Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-36533683

ABSTRACT

Stem cell derived ß-cells have demonstrated the potential to control blood glucose levels and represent a promising treatment for Type 1 diabetes (T1D). Early engraftment post-transplantation and subsequent maturation of these ß-cells are hypothesized to be limited by the initial inflammatory response, which impacts the ability to sustain normoglycemia for long periods. We investigated the survival and development of immature hPSC-derived ß-cells transplanted on poly(lactide-co-glycolide) (PLG) microporous scaffolds into the peritoneal fat, a site being considered for clinical translation. The scaffolds were modified with biotin for binding of a streptavidin-FasL (SA-FasL) chimeric protein to modulate the local immune cell responses. The presence of FasL impacted infiltration of monocytes and neutrophils and altered the immune cell polarization. Conditioned media generated from SA-FasL scaffolds explanted at day 4 post-transplant did not impact hPSC-derived ß-cell survival and maturation in vitro, while these responses were reduced with conditioned media from control scaffolds. Following transplantation, ß-cell viability and differentiation were improved with SA-FasL modification. A sustained increase in insulin positive cell ratio was observed with SA-FasL-modified scaffolds relative to control scaffolds. These results highlight that the initial immune response can significantly impact ß-cell engraftment, and modulation of cell infiltration and polarization may be a consideration for supporting long-term function at an extrahepatic site.

2.
Sci Adv ; 8(19): eabm9881, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35559682

ABSTRACT

Islet transplantation to treat insulin-dependent diabetes is greatly limited by the need for maintenance immunosuppression. We report a strategy through which cotransplantation of allogeneic islets and streptavidin (SA)-FasL-presenting microgels to the omentum under transient rapamycin monotherapy resulted in robust glycemic control, sustained C-peptide levels, and graft survival in diabetic nonhuman primates for >6 months. Surgical extraction of the graft resulted in prompt hyperglycemia. In contrast, animals receiving microgels without SA-FasL under the same rapamycin regimen rejected islet grafts acutely. Graft survival was associated with increased number of FoxP3+ cells in the graft site with no significant changes in T cell systemic frequencies or responses to donor and third-party antigens, indicating localized tolerance. Recipients of SA-FasL microgels exhibited normal liver and kidney metabolic function, demonstrating safety. This localized immunomodulatory strategy succeeded with unmodified islets and does not require long-term immunosuppression, showing translational potential in ß cell replacement for treating type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Microgels , Allografts/metabolism , Animals , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Primates , Sirolimus , Streptavidin
3.
J Immunol ; 204(10): 2840-2851, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32253240

ABSTRACT

Allogeneic islet transplantation is limited by adverse effects of chronic immunosuppression used to control rejection. The programmed cell death 1 pathway as an important immune checkpoint has the potential to obviate the need for chronic immunosuppression. We generated an oligomeric form of programmed cell death 1 ligand chimeric with core streptavidin (SA-PDL1) that inhibited the T effector cell response to alloantigens and converted T conventional cells into CD4+Foxp3+ T regulatory cells. The SA-PDL1 protein was effectively displayed on the surface of biotinylated mouse islets without a negative impact islet viability and insulin secretion. Transplantation of SA-PDL1-engineered islet grafts with a short course of rapamycin regimen resulted in sustained graft survival and function in >90% of allogeneic recipients over a 100-d observation period. Long-term survival was associated with increased levels of intragraft transcripts for innate and adaptive immune regulatory factors, including IDO-1, arginase-1, Foxp3, TGF-ß, IL-10, and decreased levels of proinflammatory T-bet, IL-1ß, TNF-α, and IFN-γ as assessed on day 3 posttransplantation. T cells of long-term graft recipients generated a proliferative response to donor Ags at a similar magnitude to T cells of naive animals, suggestive of the localized nature of tolerance. Immunohistochemical analyses showed intense peri-islet infiltration of T regulatory cells in long-term grafts and systemic depletion of this cell population resulted in prompt rejection. The transient display of SA-PDL1 protein on the surface of islets serves as a practical means of localized immunomodulation that accomplishes sustained graft survival in the absence of chronic immunosuppression with potential clinical implications.


Subject(s)
Allografts/physiology , B7-H1 Antigen/metabolism , Diabetes Mellitus, Type 1/immunology , Immunosuppression Therapy/methods , Islets of Langerhans/physiology , Streptavidin/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , B7-H1 Antigen/genetics , Cell Differentiation , Cell Survival , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Immunity/genetics , Immunomodulation , Islets of Langerhans Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Streptavidin/genetics
4.
Am J Transplant ; 20(5): 1285-1295, 2020 05.
Article in English | MEDLINE | ID: mdl-31850658

ABSTRACT

We have previously shown that pancreatic islets engineered to transiently display a modified form of FasL protein (SA-FasL) on their surface survive indefinitely in allogeneic recipients without a need for chronic immunosuppression. Mechanisms that confer long-term protection to allograft are yet to be elucidated. We herein demonstrated that immune protection evolves in two distinct phases; induction and maintenance. SA-FasL-engineered allogeneic islets survived indefinitely and conferred protection to a second set of donor-matched, but not third-party, unmanipulated islet grafts simultaneously transplanted under the contralateral kidney capsule. Protection at the induction phase involved a reduction in the frequency of proliferating alloreactive T cells in the graft-draining lymph nodes, and required phagocytes and TGF-ß. At the maintenance phase, immune protection evolved into graft site-restricted immune privilege as the destruction of long-surviving SA-FasL-islet grafts by streptozotocin followed by the transplantation of a second set of unmanipulated islet grafts into the same site from the donor, but not third party, resulted in indefinite survival. The induced immune privilege required both CD4+ CD25+ Foxp3+ Treg cells and persistent presence of donor antigens. Engineering cell and tissue surfaces with SA-FasL protein provides a practical, efficient, and safe means of localized immunomodulation with important implications for autoimmunity and transplantation.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Fas Ligand Protein , Graft Survival , Immune Privilege , Immune Tolerance
5.
Methods Mol Biol ; 1139: 269-85, 2014.
Article in English | MEDLINE | ID: mdl-24619687

ABSTRACT

Therapeutic vaccines against cancer are at their prime, owing to our comprehensive understanding of immune effector responses generated against tumor and the mechanisms employed by the progressing tumor to evade the immune system. The immune system is primed by tumor-associated antigens (TAA) that are perceived as foreign. Therefore, the identification of TAA led to the development of subunit vaccine formulations comprising defined TAA as stand-alone vaccines or in combination with immune adjuvants. Inasmuch as cancer cells express a diverse set of TAA, novel immunomodulatory approaches that not only use tumor cells as a source of diverse TAA but also convert them into competent antigen-presenting cells have significant therapeutic potential as cell-based vaccines. Toward this end, we have developed a novel protein display approach designated as ProtEx™ as a safe and efficient alternative to DNA-based gene expression to generate novel immunomodulatory molecules and display them on tumor cells for the development of cancer vaccines. This chapter describes the ProtEx™ technology and its application to the generation of tumor cell-based cancer vaccines.


Subject(s)
Cancer Vaccines/immunology , Cell Engineering/methods , Neoplasms/immunology , Neoplasms/therapy , Vaccination , Animals , Cancer Vaccines/genetics , Cell Line , Cell Transformation, Neoplastic , Chromatography, Affinity , Cloning, Molecular , Drosophila/cytology , Neoplasms/genetics , Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Streptavidin/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL