clopidogrel, were protected from pathologic osteolysis. These results demonstrate that P2RY12 is the primary ADP receptor in OCs and suggest that P2RY12 inhibition is a potential therapeutic target for pathologic bone loss.


Assuntos
Difosfato de Adenosina/fisiologia , Remodelação Óssea/fisiologia , Osteoclastos/fisiologia , Osteoporose/fisiopatologia , Receptores Purinérgicos P2Y12/fisiologia , Animais , Artrite Experimental/complicações , Neoplasias Ósseas/complicações , Neoplasias Ósseas/secundário , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/fisiopatologia , Carcinoma/complicações , Carcinoma/secundário , Adesão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Clopidogrel , Ativação Enzimática/efeitos dos fármacos , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoporose/etiologia , Osteoporose/prevenção & controle , Ovariectomia , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y12/deficiência , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/genética , Organismos Livres de Patógenos Específicos , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico , Proteínas rap1 de Ligação ao GTP/efeitos dos fármacos
10.
J Cell Biochem ; 113(1): 93-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898536

RESUMO

Microfibril-associated glycoprotein-1 (MAGP1), together with the fibrillins, are constitutive components of vertebrate microfibrils. Mice deficient in MAGP1 (murine MAGP1 knockout animals (Mfap2(-/-)); MAGP1Δ) is appropriate develop progressive osteopenia and reduced whole bone strength, and have elevated numbers of osteoclasts lining the bone surface. Our previous studies suggested that the increased osteoclast population was associated with elevated levels of receptor activator of NF-κB ligand (RANKL), a positive regulator of osteoclast differentiation. To explore the relationship between RANKL expression and osteoclast differentiation in MAGP1 deficiency, oophorectomy (OVX) was used to stimulate RANKL expression in both WT and MAGP1Δ animals. Bone loss following OVX was monitored using whole body DEXA and in vivo µCT. While WT mice exhibited significant bone loss following OVX, percent bone loss was reduced in MAGP1Δ mice. Further, serum RANKL levels rose significantly in OVX WT mice, whereas, there was only a modest increase in RANKL following OVX in the mutant mice due to already high baseline levels. Elevated RANKL expression was normalized when cultured MAGP1Δ osteoblasts were treated with a neutralizing antibody targeting free TGFß. These studies provide support for increased RANKL expression associated with MAGP1 deficiency and provide a link to altered TGF-ß signaling as a possible causative signaling pathway regulating RANKL expression in MAGP1Δ osteoblasts.


Assuntos
Reabsorção Óssea/metabolismo , Proteínas Contráteis/deficiência , Proteínas da Matriz Extracelular/deficiência , Ligante RANK/metabolismo , Animais , Doenças Ósseas Metabólicas/metabolismo , Reabsorção Óssea/genética , Diferenciação Celular , Células Cultivadas , Proteínas Contráteis/genética , Proteínas da Matriz Extracelular/genética , Feminino , Camundongos , Camundongos Knockout , Microfibrilas/fisiologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Ovariectomia , Fatores de Processamento de RNA , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
11.
Clin Exp Metastasis ; 28(1): 39-53, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20981476

RESUMO

Endoglin is a transmembrane receptor that suppresses human prostate cancer (PCa) cell invasion. Small molecule therapeutics now being tested in humans can activate endoglin signaling. It is not known whether endoglin can regulate metastatic behavior, PCa tumor growth, nor what signaling pathways are linked to these processes. This study sought to investigate the effect of endoglin on these parameters. We used a murine orthotopic model of human PCa metastasis, designed by us to measure effects at early steps in the metastatic cascade, and implanted PCa cells stably engineered to express differing levels of endoglin. We now extend this model to measure cancer cells circulating in the blood. Progressive endoglin loss led to progressive increases in the number of circulating PCa cells as well as to the formation of soft tissue metastases. Endoglin was known to suppress invasion by activating the Smad1 transcription factor. We now show that it selectively activates specific Smad1-responsive genes, including JUNB, STAT1, and SOX4. Increased tumor growth and increased Ki67 expression in tissue was seen only with complete endoglin loss. By showing that endoglin increased TGFß-mediated suppression of cell growth in vitro and TGFß-mediated signaling in tumor tissue, loss of this growth-suppressive pathway appears to be implicated at least in part for the increased size of endoglin-deficient tumors. Endoglin is shown for the first time to suppress cell movement out of primary tumor as well as the formation of distant metastasis. It is also shown to co-regulate tumor growth and metastatic behavior in human PCa.


Assuntos
Antígenos CD/metabolismo , Metástase Neoplásica/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Superfície Celular/metabolismo , Animais , Antígenos CD/sangue , Proliferação de Células , Modelos Animais de Doenças , Endoglina , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica/prevenção & controle , Neoplasias da Próstata/sangue , Receptores de Superfície Celular/sangue , Transdução de Sinais , Proteína Smad1/sangue , Proteína Smad1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas
12.
J Biol Chem ; 285(31): 23858-67, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20501659

RESUMO

MAGP1 is an extracellular matrix protein that, in vertebrates, is a ubiquitous component of fibrillin-rich microfibrils. We previously reported that aged MAGP1-deficient mice (MAGP1Delta) develop lesions that are the consequence of spontaneous bone fracture. We now present a more defined bone phenotype found in MAGP1Delta mice. A longitudinal DEXA study demonstrated age-associated osteopenia in MAGP1Delta animals and muCT confirmed reduced bone mineral density in the trabecular and cortical bone. Further, MAGP1Delta mice have significantly less trabecular bone, the trabecular microarchitecture is more fragmented, and the diaphyseal cross-sectional area is significantly reduced. The remodeling defect seen in MAGP1Delta mice is likely not due to an osteoblast defect, because MAGP1Delta bone marrow stromal cells undergo osteoblastogenesis and form mineralized nodules. In vivo, MAGP1Delta mice exhibit normal osteoblast number, mineralized bone surface, and bone formation rate. Instead, our findings suggest increased bone resorption is responsible for the osteopenia. The number of osteoclasts derived from MAGP1Delta bone marrow macrophage cells is increased relative to the wild type, and osteoclast differentiation markers are expressed at earlier time points in MAGP1Delta cells. In vivo, MAGP1Delta mice have more osteoclasts lining the bone surface. RANKL (receptor activator of NF-kappaB ligand) expression is significantly higher in MAGP1Delta bone, and likely contributes to enhanced osteoclastogenesis. However, bone marrow macrophage cells from MAGP1Delta mice show a higher propensity than do wild-type cells to differentiate to osteoclasts in response to RANKL, suggesting that they are also primed to respond to osteoclast-promoting signals. Together, our findings suggest that MAGP1 is a regulator of bone remodeling, and its absence results in osteopenia associated with an increase in osteoclast number.


Assuntos
Remodelação Óssea , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Animais , Células da Medula Óssea/citologia , Fibrilinas , Macrófagos/citologia , Masculino , Camundongos , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/metabolismo , NF-kappa B/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Mapeamento de Interação de Proteínas , Ligante RANK/metabolismo , Fatores de Processamento de RNA , Fator de Crescimento Transformador beta/metabolismo
13.
Carcinogenesis ; 31(3): 359-66, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19736306

RESUMO

Endoglin, a transmembrane glycoprotein that acts as a transforming growth factor-beta (TGF-beta) coreceptor, is downregulated in PC3-M metastatic prostate cancer cells. When restored, endoglin expression in PC3-M cells inhibits cell migration in vitro and attenuates the tumorigenicity of PC3-M cells in SCID mice, though the mechanism of endoglin regulation of migration in prostate cancer cells is not known. The current study indicates that endoglin is phosphorylated on cytosolic domain threonine residues by the TGF-beta type I receptors ALK2 and ALK5 in prostate cancer cells. Importantly, in the presence of constitutively active ALK2, endoglin did not inhibit cell migration, suggesting that endoglin phosphorylation regulated PC3-M cell migration. Therefore, our results suggest that endoglin phosphorylation is a mechanism with relevant functional consequences in prostate cancer cells. These data demonstrate for the first time that TGF-beta receptor-mediated phosphorylation of endoglin is a Smad-independent mechanism involved in the regulation of prostate cancer cell migration.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Adenocarcinoma/patologia , Antígenos CD/fisiologia , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Adenocarcinoma/metabolismo , Animais , Antígenos CD/química , Antígenos CD/genética , Proteína Morfogenética Óssea 7/farmacologia , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/transplante , Movimento Celular/fisiologia , Endoglina , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Fosfotreonina/metabolismo , Neoplasias da Próstata/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Fator de Crescimento Transformador beta1/farmacologia , Transplante Heterólogo
14.
Mol Pharmacol ; 73(1): 235-42, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17951357

RESUMO

Genistein has been shown to inhibit human prostate cancer (PCa) cell motility. Endoglin has been identified as an important suppressor of PCa cell motility, and its expression is lost during PCa progression. It is therefore important to determine whether endoglin loss affects genistein's efficacy and, if so, by what mechanism. In the current study, genistein was shown to induce reversion of endoglin-deficient cells to a low motility, endoglin-replete phenotype. Because endoglin suppresses PCa cell motility in an activin-like kinase receptor-2 (ALK2)- and Smad1-dependent manner, we sought to determine whether genistein was activating the ALK2-Smad1 pathway. Although treatment with genistein or overexpression of Smad1 or ALK2 all increased Smad1-responsive promoter activity and decreased cell motility, genistein's efficacy was abrogated by either Smad1 or ALK2 knockdown. Furthermore, transfection of cells with a kinase dead mutant of ALK2 abrogated genistein's efficacy. Together, these findings demonstrate that genistein therapeutically induces reversion to a low-motility phenotype in aggressive endoglin-deficient PCa cells. It does so by activating ALK2-Smad1 endoglin-associated signaling. These findings support the notion that individuals with low endoglin-expressing PCa will benefit from genistein treatment.


Assuntos
Antígenos CD/metabolismo , Genisteína/farmacologia , Neoplasias da Próstata/metabolismo , Receptores de Superfície Celular/metabolismo , Linhagem Celular Tumoral , Endoglina , Humanos , Masculino , Fenótipo , Neoplasias da Próstata/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA