Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 37(8): 110058, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818538

RESUMO

Mouse hematopoietic tissues contain abundant tissue-resident macrophages that support immunity, hematopoiesis, and bone homeostasis. A systematic strategy to characterize macrophage subsets in mouse bone marrow (BM), spleen, and lymph node unexpectedly reveals that macrophage surface marker staining emanates from membrane-bound subcellular remnants associated with unrelated cells. Intact macrophages are not present within these cell preparations. The macrophage remnant binding profile reflects interactions between macrophages and other cell types in vivo. Depletion of CD169+ macrophages in vivo eliminates F4/80+ remnant attachment. Remnant-restricted macrophage-specific membrane markers, cytoplasmic fluorescent reporters, and mRNA are all detected in non-macrophage cells including isolated stem and progenitor cells. Analysis of RNA sequencing (RNA-seq) data, including publicly available datasets, indicates that macrophage fragmentation is a general phenomenon that confounds bulk and single-cell analysis of disaggregated hematopoietic tissues. Hematopoietic tissue macrophage fragmentation undermines the accuracy of macrophage ex vivo molecular profiling and creates opportunity for misattribution of macrophage-expressed genes to non-macrophage cells.


Assuntos
Separação Celular/métodos , Macrófagos/citologia , Análise de Célula Única/métodos , Animais , Medula Óssea/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Homeostase , Camundongos
2.
Biomaterials ; 275: 120936, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34303178

RESUMO

Macrophage-targeted therapies, including macrophage colony-stimulating factor 1 (CSF1), have been shown to have pro-repair impacts post-fracture. Preclinical/clinical applications of CSF1 have been expedited by development of chimeric CSF1-Fc which has extended circulating half-life. Here, we used mouse models to investigate the bone regenerative potential of CSF1-Fc in healthy and osteoporotic fracture. We also explored whether combination of CSF1-Fc with interleukin (IL)-4 provided additional fracture healing benefit in osteopenic bone. Micro-computed tomography, in situ histomorphometry, and bone mechanical parameters were used to assess systemic impacts of CSF1-Fc therapy in naive mice (male and female young, adult and geriatric). An intermittent CSF1-Fc regimen was optimized to mitigate undesirable impacts on bone resorption and hepatosplenomegaly, irrespective of age or gender. The intermittent CSF1-Fc regimen was tested in a mid-diaphyseal femoral fracture model in healthy bones with treatment initiated 1-day post-fracture. Weekly CSF1-Fc did not impact osteoclasts but increased osteal macrophages and improved fracture strength. Importantly, this treatment regimen also improved fracture union and strength in an ovariectomy-model of delayed fracture repair. Combining CSF1-Fc with IL-4 initiated 1-week post-fracture reduced the efficacy of CSF1-Fc. This study describes a novel strategy to specifically achieve bone regenerative actions of CSF1-Fc that has the potential to alleviate fragility fracture morbidity and mortality.


Assuntos
Consolidação da Fratura , Fator Estimulador de Colônias de Macrófagos , Animais , Osso e Ossos , Feminino , Macrófagos , Masculino , Camundongos , Microtomografia por Raio-X
3.
J Bone Miner Res ; 36(11): 2214-2228, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34278602

RESUMO

Osteal macrophages (osteomacs) support osteoblast function and promote bone anabolism, but their contribution to osteoporosis has not been explored. Although mouse ovariectomy (OVX) models have been repeatedly used, variation in strain, experimental design and assessment modalities have contributed to no single model being confirmed as comprehensively replicating the full gamut of osteoporosis pathological manifestations. We validated an OVX model in adult C3H/HeJ mice and demonstrated that it presents with human postmenopausal osteoporosis features with reduced bone volume in axial and appendicular bone and bone loss in both trabecular and cortical bone including increased cortical porosity. Bone loss was associated with increased osteoclasts on trabecular and endocortical bone and decreased osteoblasts on trabecular bone. Importantly, this OVX model was characterized by delayed fracture healing. Using this validated model, we demonstrated that osteomacs are increased post-OVX on both trabecular and endocortical bone. Dual F4/80 (pan-macrophage marker) and tartrate-resistant acid phosphatase (TRAP) staining revealed osteomacs frequently located near TRAP+ osteoclasts and contained TRAP+ intracellular vesicles. Using an in vivo inducible macrophage depletion model that does not simultaneously deplete osteoclasts, we observed that osteomac loss was associated with elevated extracellular TRAP in bone marrow interstitium and increased serum TRAP. Using in vitro high-resolution confocal imaging of mixed osteoclast-macrophage cultures on bone substrate, we observed macrophages juxtaposed to osteoclast basolateral functional secretory domains scavenging degraded bone byproducts. These data demonstrate a role for osteomacs in supporting osteoclastic bone resorption through phagocytosis and sequestration of resorption byproducts. Overall, our data expose a novel role for osteomacs in supporting osteoclast function and provide the first evidence of their involvement in osteoporosis pathogenesis. © 2021 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Reabsorção Óssea , Osteoporose Pós-Menopausa , Animais , Osso e Ossos , Diferenciação Celular , Feminino , Humanos , Macrófagos , Camundongos , Camundongos Endogâmicos C3H , Osteoblastos , Osteoclastos , Ovariectomia
4.
J Hematol Oncol ; 14(1): 3, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33402221

RESUMO

BACKGROUND: Prior chemotherapy and/or underlying morbidity commonly leads to poor mobilisation of hematopoietic stem cells (HSC) for transplantation in cancer patients. Increasing the number of available HSC prior to mobilisation is a potential strategy to overcome this deficiency. Resident bone marrow (BM) macrophages are essential for maintenance of niches that support HSC and enable engraftment in transplant recipients. Here we examined potential of donor treatment with modified recombinant colony-stimulating factor 1 (CSF1) to influence the HSC niche and expand the HSC pool for autologous transplantation. METHODS: We administered an acute treatment regimen of CSF1 Fc fusion protein (CSF1-Fc, daily injection for 4 consecutive days) to naive C57Bl/6 mice. Treatment impacts on macrophage and HSC number, HSC function and overall hematopoiesis were assessed at both the predicted peak drug action and during post-treatment recovery. A serial treatment strategy using CSF1-Fc followed by granulocyte colony-stimulating factor (G-CSF) was used to interrogate HSC mobilisation impacts. Outcomes were assessed by in situ imaging and ex vivo standard and imaging flow cytometry with functional validation by colony formation and competitive transplantation assay. RESULTS: CSF1-Fc treatment caused a transient expansion of monocyte-macrophage cells within BM and spleen at the expense of BM B lymphopoiesis and hematopoietic stem and progenitor cell (HSPC) homeostasis. During the recovery phase after cessation of CSF1-Fc treatment, normalisation of hematopoiesis was accompanied by an increase in the total available HSPC pool. Multiple approaches confirmed that CD48-CD150+ HSC do not express the CSF1 receptor, ruling out direct action of CSF1-Fc on these cells. In the spleen, increased HSC was associated with expression of the BM HSC niche macrophage marker CD169 in red pulp macrophages, suggesting elevated spleen engraftment with CD48-CD150+ HSC was secondary to CSF1-Fc macrophage impacts. Competitive transplant assays demonstrated that pre-treatment of donors with CSF1-Fc increased the number and reconstitution potential of HSPC in blood following a HSC mobilising regimen of G-CSF treatment. CONCLUSION: These results indicate that CSF1-Fc conditioning could represent a therapeutic strategy to overcome poor HSC mobilisation and subsequently improve HSC transplantation outcomes.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Animais , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hematopoese/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/farmacologia
5.
Blood ; 132(7): 735-749, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29945953

RESUMO

Distinct subsets of resident tissue macrophages are important in hematopoietic stem cell niche homeostasis and erythropoiesis. We used a myeloid reporter gene (Csf1r-eGFP) to dissect the persistence of bone marrow and splenic macrophage subsets following lethal irradiation and autologous hematopoietic stem cell transplantation in a mouse model. Multiple recipient bone marrow and splenic macrophage subsets survived after autologous hematopoietic stem cell transplantation with organ-specific persistence kinetics. Short-term persistence (5 weeks) of recipient resident macrophages in spleen paralleled the duration of extramedullary hematopoiesis. In bone marrow, radiation-resistant recipient CD169+ resident macrophages and erythroid-island macrophages self-repopulated long-term after transplantation via autonomous cell division. Posttransplant peak expansion of recipient CD169+ resident macrophage number in bone marrow aligned with the persistent engraftment of phenotypic long-term reconstituting hematopoietic stem cells within bone marrow. Selective depletion of recipient CD169+ macrophages significantly compromised the engraftment of phenotypic long-term reconstituting hematopoietic stem cells and consequently impaired hematopoietic reconstitution. Recipient bone marrow resident macrophages are essential for optimal hematopoietic stem cell transplantation outcomes and could be an important consideration in the development of pretransplant conditioning therapies and/or chemoresistance approaches.


Assuntos
Medula Óssea/metabolismo , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Macrófagos/metabolismo , Lesões Experimentais por Radiação/metabolismo , Animais , Autoenxertos , Medula Óssea/patologia , Sobrevivência Celular , Células-Tronco Hematopoéticas/patologia , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/terapia
6.
Stem Cell Reports ; 5(5): 682-689, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26527386

RESUMO

In this article, Millard and colleagues show that intrauterine bone marrow transplantation in the oim/oim mouse model of osteogenesis imperfecta yields hematopoietic microchimerism in the absence of donor osteopoiesis or phenotypic improvement. Bone-associated donor cells were not bone-forming osteoblasts, but osteoclasts (bone resorbing cells of the hematopoietic lineage) and osteal macrophages (bone regulatory cells of the hematopoietic lineage).


Assuntos
Células da Medula Óssea/citologia , Transplante de Medula Óssea , Transplante de Células-Tronco Hematopoéticas , Osteoblastos/citologia , Osteoclastos/citologia , Osteogênese Imperfeita/cirurgia , Animais , Camundongos
7.
Am J Pathol ; 184(12): 3192-204, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25285719

RESUMO

The distribution, phenotype, and requirement of macrophages for fracture-associated inflammation and/or early anabolic progression during endochondral callus formation were investigated. A murine femoral fracture model [internally fixed using a flexible plate (MouseFix)] was used to facilitate reproducible fracture reduction. IHC demonstrated that inflammatory macrophages (F4/80(+)Mac-2(+)) were localized with initiating chondrification centers and persisted within granulation tissue at the expanding soft callus front. They were also associated with key events during soft-to-hard callus transition. Resident macrophages (F4/80(+)Mac-2(neg)), including osteal macrophages, predominated in the maturing hard callus. Macrophage Fas-induced apoptosis transgenic mice were used to induce macrophage depletion in vivo in the femoral fracture model. Callus formation was completely abolished when macrophage depletion was initiated at the time of surgery and was significantly reduced when depletion was delayed to coincide with initiation of early anabolic phase. Treatment initiating 5 days after fracture with the pro-macrophage cytokine colony stimulating factor-1 significantly enhanced soft callus formation. The data support that inflammatory macrophages were required for initiation of fracture repair, whereas both inflammatory and resident macrophages promoted anabolic mechanisms during endochondral callus formation. Overall, macrophages make substantive and prolonged contributions to fracture healing and can be targeted as a therapeutic approach for enhancing repair mechanisms. Thus, macrophages represent a viable target for the development of pro-anabolic fracture treatments with a potentially broad therapeutic window.


Assuntos
Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura , Macrófagos/metabolismo , Osteogênese/fisiologia , Periósteo/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Citocinas/metabolismo , Progressão da Doença , Citometria de Fluxo , Fixação de Fratura , Imuno-Histoquímica , Inflamação , Fixadores Internos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/citologia , Fenótipo
8.
Exp Hematol ; 42(7): 547-61.e4, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24721610

RESUMO

Similarly to other tissues, the bone marrow contains subsets of resident tissue macrophages, which are essential to maintain bone formation, functional hematopoietic stem cell (HSC) niches, and erythropoiesis. Pharmacologic doses of granulocyte colony-stimulating factor (G-CSF) mobilize HSC in part by interfering with the HSC niche-supportive function of BM resident macrophages. Because bone marrow macrophages are key to both maintenance of HSC within their niche and erythropoiesis, we investigated the effect of mobilizing doses of G-CSF on erythropoiesis in mice. We now report that G-CSF blocks medullar erythropoiesis by depleting the erythroid island macrophages we identified as co-expressing F4/80, vascular cell adhesion molecule-1, CD169, Ly-6G, and the ER-HR3 erythroid island macrophage antigen. Both broad macrophage depletion, achieved by injecting clodronate-loaded liposomes, and selective depletion of CD169(+) macrophages, also concomitantly depleted F4/80(+)VCAM-1(+)CD169(+)ER-HR3(+)Ly-6G(+) erythroid island macrophages and blocked erythropoiesis. This more precise phenotypic definition of erythroid island macrophages will enable studies on their biology and function in normal settings and on diseases associated with anemia. Finally, this study further illustrates that macrophages are a potent relay of innate immunity and inflammation on bone, hematopoietic, and erythropoietic maintenance. Agents that affect these macrophages, such as G-CSF, are likely to affect these three processes concomitantly.


Assuntos
Eritropoese/fisiologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Macrófagos/fisiologia , Animais , Camundongos , Molécula 1 de Adesão de Célula Vascular/fisiologia
9.
Am J Pathol ; 182(5): 1501-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23499466

RESUMO

Previous studies have generated conflicting results regarding the contribution of B cells to bone formation during physiology and repair. Here, we have investigated the role of B cells in osteoblast-mediated intramembranous anabolic bone modeling. Immunohistochemistry for CD45 receptor expression indicated that B cells had no propensity or aversion for endosteal regions or sites of bone modeling and/or remodeling in wild-type mice. In the endocortical diaphyseal region, quantitative immunohistology demonstrated that young wild-type and B-cell deficient mice had similar amounts of osteocalcin(+) osteoblast bone modeling surface. The degree of osteoblast-associated osteomac canopy was also comparable in these mice inferring that bone modeling cellular units were preserved in the absence of B cells. In a tibial injury model, only rare CD45 receptor positive B cells were located within areas of high anabolic activity, including minimal association with osterix(+) osteoblast-lineage committed mesenchymal cells in wild-type mice. Quantitative immunohistology demonstrated that collagen type I matrix deposition and macrophage and osteoclast distribution within the injury site were not compromised by the absence of B cells. Overall, osteoblast distribution during normal growth and bone healing via intramembranous ossification proceeded normally in the absence of B cells. These observations support that in vivo, these lymphoid cells have minimal influence, or at most, make redundant contributions to osteoblast function during anabolic bone modeling via intramembranous mechanisms.


Assuntos
Linfócitos B/patologia , Depleção Linfocítica , Osteogênese , Tíbia/lesões , Tíbia/patologia , Cicatrização , Animais , Medula Óssea/patologia , Remodelação Óssea , Microambiente Celular , Diáfises/patologia , Modelos Animais de Doenças , Antígenos Comuns de Leucócito/metabolismo , Membranas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ossificação Heterotópica/patologia , Ossificação Heterotópica/fisiopatologia , Tíbia/fisiopatologia
10.
Bonekey Rep ; 2: 373, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-25035807

RESUMO

Macrophages have reemerged to prominence with widened understanding of their pleiotropic contributions to many biologies and pathologies. This includes clear advances in revealing their importance in wound healing. Here we have focused on the current state of knowledge with respect to bone repair, which has received relatively little scientific attention compared with its soft-tissue counterparts. Our detailed characterization of resident tissue macrophages residing in bone-lining tissues (osteomacs), including their pro-anabolic function, exposed a more prominent role for these cells in bone biology than previously anticipated. Recent studies have confirmed the importance of macrophages in early inflammatory processes that establish the healing cascade after bone fracture. Emerging data support that macrophage influence extends into both anabolic and catabolic phases of repair, suggesting that these cells have prolonged and diverse functions during fracture healing. More research is needed to clarify macrophage phase-specific contributions, temporospatial subpopulation variance and macrophage specific-molecular mediators. There is also clear motivation for determining whether macrophage alterations underlie compromised fracture healing. Overall, there is strong justification to pursue strategies targeting macrophages and/or their products for improving normal bone healing and overcoming failed repair.

11.
J Bone Miner Res ; 26(7): 1517-32, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21305607

RESUMO

Bone-lining tissues contain a population of resident macrophages termed osteomacs that interact with osteoblasts in vivo and control mineralization in vitro. The role of osteomacs in bone repair was investigated using a mouse tibial bone injury model that heals primarily through intramembranous ossification and progresses through all major phases of stabilized fracture repair. Immunohistochemical studies revealed that at least two macrophage populations, F4/80(+) Mac-2(-/low) TRACP(-) osteomacs and F4/80(+) Mac-2(hi) TRACP(-) inflammatory macrophages, were present within the bone injury site and persisted throughout the healing time course. In vivo depletion of osteomacs/macrophages (either using the Mafia transgenic mouse model or clodronate liposome delivery) or osteoclasts (recombinant osteoprotegerin treatment) established that osteomacs were required for deposition of collagen type 1(+) (CT1(+)) matrix and bone mineralization in the tibial injury model, as assessed by quantitative immunohistology and micro-computed tomography. Conversely, administration of the macrophage growth factor colony-stimulating factor 1 (CSF-1) increased the number of osteomacs/macrophages at the injury site significantly with a concurrent increase in new CT1(+) matrix deposition and enhanced mineralization. This study establishes osteomacs as participants in intramembranous bone healing and as targets for primary anabolic bone therapies.


Assuntos
Macrófagos/metabolismo , Tíbia/lesões , Tíbia/patologia , Cicatrização , Fosfatase Ácida/metabolismo , Animais , Matriz Óssea/efeitos dos fármacos , Matriz Óssea/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Modelos Animais de Doenças , Inflamação/patologia , Isoenzimas/metabolismo , Lipossomos/administração & dosagem , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Membranas/efeitos dos fármacos , Membranas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteogênese/efeitos dos fármacos , Osteoprotegerina/farmacologia , Propriedades de Superfície/efeitos dos fármacos , Fosfatase Ácida Resistente a Tartarato , Tíbia/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
12.
Blood ; 116(23): 4815-28, 2010 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-20713966

RESUMO

In the bone marrow, hematopoietic stem cells (HSCs) reside in specific niches near osteoblast-lineage cells at the endosteum. To investigate the regulation of these endosteal niches, we studied the mobilization of HSCs into the bloodstream in response to granulocyte colony-stimulating factor (G-CSF). We report that G-CSF mobilization rapidly depletes endosteal osteoblasts, leading to suppressed endosteal bone formation and decreased expression of factors required for HSC retention and self-renewal. Importantly, G-CSF administration also depleted a population of trophic endosteal macrophages (osteomacs) that support osteoblast function. Osteomac loss, osteoblast suppression, and HSC mobilization occurred concomitantly, suggesting that osteomac loss could disrupt endosteal niches. Indeed, in vivo depletion of macrophages, in either macrophage Fas-induced apoptosis (Mafia) transgenic mice or by administration of clodronate-loaded liposomes to wild-type mice, recapitulated the: (1) loss of endosteal osteoblasts and (2) marked reduction of HSC-trophic cytokines at the endosteum, with (3) HSC mobilization into the blood, as observed during G-CSF administration. Together, these results establish that bone marrow macrophages are pivotal to maintain the endosteal HSC niche and that the loss of such macrophages leads to the egress of HSCs into the blood.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Nicho de Células-Tronco/metabolismo , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular , Linhagem da Célula , Movimento Celular/fisiologia , Separação Celular , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Biol Chem ; 285(33): 25103-8, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20501658

RESUMO

Physiological bone remodeling is a highly coordinated process responsible for bone resorption and formation and is necessary to repair damaged bone and to maintain mineral homeostasis. In addition to the traditional bone cells (osteoclasts, osteoblasts, and osteocytes) that are necessary for bone remodeling, several immune cells have also been implicated in bone disease. This minireview discusses physiological bone remodeling, outlining the traditional bone biology dogma in light of emerging osteoimmunology data. Specifically discussed in detail are the cellular and molecular mechanisms of bone remodeling, including events that orchestrate the five sequential phases of bone remodeling: activation, resorption, reversal, formation, and termination.


Assuntos
Remodelação Óssea/fisiologia , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/fisiopatologia , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Osteogênese/fisiologia
14.
Genomics ; 94(4): 233-40, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19573590

RESUMO

Mutations in the FBN1 gene, encoding the extracellular matrix protein fibrillin-1, result in the dominant connective tissue disease Marfan syndrome. Marfan syndrome has a variable phenotype, even within families carrying the same FBN1 mutation. Differences in gene expression resulting from sequence differences in the promoter region of the FBN1 gene are likely to be involved in causing this phenotypic variability. In this report, we present an analysis of FBN1 transcription start site (TSS) use in mouse and human tissues. We found that transcription of FBN1 initiated primarily from a single CpG-rich promoter which was highly conserved in mammals. It contained potential binding sites for a number of factors implicated in mesenchyme differentiation and gene expression. The human osteosarcoma line MG63 had high levels of FBN1 mRNA and secreted fibrillin-1 protein to form extracellular matrix fibres. The human embryonic kidney line HEK293 and two breast cancer lines MCF7 and MDA-MB-231 had levels of FBN1 mRNA 1000 fold lower and produced negligible amounts of fibrillin-1 protein. Therefore MG63 appears to be the optimal cell line for examining tissue-specific, biologically relevant promoter activity for FBN1. In reporter assays, the conserved promoter region was more active in MG63 cells than in non-FBN1-expressing lines but additional elements outside the proximal promoter are probably required for optimal tissue-specific expression. Understanding the regulation of the FBN1 gene may lead to alternative therapeutic strategies for Marfan syndrome.


Assuntos
Biologia Computacional/métodos , Síndrome de Marfan/genética , Proteínas dos Microfilamentos/genética , Regiões Promotoras Genéticas , Animais , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Ilhas de CpG , Éxons , Fibrilina-1 , Fibrilinas , Imunofluorescência , Genes Dominantes , Genes Reporter , Humanos , Imuno-Histoquímica , Rim/citologia , Luciferases/metabolismo , Camundongos , Dados de Sequência Molecular , Osteoblastos/metabolismo , Ligação Proteica , RNA Mensageiro/metabolismo , Análise de Sequência de Proteína , Sítio de Iniciação de Transcrição
15.
Blood ; 113(22): 5644-9, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19336758

RESUMO

We have quantified the relative contribution of donor antigen-presenting cell populations to alloantigen presentation after bone marrow transplantation (BMT) by using transgenic T cells that can respond to host-derived alloantigen presented within the donor major histocompatibility complex. We also used additional transgenic/knockout donor mice and/or monoclonal antibodies that allowed conditional depletion of conventional dendritic cells (cDCs), plasmacytoid DC (pDCs), macrophages, or B cells. Using these systems, we demonstrate that donor cDCs are the critical population presenting alloantigen after BMT, whereas pDCs and macrophages do not make a significant contribution in isolation. In addition, alloantigen presentation was significantly enhanced in the absence of donor B cells, confirming a regulatory role for these cells early after transplantation. These data have major implications for the design of therapeutic strategies post-BMT, and suggest that cDC depletion and the promotion of B-cell reconstitution may be beneficial tools for the control of alloreactivity.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Transplante de Medula Óssea/imunologia , Células Dendríticas/imunologia , Isoantígenos/imunologia , Experimentação Animal , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Antígenos CD11/genética , Células Dendríticas/fisiologia , Feminino , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
J Biol Chem ; 283(11): 6790-8, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18165223

RESUMO

Interleukin-18 (IL-18) can regulate osteoblast and osteoclast function. We have identified, using cDNA microarray technology, that IL-18 expression is increased in UMR 106-01 rat osteoblastic cells in response to parathyroid hormone (PTH) treatment. Confirmation of these data using real-time reverse transcription-PCR showed that steady-state levels of IL-18 mRNA increased by 2 h (3-fold), peaked by 4 h (10-fold), and had diminished after 12 h (4.4-fold) and that this regulation was via the protein kinase A signaling pathway and did not involve activation of the PKC signal cascade. PTH regulation of IL-18 was confirmed at the protein level, and analysis of differentiating primary rat calvarial osteoblasts verified that both IL-18 mRNA and protein are regulated by PTH in primary rat osteoblasts. Promoter reporter assays revealed that PTH regulated the upstream IL-18 promoter and induced the exon 1 containing 1.1-kb IL-18 mRNA transcript in primary osteoblast cells. The in vivo physiological role of IL-18 in the anabolic actions of PTH on bone was then assessed using IL-18 knock-out mice. Female IL-18 null mice and wild-type littermate controls were injected with vehicle or 8 microg/100 g of human 1-38 PTH for 4 weeks. In IL-18 knock-out animals the anabolic effect of PTH (determined by bone mineral density changes in the proximal tibia) was abolished in trabecular bone but not in the cortical component. These data characterize the PTH regulation of IL-18 expression in osteoblastic cells and suggest that this cytokine is involved in the anabolic actions of PTH.


Assuntos
Osso e Ossos/metabolismo , Regulação da Expressão Gênica , Interleucina-18/fisiologia , Hormônio Paratireóideo/metabolismo , Animais , Feminino , Humanos , Interleucina-18/genética , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Modelos Genéticos , Osteoblastos/metabolismo , Regiões Promotoras Genéticas , Ratos
17.
J Biomater Sci Polym Ed ; 18(9): 1101-23, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17931502

RESUMO

Osteoblast proliferation is sensitive to material surface properties. In this study, the proliferation of MC3T3 E1-S14 osteoblastic cells on poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) films with different surface characteristics was investigated with the aim of evaluating the cause of a lag in cell growth previously observed. The solvent-cast films were prepared using three different solvents/solvent mixtures which produced PHBV films with both a rough (at the air interface) and smooth (at the glass interface) surface. Investigation of the surface roughness by scanning electron and scanning probe microscopy revealed that the surfaces had features that were different in both average lateral size and average amplitude (Ra 20-200 nm). Water contact angles showed that all surfaces were hydrophobic in nature (thetaA in the range 69-82 degrees ). The lateral distribution of surface crystallinity of the films was evaluated by use of micro-attenuated total reflectance Fourier transform infrared (ATR-FT-IR) by determining the surface crystallinity index (CI) which was found to differ between samples. MC3T3-E1-S14 osteoblasts were cultured on the six surfaces and proliferation was determined. After 2 days, cell proliferation on all surfaces was significantly less than on the control substrate; however, after 4 days cell proliferation was optimal on three surfaces. It was concluded that the initial lag on all substrates was due to the hydrophobic nature of the substrates. The ability of the cells to recover on the materials was attributed to the degree of heterogeneity of the crystallinity and surface roughness: samples with a roughness of 80 nm were found to support cell proliferation. In addition, the lateral surface features influenced the proliferation of osteoblasts on the PHBV film surface.


Assuntos
Osteoblastos/citologia , Poliésteres/química , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Forma Celular , Interações Hidrofóbicas e Hidrofílicas , Lipopolissacarídeos/farmacologia , Camundongos , Microscopia Eletrônica de Varredura , Osteoblastos/efeitos dos fármacos , Solventes , Espectroscopia de Infravermelho com Transformada de Fourier , Propriedades de Superfície , Difração de Raios X
18.
Trends Endocrinol Metab ; 15(2): 60-5, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15036251

RESUMO

Parathyroid hormone (PTH) is the major hormone regulating calcium metabolism. It is also the only FDA-approved drug for osteoporosis treatment that stimulates bone formation when injected daily. However, continuous infusion of PTH causes severe bone loss in line with its known catabolic effects. Many studies to understand the dual effects of PTH have been carried out, and in recent years a growing number of molecular and cellular mechanisms underlying these effects have emerged. Here, we outline the present knowledge and conclude that the kinetics of administration and subsequent signaling probably account for the divergent actions of the hormone.


Assuntos
Remodelação Óssea/fisiologia , Osso e Ossos/metabolismo , Osteoporose/metabolismo , Hormônio Paratireóideo/farmacologia , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/fisiologia , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/fisiopatologia , Cálcio/metabolismo , Feminino , Humanos , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/administração & dosagem
19.
Drugs ; 62(15): 2185-91, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12381218

RESUMO

The benefit of HMG-CoA reductase inhibitors (statins) to the cardiovascular system is now well established and these drugs are being used extensively to treat hypercholesterolaemia clinically. However, as clinical outcomes become available it appears that statins are proving more beneficial than expected and thus it is being proposed that the actions of statins go beyond their ability to lower serum cholesterol levels. The report that statins can interact directly with lymphocyte function-associated antigen (LFA)-1 and prevent it engaging with the intracellular adhesion molecule (ICAM)-1 receptor on T cells is a novel mechanism of statin action and provides convincing evidence that these compounds can regulate biological systems other than by the cholesterol synthesis pathway. Immunosuppression to prevent organ transplant rejection is one application for which statins are currently being assessed. The clinical evidence is conflicting and does not convincingly reflect whether statins are beneficial as immunomodulators. However, in vivo studies investigating the cellular actions of statins have identified two mechanisms by which statins can potentially modulate an in vivo immune response. Firstly, statins regulate inducible class II major histocompatibility complex (MHC) expression on macrophages and endothelial cells. Secondly, statins can inhibit LFA-1 adhesion to ICAM-1 and thus regulate T cell activation. These findings suggest that statins have the potential to regulate an immune response in vivo and that more investigation is essential in order to explain the opposing clinical data.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...