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.
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
2.
J Immunol ; 187(11): 5901-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22068235

ABSTRACT

Allogeneic islet transplantation is an important therapeutic approach for the treatment of type 1 diabetes. Clinical application of this approach, however, is severely curtailed by allograft rejection primarily initiated by pathogenic effector T cells regardless of chronic use of immunosuppression. Given the role of Fas-mediated signaling in regulating effector T cell responses, we tested if pancreatic islets can be engineered ex vivo to display on their surface an apoptotic form of Fas ligand protein chimeric with streptavidin (SA-FasL) and whether such engineered islets induce tolerance in allogeneic hosts. Islets were modified with biotin following efficient engineering with SA-FasL protein that persisted on the surface of islets for >1 wk in vitro. SA-FasL-engineered islet grafts established euglycemia in chemically diabetic syngeneic mice indefinitely, demonstrating functionality and lack of acute toxicity. Most importantly, the transplantation of SA-FasL-engineered BALB/c islet grafts in conjunction with a short course of rapamycin treatment resulted in robust localized tolerance in 100% of C57BL/6 recipients. Tolerance was initiated and maintained by CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells, as their depletion early during tolerance induction or late after established tolerance resulted in prompt graft rejection. Furthermore, Treg cells sorted from graft-draining lymph nodes, but not spleen, of long-term graft recipients prevented the rejection of unmodified allogeneic islets in an adoptive transfer model, further confirming the Treg role in established tolerance. Engineering islets ex vivo in a rapid and efficient manner to display on their surface immunomodulatory proteins represents a novel, safe, and clinically applicable approach with important implications for the treatment of type 1 diabetes.


Subject(s)
Fas Ligand Protein/immunology , Immune Tolerance/immunology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/surgery , Graft Rejection/immunology , Graft Rejection/prevention & control , Immunohistochemistry , Indicators and Reagents , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Streptavidin/immunology , Transplantation, Homologous
3.
Vaccine ; 28(2): 512-22, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19836479

ABSTRACT

Agonistic Abs to select costimulatory members of CD28 and TNFR family have shown efficacy in various preclinical cancer immunotherapeutic settings. However, the use of agonistic Abs is often associated with severe toxicity due to non-specific activation of lymphocytes. We hypothesized that natural costimulatory ligands may serve as more potent and safer alternative to agonistic Abs for immunotherapy. In this communication, we focused on 4-1BBL as the molecule of choice because of the pleiotropic effects of 4-1BB signaling in the immune system and the demonstrated therapeutic efficacy of 4-1BB agonistic Abs in preclinical cancer and infection models. We report that a novel form of soluble ligand, SA-4-1BBL, delivered more potent and qualitatively different signals to T cells than an agonistic Ab. Importantly, while treatment of naïve mice with the agonistic Ab resulted in severe toxicity, as assessed by enlarged spleen and peripheral LNs, non-specific T cell proliferation, hepatitis, and systemic inflammatory cytokine production, treatment with SA-4-1BBL lacked these immune anomalies. Agonistic Ab treatment produced full toxicity in FcgammaR(-/-) or complement C1q(-/-) or C3(-/-) knockout mice, suggesting lack of involvement of stimulatory FcgammaRs or complement system in the observed toxicity. Naïve and memory T cells served as direct targets of anti-4-1BB Ab-mediated toxicity. Potent immunostimulatory activity combined with lack of toxicity rationalizes further development of soluble SA-4-1BBL as an immunomodulatory component of therapeutic vaccines against cancer and chronic infections.


Subject(s)
4-1BB Ligand/metabolism , Antibodies/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , 4-1BB Ligand/adverse effects , Adjuvants, Immunologic/adverse effects , Animals , Antibodies/adverse effects , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , T-Lymphocytes/immunology
4.
Cancer Res ; 69(10): 4319-26, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435920

ABSTRACT

Vaccines represent an attractive treatment modality for the management of cancer primarily because of their specificity and generation of immunologic memory important for controlling recurrences. However, the efficacy of therapeutic vaccines may require formulations that not only generate effective immune responses but also overcome immune evasion mechanisms employed by progressing tumor. Costimulatory molecules play critical roles in modulating innate, adaptive, and regulatory immunity and have potential to serve as effective immunomodulatory components of therapeutic vaccines. In this study, we tested the function of a novel soluble form of 4-1BB ligand (4-1BBL) costimulatory molecule in modulating innate, adaptive, and regulatory immunity and assessed its therapeutic efficacy in the HPV-16 E7-expressing TC-1 cervical cancer and survivin-expressing 3LL lung carcinoma mouse models. Vaccination with 4-1BBL activated dendritic cells and enhanced antigen uptake, generated CD8(+) T-cell effector/memory responses, and endowed T effector cells refractory to suppression by CD4(+)CD25(+)FoxP3(+) T regulatory cells. Immunization with 4-1BBL in combination with an E7 peptide or survivin protein resulted in eradication of TC-1 and 3LL tumors, respectively. 4-1BBL was more effective than TLR agonists LPS, MPL, and CpG and an agonistic 4-1BB antibody as a component of E7 peptide-based therapeutic vaccine for the generation of immune responses and eradication of TC-1 established tumors in the absence of detectable toxicity. Therapeutic efficacy was associated with reversal of tumor-mediated nonresponsiveness/anergy as well as establishment of long-term CD8(+) T-cell memory. Potent pleiotropic immunomodulatory activities combined with lack of toxicity highlight the potential of 4-1BBL molecule as an effective component of therapeutic cancer vaccines.


Subject(s)
4-1BB Ligand/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Animals , Female , Humans , Immunologic Memory/drug effects , Inhibitor of Apoptosis Proteins , Lung Neoplasms/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/therapeutic use , Repressor Proteins , Survivin , Uterine Cervical Neoplasms/immunology
5.
J Immunol ; 181(2): 931-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606644

ABSTRACT

The critical role played by Fas ligand (FasL) in immune homeostasis renders this molecule an attractive target for immunomodulation to achieve tolerance to auto- and transplantation Ags. Immunomodulation with genetically modified cells expressing FasL was shown to induce tolerance to alloantigens. However, genetic modification of primary cells in a rapid, efficient, and clinically applicable manner proved challenging. Therefore, we tested the efficacy of donor splenocytes rapidly and efficiently engineered to display on their surface a chimeric form of FasL protein (SA-FasL) for tolerance induction to cardiac allografts. The i.p. injection of ACI rats with Wistar-Furth rat splenocytes displaying SA-FasL on their surface resulted in tolerance to donor, but not F344 third-party cardiac allografts. Tolerance was associated with apoptosis of donor reactive T effector cells and induction/expansion of CD4(+)CD25(+)FoxP3(+) T regulatory (Treg) cells. Treg cells played a critical role in the observed tolerance as adoptive transfer of sorted Treg cells from long-term graft recipients into naive unmanipulated ACI rats resulted in indefinite survival of secondary Wistar-Furth grafts. Immunomodulation with allogeneic cells rapidly and efficiently engineered to display on their surface SA-FasL protein provides an effective and clinically applicable means of cell-based therapy with potential application to regenerative medicine, transplantation, and autoimmunity.


Subject(s)
Fas Ligand Protein/metabolism , Heart Transplantation/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Transplantation Tolerance , Adoptive Transfer , Animals , Cell Proliferation , Fas Ligand Protein/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Rats , Rats, Inbred ACI , Rats, Inbred F344 , Rats, Inbred WF , Recombinant Fusion Proteins/metabolism , Spleen/cytology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...