Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Exp Biol Med (Maywood) ; 241(10): 1084-97, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27229903

RESUMO

Monocytes and macrophages play a critical role in tissue development, homeostasis, and injury repair. These innate immune cells participate in guiding vascular remodeling, stimulation of local stem and progenitor cells, and structural repair of tissues such as muscle and bone. Therefore, there is a great interest in harnessing this powerful endogenous cell source for therapeutic regeneration through immunoregenerative biomaterial engineering. These materials seek to harness specific subpopulations of monocytes/macrophages to promote repair by influencing their recruitment, positioning, differentiation, and function within a damaged tissue. Monocyte and macrophage phenotypes span a continuum of inflammatory (M1) to anti-inflammatory or pro-regenerative cells (M2), and their heterogeneous functions are highly dependent on microenvironmental cues within the injury niche. Increasing evidence suggests that division of labor among subpopulations of monocytes and macrophages could allow for harnessing regenerative functions over inflammatory functions of myeloid cells; however, the complex balance between necessary functions of inflammatory versus regenerative myeloid cells remains to be fully elucidated. Historically, biomaterial-based therapies for promoting tissue regeneration were designed to minimize the host inflammatory response; although, recent appreciation for the roles that innate immune cells play in tissue repair and material integration has shifted this paradigm. A number of opportunities exist to exploit known signaling systems of specific populations of monocytes/macrophages to promote repair and to better understand the biological and pathological roles of myeloid cells. This review seeks to outline the characteristics of distinct populations of monocytes and macrophages, identify the role of these cells within diverse tissue injury niches, and offer design criteria for immunoregenerative biomaterials given the intrinsic inflammatory response to their implantation.


Assuntos
Materiais Biocompatíveis/farmacologia , Regeneração Tecidual Guiada/métodos , Fatores Imunológicos/farmacologia , Macrófagos/fisiologia , Monócitos/fisiologia , Ferimentos e Lesões/terapia , Animais , Materiais Biocompatíveis/metabolismo , Humanos , Fatores Imunológicos/metabolismo , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos
2.
Biomaterials ; 64: 98-107, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26125501

RESUMO

Bone grafting procedures are performed to treat wounds incurred during wartime trauma, accidents, and tumor resections. Endogenous mechanisms of repair are often insufficient to ensure integration between host and donor bone and subsequent restoration of function. We investigated the role that bone marrow-derived cells play in bone regeneration and sought to increase their contributions by functionalizing bone allografts with bioactive lipid coatings. Polymer-coated allografts were used to locally deliver the immunomodulatory small molecule FTY720 in tibial defects created in rat bone marrow chimeras containing genetically-labeled bone marrow for monitoring cell origin and fate. Donor bone marrow contributed significantly to both myeloid and osteogenic cells in remodeling tissue surrounding allografts. FTY720 coatings altered the phenotype of immune cells two weeks post-injury, which was associated with increased vascularization and bone formation surrounding allografts. Consequently, degradable polymer coating strategies that deliver small molecule growth factors such as FTY720 represent a novel therapeutic strategy for harnessing endogenous bone marrow-derived progenitors and enhancing healing in load-bearing bone defects.


Assuntos
Transplante Ósseo , Cloridrato de Fingolimode/administração & dosagem , Fatores Imunológicos/administração & dosagem , Aloenxertos , Animais , Células da Medula Óssea/citologia , Osso e Ossos/fisiologia , Linhagem da Célula , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Ácido Láctico/administração & dosagem , Masculino , Mielopoese/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ácido Poliglicólico/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Quimera por Radiação , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Regeneração , Estresse Mecânico , Tíbia/lesões , Tíbia/cirurgia , Microtomografia por Raio-X
3.
Curr Pharm Des ; 19(19): 3403-19, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23432670

RESUMO

Regeneration of bone requires the coordinated processes of angiogenesis and osteogenesis. These repair mechanisms are closely linked through both direct cell-cell contact and indirect paracrine signaling among osteoblasts, endothelial cells, and other cell types. The vasculature provides a source of nutrients, oxygen, metabolic substrates, and access for circulating cells that help to support new bone formation. The complexity of the endogenous signaling axis that promotes angiogenesis provides numerous opportunities for therapeutic intervention ranging from progenitor cell mobilization to endothelial proliferation and sprouting. Small molecules are particularly appealing for regenerative medicine applications because many exhibit extended in vivo stability, low cost, and scalable production. Innovative techniques for developing small molecules such as high throughput functional assays and broad-spectrum database analysis techniques have led to the development of new compounds and the identification of novel applications of existing drugs. In addition, rapid advances in biomaterials design and synthesis provide platforms to deliver therapeutic small molecules to sites of bone injury. This review presents an overview of current strategies for harnessing endogenous healing mechanisms using small molecules by targeting angiogenesis, osteogenesis, or both.


Assuntos
Produtos Biológicos/farmacologia , Regeneração Óssea/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Produtos Biológicos/administração & dosagem , Produtos Biológicos/química , Produtos Biológicos/uso terapêutico , Osso e Ossos/irrigação sanguínea , Fraturas Ósseas/tratamento farmacológico , Humanos , Estrutura Molecular , Doenças Musculoesqueléticas/tratamento farmacológico , Osteogênese/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/uso terapêutico
4.
Biomaterials ; 34(38): 9853-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24064148

RESUMO

Endogenous signals originating at the site of injury are involved in the paracrine recruitment, proliferation, and differentiation of circulating progenitor and diverse inflammatory cell types. Here, we investigate a strategy to exploit endogenous cell recruitment mechanisms to regenerate injured bone by local targeting and activation of sphingosine-1-phosphate (S1P) receptors. A mandibular defect model was selected for evaluating regeneration of bone following trauma or congenital disease. The particular challenges of mandibular reconstruction are inherent in the complex anatomy and function of the bone given that the area is highly vascularized and in close proximity to muscle. Nanofibers composed of poly(DL-lactide-co-glycolide) (PLAGA) and polycaprolactone (PCL) were used to delivery FTY720, a targeted agonist of S1P receptors 1 and 3. In vitro culture of bone progenitor cells on drug-loaded constructs significantly enhanced SDF1α mediated chemotaxis of bone marrow mononuclear cells. In vivo results show that local delivery of FTY720 from composite nanofibers enhanced blood vessel ingrowth and increased recruitment of M2 alternatively activated macrophages, leading to significant osseous tissue ingrowth into critical sized defects after 12 weeks of treatment. These results demonstrate that local activation of S1P receptors is a regenerative cue resulting in recruitment of wound healing or anti-inflammatory macrophages and bone healing. Use of such small molecule therapy can provide an alternative to biological factors for the clinical treatment of critical size craniofacial defects.


Assuntos
Macrófagos/metabolismo , Mandíbula , Nanofibras/química , Receptores de Lisoesfingolipídeo/metabolismo , Cicatrização/fisiologia , Animais , Cloridrato de Fingolimode , Lisofosfolipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Poliésteres/química , Propilenoglicóis/química , Ratos , Esfingosina/análogos & derivados , Esfingosina/química
5.
Acta Biomater ; 8(1): 31-40, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21920469

RESUMO

Poly(ethylene glycol) (PEG) microspheres were assembled around HL-1 cardiomyocytes to produce highly porous modular scaffolds. In this study we took advantage of the immiscibility of PEG and dextran to improve upon our previously described modular scaffold fabrication methods. Phase separating the PEG microspheres in dextran solutions caused them to rapidly deswell and crosslink together, eliminating the need for serum protein-based crosslinking. This also led to a dramatic increase in the stiffness of the scaffolds and greatly improved the handling characteristics. HL-1 cardiomyocytes were present during microsphere crosslinking in the cytocompatible dextran solution, exhibiting high cell viability following scaffold formation. Over the course of 2 weeks a 9-fold expansion in cell number was observed. The cardiac functional markers sarcomeric α-actinin and connexin 43 were expressed at 13 and 24 days after scaffold formation. HL-1 cells were spontaneously depolarizing 38 days after scaffold formation, which was visualized by confocal microscopy using a calcium-sensitive dye. Electrical stimulation resulted in synchronization of activation peaks throughout the scaffolds. These findings demonstrate that PEG microsphere scaffolds fabricated in the presence of dextran can support the long-term three-dimensional culture of cells, suggesting applications in cardiovascular tissue engineering.


Assuntos
Materiais Biocompatíveis/química , Microesferas , Miócitos Cardíacos/citologia , Polietilenoglicóis/química , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Reagentes de Ligações Cruzadas/química , Dextranos/química , Estimulação Elétrica , Teste de Materiais , Camundongos , Miócitos Cardíacos/metabolismo , Polietilenoglicóis/metabolismo , Porosidade , Estresse Mecânico , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA