Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Aldo A. Rossini is active.

Publication


Featured researches published by Aldo A. Rossini.


Journal of Clinical Investigation | 1998

Long-term survival of skin allografts induced by donor splenocytes and anti-CD154 antibody in thymectomized mice requires CD4(+) T cells, interferon-gamma, and CTLA4.

Thomas G. Markees; Nancy E. Phillips; Ethel J. Gordon; Randolph J. Noelle; Leonard D. Shultz; John P. Mordes; Dale L. Greiner; Aldo A. Rossini

Treatment of C57BL/6 mice with one transfusion of BALB/c spleen cells and anti-CD154 (anti-CD40-ligand) antibody permits BALB/c islet grafts to survive indefinitely and BALB/c skin grafts to survive for approximately 50 d without further intervention. The protocol induces long-term allograft survival, but the mechanism is unknown. We now report: (a) addition of thymectomy to the protocol permitted skin allografts to survive for > 100 d, suggesting that graft rejection in euthymic mice results from thymic export of alloreactive T cells. (b) Clonal deletion is not the mechanism of underlying long-term graft survival, as recipient thymectomized mice were immunocompetent and harbor alloreactive T cells. (c) Induction of skin allograft acceptance initially depended on the presence of IFN-gamma, CTLA4, and CD4(+) T cells. Addition of anti-CTLA4 or anti-IFN-gamma mAb to the protocol was associated with prompt graft rejection, whereas anti-IL-4 mAb had no effect. The role of IFN-gamma was confirmed using knockout mice. (d) Graft survival was associated with the absence of IFN-gamma in the graft. (e) Long-term graft maintenance required the continued presence of CD4(+) T cells. The results suggest that, with modification, our short-term protocol may yield a procedure for the induction of long-term graft survival without prolonged immunosuppression.


Journal of Immunology | 2000

Treatment of Allograft Recipients with Donor-Specific Transfusion and Anti-CD154 Antibody Leads to Deletion of Alloreactive CD8+ T Cells and Prolonged Graft Survival in a CTLA4-Dependent Manner

Neal N. Iwakoshi; John P. Mordes; Thomas G. Markees; Nancy E. Phillips; Aldo A. Rossini; Dale L. Greiner

A two-element protocol consisting of one donor-specific transfusion (DST) plus a brief course of anti-CD154 mAb greatly prolongs the survival of murine islet, skin, and cardiac allografts. To study the mechanism of allograft survival, we determined the fate of tracer populations of alloreactive transgenic CD8+ T cells in a normal microenvironment. We observed that DST plus anti-CD154 mAb prolonged allograft survival and deleted alloreactive transgenic CD8+ T cells. Neither component alone did so. Skin allograft survival was also prolonged in normal recipients treated with anti-CD154 mAb plus a depleting anti-CD8 mAb and in C57BL/6-CD8 knockout mice treated with anti-CD154 mAb monotherapy. We conclude that, in the presence of anti-CD154 mAb, DST leads to an allotolerant state, in part by deleting alloreactive CD8+ T cells. Consistent with this conclusion, blockade of CTLA4, which is known to abrogate the effects of DST and anti-CD154 mAb, prevented the deletion of alloreactive transgenic CD8+ T cells. These results document for the first time that peripheral deletion of alloantigen-specific CD8+ T cells is an important mechanism through which allograft survival can be prolonged by costimulatory blockade. We propose a unifying mechanism to explain allograft prolongation by DST and blockade of costimulation.


The American Journal of Medicine | 1981

Animal models of diabetes

John P. Mordes; Aldo A. Rossini

Spontaneous diabetes is a common occurrence in many animal species. In addition, animals can be rendered diabetic by a wide variety of experimental procedures. Diabetic animals may be regarded as models of the disease in man. However, such animals display a wide diversity of pathophysiology, and, in fact, no animal syndrome corresponds precisely to any type of diabetes in human subjects. The most common diabetes syndromes in animals occur in the context of obesity, hyperinsulinemia and insulin resistance. Many such syndromes remit spontaneously. Dietary restriction and weight reduction effectively reverse some of these syndromes, but in other cases only partial correction of the syndrome occurs. Diabetes in lean animals is less common. The diabetes of lean animals is more frequently characterized by hypoinsulinemia, ketosis and insulin dependence than is the case with obese animals. Diabetes may be produced experimentally by means of surgery, viral infection or the administration of various hormones and chemical agents. Both the spontaneous and experimental animal models have been used effectively to study the etiologies, complications, treatments and prevention of diabetes.


Diabetes | 1976

Studies on Streptozotocin Diabetes

Om P. Ganda; Aldo A. Rossini; Arthur A. Like

Both alloxan and streptozotocin produce β-cell necrosis in the rat. Previous studies have shown protection against alloxan toxicity by D-glucose, D-mannose, and the nonmetabolized analogue 3-0-methyl-D-glucose and removal of this protective effect by D-mannoheptulose. The effect of several agents (i.v. infusion) against the β-cell toxic effect of streptozotocin (60 mg./kg. i.v. in 24-hour-fasted 200-gm. male rats) was studied. Protection was determined by plasma glucose concentrations 24 and 48 hours later and, in certain experiments, by histologic examination of the islets. D-glucose and D-mannose provided no protection. Similarly, D-galactose, D-fructose, α-methyl-D-glucoside, D-L-glyceraldehyde, D-xylose, and D-glucosamine had no effect. However, 3-0-methyl-D-glucose administered immediately before streptozotocin resulted in progressive inhibition of β-cell toxicity with complete protection at 0.83 mMoles per rat. The protective effect of 3-0-methyl-D-glucose was not altered by mannoheptulose. 2-Deoxy-D-glucose, which has no effect against alloxan, provided nearly complete protection against streptozotocin at 2.2 mMoles per rat. The effects of 3-0-methyl-D-glucose and 2-deoxy-D-glucose were additive and were not altered by glucose. Furthermore, the 3-0-methyl-D-glucose as well as 2-deoxy-D-glucose protective effects were still present, albeit attenuated, when these agents were given following the administration of streptozotocin. This is in contrast to alloxan, against which 3-0-methyl-D-glucose provides protection only when given before alloxan. 3-0-Methyl-D-glucose is the only carbohydrate protective against both streptozotocin and alloxan in the rat. However, several silent differences seem to exist between the mechanisms of beta-cytotoxic effects of these two diabetogenic compounds.


Clinical and Experimental Immunology | 2009

Human peripheral blood leucocyte non‐obese diabetic‐severe combined immunodeficiency interleukin‐2 receptor gamma chain gene mouse model of xenogeneic graft‐versus‐host‐like disease and the role of host major histocompatibility complex

Michael A. King; Laurence D. Covassin; Michael A. Brehm; Waldemar J. Racki; Todd Pearson; Jean Leif; Joseph Laning; W. Fodor; Oded Foreman; Lisa M. Burzenski; Thomas H. Chase; Bruce Gott; Aldo A. Rossini; Rita Bortell; Leonard D. Shultz; Dale L. Greiner

Immunodeficient non‐obese diabetic (NOD)‐severe combined immune‐deficient (scid) mice bearing a targeted mutation in the gene encoding the interleukin (IL)‐2 receptor gamma chain gene (IL2rγnull) engraft readily with human peripheral blood mononuclear cells (PBMC). Here, we report a robust model of xenogeneic graft‐versus‐host‐like disease (GVHD) based on intravenous injection of human PBMC into 2 Gy conditioned NOD‐scid IL2rγnull mice. These mice develop xenogeneic GVHD consistently (100%) following injection of as few as 5 × 106 PBMC, regardless of the PBMC donor used. As in human disease, the development of xenogeneic GVHD is highly dependent on expression of host major histocompatibility complex class I and class II molecules and is associated with severely depressed haematopoiesis. Interrupting the tumour necrosis factor‐α signalling cascade with etanercept, a therapeutic drug in clinical trials for the treatment of human GVHD, delays the onset and progression of disease. This model now provides the opportunity to investigate in vivo mechanisms of xenogeneic GVHD as well as to assess the efficacy of therapeutic agents rapidly.


Diabetes | 1977

Genetic Influence of the Streptozotocin-induced Insulitis and Hyperglycemia

Aldo A. Rossini; Michael C. Appel; Williams Rm; Arthur A. Like

Multiple injections of subdiabetogenic doses of streptozotodn (SZ) to CD-I male mice produce a diabetic syndrome that includes a cell-mediated immune reaction against the pancreatic islet. The importance of the host genetic background in the pathogenesis of this model of diabetes was studied by comparing various inbred strains of mice. Of eight strains of mice studied, only C57BL/KsJ developed insulitis and hyperglycemie comparable to that observed in CD-I mice. In two mouse strains (DBA/2J and BALB/cJ) having an haplotype similar to the C57BL/KsJ, only mild insulitis and glucose intolerance were observed. These data suggest that major histocompatibility complex genes, as presently defined, cannot be the only determinant of the severity of hyperglycemia and insulitis in this model.


Journal of Immunology | 2005

TLR Activation Synergizes with Kilham Rat Virus Infection to Induce Diabetes in BBDR Rats

Danny Zipris; Egil Lien; Jenny X. Xie; Dale L. Greiner; John P. Mordes; Aldo A. Rossini

Virus infection is hypothesized to be an important environmental “trigger” of type 1 diabetes in humans. We used the BBDR rat model to investigate the relationship between viral infection and autoimmune diabetes. BBDR rats are diabetes-free in viral Ab-free housing, but the disease develops in ∼30% of BBDR rats infected with Kilham rat virus (KRV) through a process that does not involve infection of pancreatic β cells. Pretreatment with polyinosinic-polycytidylic (poly(I:C)), a ligand of TLR3, acts synergistically to induce diabetes in 100% of KRV-infected rats. The mechanisms by which KRV induces diabetes and TLR3 ligation facilitates this process are not clear. In this study, we demonstrate that activation of the innate immune system plays a crucial role in diabetes induction. We report that multiple TLR agonists synergize with KRV infection to induce diabetes in BBDR rats, as do heat-killed Escherichia coli or Staphylococcus aureus (natural TLR agonists). KRV infection increases serum IL-12 p40 in a strain-specific manner, and increases IL-12 p40, IFN-γ-inducible protein-10, and IFN-γ mRNA transcript levels, particularly in the pancreatic lymph nodes of BBDR rats. Infection with vaccinia virus or H-1 parvovirus induced less stimulation of the innate immune system and failed to induce diabetes in BBDR rats. Our results suggest that the degree to which the innate immune system is activated by TLRs is important for expression of virus-induced diabetes in genetically susceptible hosts.


Journal of Immunology | 2003

Direct Visualization of Cross-Reactive Effector and Memory Allo-Specific CD8 T Cells Generated in Response to Viral Infections

Michael A. Brehm; Thomas G. Markees; Keith A. Daniels; Dale L. Greiner; Aldo A. Rossini; Raymond M. Welsh

CD8 T cell cross-reactivity between heterologous viruses has been shown to provide protective immunity, induce immunopathology, influence the immunodominance of epitope-specific T cell responses, and shape the overall memory population. Virus infections also induce cross-reactive allo-specific CTL responses. In this study, we quantified the allo-specific CD8 T cells elicited by infection of C57BL/6 (B6) mice with lymphocytic choriomeningitis virus (LCMV). Cross-reactive LCMV-specific CD8 T cells were directly visualized using LCMV peptide-charged MHC tetramers to costain T cells that were stimulated to produce intracellular IFN-γ in response to allogeneic target cells. The cross-reactivity between T cells specific for LCMV and allogeneic Ags was broad-based, in that it involved multiple LCMV-derived peptides, but there were distinctive patterns of reactivity against allogeneic cells with different haplotypes. Experiments indicated that this cross-reactivity was not due to the expression of two TCR per cell, and that the patterns of allo-reactivity changed during sequential infection with heterologous viruses. The allo-specific CD8 T cells generated by LCMV infection were maintained at relatively high frequencies in the memory pool, indicating that memory allo-specific CD8 T cell populations can arise as a consequence of viral infections. Mice previously infected with LCMV and harboring allo-specific memory T cells were refractory to the induction of tolerance to allogeneic skin grafts.


Journal of Virology | 2000

Virus-Induced Abrogation of Transplantation Tolerance Induced by Donor-Specific Transfusion and Anti-CD154 Antibody

Raymond M. Welsh; Thomas G. Markees; Bruce A. Woda; Keith A. Daniels; Michael A. Brehm; John P. Mordes; Dale L. Greiner; Aldo A. Rossini

ABSTRACT Treatment with a 2-week course of anti-CD154 antibody and a single transfusion of donor leukocytes (a donor-specific transfusion or DST) permits skin allografts to survive for >100 days in thymectomized mice. As clinical trials of this methodology in humans are contemplated, concern has been expressed that viral infection of graft recipients may disrupt tolerance to the allograft. We report that acute infection with lymphocytic choriomeningitis virus (LCMV) induced allograft rejection in mice treated with DST and anti-CD154 antibody if inoculated shortly after transplantation. Isografts resisted LCMV-induced rejection, and the interferon-inducing agent polyinosinic:polycytidylic acid did not induce allograft rejection, suggesting that the effect of LCMV is not simply a consequence of nonspecific inflammation. Administration of anti-CD8 antibody to engrafted mice delayed LCMV-induced allograft rejection. Pichinde virus also induced acute allograft rejection, but murine cytomegalovirus and vaccinia virus (VV) did not. Injection of LCMV ∼50 days after tolerance induction and transplantation had minimal effect on subsequent allograft survival. Treatment with DST and anti-CD154 antibody did not interfere with clearance of LCMV, but a normally nonlethal high dose of VV during tolerance induction and transplantation killed graft recipients. We conclude that DST and anti-CD154 antibody induce a tolerant state that can be broken shortly after transplantation by certain viral infections. Clinical application of transplantation tolerance protocols may require patient isolation to facilitate the procedure and to protect recipients.


Journal of Immunology | 2006

TLR Agonists Abrogate Costimulation Blockade-Induced Prolongation of Skin Allografts

Thomas B. Thornley; Michael A. Brehm; Thomas G. Markees; Leonard D. Shultz; John P. Mordes; Raymond M. Welsh; Aldo A. Rossini; Dale L. Greiner

Costimulation blockade protocols are effective in prolonging allograft survival in animal models and are entering clinical trials, but how environmental perturbants affect graft survival remains largely unstudied. We used a costimulation blockade protocol consisting of a donor-specific transfusion and anti-CD154 mAb to address this question. We observed that lymphocytic choriomeningitis virus infection at the time of donor-specific transfusion and anti-CD154 mAb shortens allograft survival. Lymphocytic choriomeningitis virus 1) activates innate immunity, 2) induces allo-cross-reactive T cells, and 3) generates virus-specific responses, all of which may adversely affect allograft survival. To investigate the role of innate immunity, mice given costimulation blockade and skin allografts were coinjected with TLR2 (Pam3Cys), TLR3 (polyinosinic:polycytidylic acid), TLR4 (LPS), or TLR9 (CpG) agonists. Costimulation blockade prolonged skin allograft survival that was shortened after coinjection by TLR agonists. To investigate underlying mechanisms, we used “synchimeric” mice which circulate trace populations of anti-H2b transgenic alloreactive CD8+ T cells. In synchimeric mice treated with costimulation blockade, coadministration of all four TLR agonists prevented deletion of alloreactive CD8+ T cells and shortened skin allograft survival. These alloreactive CD8+ T cells 1) expressed the proliferation marker Ki-67, 2) up-regulated CD44, and 3) failed to undergo apoptosis. B6.TNFR2−/− and B6.IL-12R−/− mice treated with costimulation blockade plus LPS also exhibited short skin allograft survival whereas similarly treated B6.CD8α−/− and TLR4−/− mice exhibited prolonged allograft survival. We conclude that TLR signaling abrogates the effects of costimulation blockade by preventing alloreactive CD8+ T cell apoptosis through a mechanism not dependent on TNFR2 or IL-12R signaling.

Collaboration


Dive into the Aldo A. Rossini's collaboration.

Top Co-Authors

Avatar

John P. Mordes

University of Massachusetts Boston

View shared research outputs
Top Co-Authors

Avatar

Dale L. Greiner

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Eugene S. Handler

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Leonard D. Shultz

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael C. Appel

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Rita Bortell

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Thomas G. Markees

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Arthur A. Like

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Nancy E. Phillips

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Todd Pearson

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge