Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Biomaterials ; 80: 11-19, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26701143

ABSTRACT

Biomaterial scaffolds are central to many regenerative strategies as they create a space for infiltration of host tissue and provide a platform to deliver growth factors and progenitor cells. However, biomaterial implantation results in an unavoidable inflammatory response, which can impair tissue regeneration and promote loss or dysfunction of transplanted cells. We investigated localized TGF-ß1 delivery to modulate this immunological environment around scaffolds and transplanted cells. TGF-ß1 was delivered from layered scaffolds, with protein entrapped within an inner layer and outer layers designed for cell seeding and host tissue integration. Scaffolds were implanted into the epididymal fat pad, a site frequently used for cell transplantation. Expression of cytokines TNF-α, IL-12, and MCP-1 were decreased by at least 40% for scaffolds releasing TGF-ß1 relative to control scaffolds. This decrease in inflammatory cytokine production corresponded to a 60% decrease in leukocyte infiltration. Transplantation of islets into diabetic mice on TGF-ß1 scaffolds significantly improved the ability of syngeneic islets to control blood glucose levels within the first week of transplant and delayed rejection of allogeneic islets. Together, these studies emphasize the ability of localized TGF-ß1 delivery to modulate the immune response to biomaterial implants and enhance cell function in cell-based therapies.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/therapeutic use , Animals , Cells, Cultured , Chemokine CCL2/immunology , Diabetes Mellitus, Experimental/immunology , Drug Delivery Systems/methods , Immunomodulation/drug effects , Interleukin-12/immunology , Male , Mice , Mice, Inbred C57BL , Porosity , Tumor Necrosis Factor-alpha/immunology
2.
Biomaterials ; 35(6): 2024-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24309498

ABSTRACT

Biomaterial scaffolds are central to many tissue engineering strategies as they create a space for tissue growth and provide a support for cell adhesion and migration. However, biomaterial implantation results in unavoidable injury resulting in an inflammatory response, which can impair integration with the host and tissue regeneration. Toward the goal of reducing inflammation, we investigated the hypothesis that a lentiviral gene therapy-based approach to localized and sustained IL-10 expression at a scaffold could modulate the number, relative proportions, and cytokine production of infiltrating leukocyte populations. Flow cytometry was used to quantify infiltration of six leukocyte populations for 21 days following implantation of PLG scaffolds into intraperitoneal fat. Leukocytes with innate immune functions (i.e., macrophages, dendritic cells, neutrophils) were most prevalent at early time points, while T lymphocytes became prevalent by day 14. Reporter gene delivery indicated that transgene expression persisted at the scaffold for up to 28 days and macrophages were the most common leukocyte transduced, while transduced dendritic cells expressed the greatest levels of transgene. IL-10 delivery decreased leukocyte infiltration by 50% relative to controls, increased macrophage IL-10 expression, and decreased macrophage, dendritic cell, and CD4 T cell IFN-γ expression. Thus, IL-10 gene delivery significantly decreased inflammation following scaffold implant into the intraperitoneal fat, in part by modulating cytokine expression of infiltrating leukocytes.


Subject(s)
Gene Expression Regulation , Interleukin-10/metabolism , Leukocytes/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Flow Cytometry , Genetic Therapy , Immunomodulation/physiology , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...