Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Calcif Tissue Int ; 94(1): 78-87, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24101233

RESUMEN

Throughout life, a balance exists within the marrow cavity between adipose tissue and bone. Each tissue derives from a common progenitor cell known both as a "bone marrow-derived multipotent stromal cell" and as a "mesenchymal stem cell" (BMSC). The majority of in vitro and in vivo data suggest that BMSCs differentiate into adipocytes or osteoblasts in a reciprocal manner. For example, while ligand induction of the transcription factors peroxisome proliferator-activated receptor γ initiates BMSC adipogenesis, it suppresses osteogenesis. Nevertheless, this hypothesis may oversimplify a complex regulatory paradigm. The picture may be further complicated by the systemic impact of extramedullary adipose depots on bone via the secretion of protein adipokines and lipid metabolites. This review focuses on past and current literature examining the mechanisms governing the adipose-bone interface.


Asunto(s)
Adipocitos/metabolismo , Remodelación Ósea/fisiología , Adipocitos/citología , Animales , Humanos , Células Madre Mesenquimatosas/citología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Factores de Transcripción/metabolismo
2.
Transfus Med Hemother ; 35(3): 228-238, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21547120

RESUMEN

SUMMARY: Mesenchymal stem cells (MSCs) represent a class of multipotent progenitor cells that have been isolated from multiple tissue sites. Of these, adipose tissue and bone marrow offer advantages in terms of access, abundance, and the extent of their documentation in the literature. This review focuses on the in vitro differentiation capability of cells derived from adult human tissue. Multiple, independent studies have demonstrated that MSCs can commit to mesodermal (adipocyte, chondrocyte, hematopoietic support, myocyte, osteoblast, tenocyte), ectodermal (epithelial, glial, neural), and endodermal (hepatocyte, islet cell) lineages. The limitations and promises of these studies in the context of tissue engineering are discussed.

3.
Bone ; 39(6): 1361-72, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16904389

RESUMEN

Whereas continuous PTH infusion increases bone resorption and bone loss, intermittent PTH treatment stimulates bone formation, in part, via reactivation of quiescent bone surfaces and reducing osteoblast apoptosis. We investigated the possibility that intermittent and continuous PTH treatment also differentially regulates osteogenic and adipocytic lineage commitment of bone marrow stromal progenitor/mesenchymal stem cells (MSC). The MSC were cultured under mildly adipogenic conditions in medium supplemented with dexamethasone, insulin, isobutyl-methylxanthine and troglitazone (DIIT), and treated with 50 nM human PTH(1-34) for either 1 h/day or continuously (PTH replenished every 48 h). After 6 days, cells treated with PTH for 1 h/day retained their normal fibroblastic appearance whereas those treated continuously adopted a polygonal, irregular morphology. After 12-18 days numerous lipid vacuole and oil red O-positive adipocytes had developed in cultures treated with DIIT alone, or with DIIT and continuous PTH. In contrast, adipocyte number was reduced and alkaline phosphatase staining increased in the cultures treated with DIIT and 1 h/day PTH, indicating suppression of adipogenesis and possible promotion of early osteoblastic differentiation. Furthermore, intermittent but not continuous PTH treatment suppressed markers of differentiated adipocytes such as mRNA expression of lipoprotein lipase and PPARgamma as well as glycerol 3-phosphate dehydrogenase activity. All of these effects of intermittent PTH were also produced by a 1 h/day treatment with AH3960 (30 microM), a small molecule, non-peptide agonist of the PTH1 receptor. AH3960, like PTH, activates both the cAMP and calcium signaling pathways. Treatment with the adenylyl cyclase activator forskolin for 1 h/day, mimicked the anti-adipogenic effect of intermittent PTH, whereas pretreatment with the protein kinase-A inhibitor H89 prior to intermittent PTH resulted in almost complete conversion to adipocytes. In contrast, the MAP kinase inhibitor PD 98059 failed to prevent the anti-adipocytic effect of intermittent PTH, suggesting that the inhibitory effect of PTH on adipocyte differentiation is predominantly cAMP-dependent. These results demonstrate a differential effect of PTH1 receptor agonists on the adipocytic commitment and differentiation of adult human bone marrow mesenchymal stem cells. This response may represent an additional mechanism that contributes to the overall bone anabolic action of intermittent PTH.


Asunto(s)
Adipocitos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Teriparatido/farmacología , Adenilil Ciclasas/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Fosfatasa Alcalina/metabolismo , Barbitúricos/farmacología , Secuencia de Bases , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , AMP Cíclico/metabolismo , Cartilla de ADN/genética , Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Glicerolfosfato Deshidrogenasa/metabolismo , Humanos , Metabolismo de los Lípidos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/genética , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
4.
Bone ; 37(2): 159-69, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15921971

RESUMEN

In this paper, we propose a mathematical model for parathyroid hormone receptor (PTH1R) kinetics, focusing on the receptor's response to PTH dosing to discern bone formation responses from bone resorption. The PTH1R is a major target for new osteoporosis treatments, as pulsatile PTH dosing has been shown to induce net bone formation in both animals and humans, and PTH(1-34) was recently FDA approved for the treatment of post-menopausal osteoporosis. PTH has also been shown to cause net bone loss when given continuously, so that the net action of PTH on bone is dependent on the dosing pattern. We have developed a simplified two-state receptor kinetics model for the PTH1R, based on the concepts of Segel et al., to distinguish the activity of active and inactive receptor and receptor-ligand complexes. The goal is to develop a plausible model of the minimal essential biological relationships necessary for understanding the responses to PTH dosing. A two-state model is able to effectively discriminate between continuous and pulsatile PTH dosing using the active species as surrogates for the downstream anabolic response. For continuous PTH dosing, the model predicts a desensitized system dominated by the inactive receptor and complex, consistent with downstream net bone loss that has been demonstrated experimentally. Using pulsatile PTH dosing, the model system predicts a highly sensitized state dominated by the active receptor and complex, corresponding to net bone formation. These results are consistent with the hypothesis that the kinetics of the receptor plays a critical role in the downstream effects of PTH dosing. Moreover, these results indicate that within a range of biologically relevant PTH doses, the two-state model is able to capture the differential behavior of the system for both continuous and pulsatile PTH dosing. The development of such a model provides a rational basis for developing more biologically extensive models that may support the design of optimal dosing strategies for PTH-based anti-osteoporosis treatments. Moreover, this model provides a unique starting point from which to design experiments investigating PTH receptor biology.


Asunto(s)
Resorción Ósea/terapia , Modelos Biológicos , Hormona Paratiroidea/administración & dosificación , Receptor de Hormona Paratiroídea Tipo 1/fisiología , Animales , Humanos , Cinética , Hormona Paratiroidea/uso terapéutico , Transducción de Señal
5.
Curr Opin Pharmacol ; 4(3): 290-4, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15140422

RESUMEN

The increase in marrow adipogenesis associated with osteoporosis and age-related osteopenia is well known clinically. However, we are only now beginning to understand the mechanisms that control the differentiation of mesenchymal stem cells to either osteoblasts or adipocytes. Recent work with gene silencing and overexpression has provided insight into critical pathways that determine the fate of these multipotential cells. One of these pathways - that of the nuclear hormone receptor peroxisome proliferator activated receptor-gamma - when activated, promotes adipogenesis and inhibits osteogenesis. This in vitro mechanism of action has been confirmed in vivo using ligands to this receptor. Discovery of this and other targets and pathways, such as Wnt signaling, notch/delta/jagged ligands and receptors, and RhoA gene expression, provides new insights into mesenchymal stem cell differentiation. These pathways provide exciting future pharmacological targets with which to enhance bone formation and therefore reduce the risk of fracture.


Asunto(s)
Adipocitos/fisiología , Envejecimiento/metabolismo , Células de la Médula Ósea/fisiología , Osteoblastos/fisiología , Osteoporosis/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Adipocitos/metabolismo , Animales , Femenino , Humanos , Osteoblastos/metabolismo , Osteoporosis/etiología , Osteoporosis/prevención & control , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
6.
Clin Biochem ; 45(12): 874-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22429519

RESUMEN

OBJECTIVES: The authors have set out to evaluate the literature relevant to the dynamic regulation of adipogenesis and osteogenesis. DESIGN AND METHODS: A detailed search of the past and recent literature was conducted on Pubmed using a combination of keywords including: adipogenesis, bone marrow, hematopoiesis, mesenchymal stromal/stem cell, and osteogenesis. RESULTS: Throughout one's lifespan, the bone marrow microenvironment provides a unique niche for mesenchymal stromal/stem cells (BMSCs) and hematopoietic stem cells (HSCs). The marrow changes as a function of biological age and pathophysiology. Historically, clinical biochemistry has observed these changes from an HSC and hematological perspective. Nevertheless, these changes also reflect the balance between BMSC adipogenic and osteogenic processes which can display an inverse or reciprocal relationship. Multiple hormonal factors and nuclear hormone receptor ligands and drugs are responsible for BMSC lineage selection. Data from a number of laboratories now implicates endocrine feedback loops between extramedullary adipose depots and the central nervous system. CONCLUSIONS: This concise review provides a perspective on the mechanisms regulating BMSC differentiation in the context of biological aging, obesity, and osteoporosis.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Células de la Médula Ósea/fisiología , Huesos/metabolismo , Células Madre Mesenquimatosas/fisiología , Tejido Adiposo Blanco/patología , Envejecimiento/metabolismo , Animales , Huesos/patología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Obesidad/metabolismo , Obesidad/patología , Osteoporosis/metabolismo , Osteoporosis/patología
7.
Curr Pharm Des ; 17(4): 332-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21375497

RESUMEN

Information relating to the biology, culture expansion, and mechanisms relating to adipose-derived cells has advanced significantly in the past decade. Both the heterogeneous stromal vascular fraction (SVF) and more homogeneous adipose-derived stem cells (ASC) offer unique opportunities as novel cell-based therapeutics and as traditional pharmaceutical discovery tools. This review highlights the cytokine secretory functions of ASC and SVF cells as well as their potential use as immunomodulators and gene delivery vehicles. These functions make it feasible to exploit adipose-derived cells in the treatment of ischemic, musculoskeletal, and oncological disorders. With appropriate commercial development and in vivo validation, ASC and SVF cells will have a significant therapeutic impact in the future.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad , Diseño de Fármacos , Medicina Regenerativa/métodos , Células Madre/fisiología , Células del Estroma/fisiología , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/citología , Animales , Humanos , Células del Estroma/citología
8.
J Cell Biochem ; 98(2): 251-66, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16479589

RESUMEN

The relationship between bone and fat formation within the bone marrow microenvironment is complex and remains an area of active investigation. Classical in vitro and in vivo studies strongly support an inverse relationship between the commitment of bone marrow-derived mesenchymal stem cells or stromal cells to the adipocyte and osteoblast lineage pathways. In this review, we focus on the recent literature exploring the mechanisms underlying these differentiation events and discuss their implications relevant to osteoporosis and regenerative medicine.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Huesos/metabolismo , Células Madre Mesenquimatosas/citología , Adipocitos/citología , Animales , Densidad Ósea/fisiología , Enfermedades Óseas Metabólicas/fisiopatología , Diferenciación Celular , Niño , Colesterol/metabolismo , Ejercicio Físico/fisiología , Hematopoyesis/fisiología , Humanos , Obesidad/metabolismo , Osteogénesis/fisiología , Osteoporosis/fisiopatología , Inhibidores de Proteasas/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal/fisiología
9.
J Cell Physiol ; 205(1): 133-40, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15828017

RESUMEN

Apoptosis may play a role in osteoarthritis (OA). Apoptosis can proceed via two different pathways depending on the stimulus. However, both pathways converge upon the effector caspases, caspases-3 and -7. In some systems inhibition of caspases-3 and -7 can prevent apoptosis and may therefore have important therapeutic implications. To confirm this, apoptosis was induced in canine chondrocytes with mitomycin-c (MMC), either in the presence or absence of the general caspase inhibitor, Z-VAD FMK, or a specific caspase-3/7 inhibitor. Z-VAD FMK prevented MMC induced cell death. In contrast, inhibition of caspases-3 and -7 in the presence of MMC induced morphological changes that could be described as necrotic-like or paraptotic-like but did not prevent cell death. The addition of an inhibitor of caspase-8 or caspase-9 along with inhibitor of caspase-3/7 was required to reduce cell death. The morphological changes did not occur in the presence of the caspase-3/7 inhibitor alone and could be prevented by addition of Z-VAD FMK. These data lead to the conclusion that, if the apoptotic program cannot be completed, the cells are pushed into a necrotic or other nonapoptotic mode of death which may involve caspase-8 and/or caspase-9.


Asunto(s)
Inhibidores de Caspasas , Condrocitos/citología , Condrocitos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Mitomicina/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 7 , Caspasas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Condrocitos/enzimología , Perros , Microscopía Electrónica de Transmisión , Fenotipo
10.
Endocrine ; 23(2-3): 183-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15146099

RESUMEN

Until recently, adipose tissue was considered to serve only as a triglyceride reservoir and was relegated to a passive endocrine role. With the discovery of leptin and other adipokines, adipose tissue is now recognized as an active participant in systemic metabolism. This review focuses on the complex relationship existing between adipose tissue and bone metabolism and differentiation. It explores the paradigms that have shaped the past decade's research and what these findings forecast for the future. Particular attention is given to the multipotent adult stem cell populations that reside within bone and fat. These adult stem cells have critical importance to the emerging field of tissue engineering and regenerative medicine.


Asunto(s)
Tejido Adiposo/anatomía & histología , Tejido Adiposo/fisiología , Huesos/anatomía & histología , Huesos/fisiología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Células de la Médula Ósea/citología , Huesos/citología , Huesos/metabolismo , Diferenciación Celular , Humanos , Mesodermo/citología , Modelos Biológicos , Osteoblastos/citología , Células Madre/citología , Células del Estroma/citología , Relación Estructura-Actividad
11.
Expert Opin Ther Targets ; 6(6): 679-89, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12472380

RESUMEN

Advances in genomics and proteomics have revolutionised the drug discovery process and target validation. Identification of novel therapeutic targets for chronic skeletal diseases is an extremely challenging process based on the difficulty of obtaining high-quality human diseased versus normal tissue samples. The quality of tissue and genomic information obtained from the sample is critical to identifying disease-related genes. Using a genomics-based approach, novel genes or genes with similar homology to existing genes can be identified from cDNA libraries generated from normal versus diseased tissue. High-quality cDNA libraries are prepared from uncontaminated homogeneous cell populations harvested from tissue sections of interest. Localised gene expression analysis and confirmation are obtained through in situ hybridisation or immunohistochemical studies. Cells overexpressing the recombinant protein are subsequently designed for primary cell-based high-throughput assays that are capable of screening large compound banks for potential hits. Afterwards, secondary functional assays are used to test promising compounds. The same overexpressing cells are used in the secondary assay to test protein activity and functionality as well as screen for small-molecule agonists or antagonists. Once a hit is generated, a structure-activity relationship of the compound is optimised for better oral bioavailability and pharmacokinetics allowing the compound to progress into development. Parallel efforts from proteomics, as well as genetics/transgenics, bioinformatics and combinatorial chemistry, and improvements in high-throughput automation technologies, allow the drug discovery process to meet the demands of the medicinal market. This review discusses and illustrates how different approaches are incorporated into the discovery and validation of novel targets and, consequently, the development of potentially therapeutic agents in the areas of osteoporosis and osteoarthritis. While current treatments exist in the form of hormone replacement therapy, antiresorptive and anabolic agents for osteoporosis, there are no disease-modifying therapies for the treatment of the most common human joint disease, osteoarthritis. A massive market potential for improved options with better safety and efficacy still remains. Therefore, the application of genomics and proteomics for both diseases should provide much needed novel therapeutic approaches to treating these major world health problems.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Animales , Proteínas Portadoras , Caspasas/fisiología , Catepsina K , Catepsinas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas de Unión al ADN/fisiología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Proteína 1 de la Respuesta de Crecimiento Precoz , Genómica , Glicoproteínas/fisiología , Humanos , Proteínas Inmediatas-Precoces/fisiología , Glicoproteínas de Membrana , Ratones , Ratones Noqueados , Ratones Transgénicos , Estructura Molecular , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoprotegerina , Proteómica , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Transcripción/fisiología , Técnicas del Sistema de Dos Híbridos , Familia-src Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA