Network


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

Hotspot


Dive into the research topics where Andi Qipo is active.

Publication


Featured researches published by Andi Qipo.


Journal of Experimental Medicine | 2006

Tissue expression of PD-L1 mediates peripheral T cell tolerance

Mary E. Keir; Spencer C. Liang; Indira Guleria; Yvette Latchman; Andi Qipo; Lee A. Albacker; Maria Koulmanda; Gordon J. Freeman; Mohamed H. Sayegh; Arlene H. Sharpe

Programmed death 1 (PD-1), an inhibitory receptor expressed on activated lymphocytes, regulates tolerance and autoimmunity. PD-1 has two ligands: PD-1 ligand 1 (PD-L1), which is expressed broadly on hematopoietic and parenchymal cells, including pancreatic islet cells; and PD-L2, which is restricted to macrophages and dendritic cells. To investigate whether PD-L1 and PD-L2 have synergistic or unique roles in regulating T cell activation and tolerance, we generated mice lacking PD-L1 and PD-L2 (PD-L1/PD-L2−/− mice) and compared them to mice lacking either PD-L. PD-L1 and PD-L2 have overlapping functions in inhibiting interleukin-2 and interferon-γ production during T cell activation. However, PD-L1 has a unique and critical role in controlling self-reactive T cells in the pancreas. Our studies with bone marrow chimeras demonstrate that PD-L1/PD-L2 expression only on antigen-presenting cells is insufficient to prevent the early onset diabetes that develops in PD-L1/PD-L2−/− non-obese diabetic mice. PD-L1 expression in islets protects against immunopathology after transplantation of syngeneic islets into diabetic recipients. PD-L1 inhibits pathogenic self-reactive CD4+ T cell–mediated tissue destruction and effector cytokine production. These data provide evidence that PD-L1 expression on parenchymal cells rather than hematopoietic cells protects against autoimmune diabetes and point to a novel role for PD-1–PD-L1 interactions in mediating tissue tolerance.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Curative and β cell regenerative effects of α1-antitrypsin treatment in autoimmune diabetic NOD mice

Maria Koulmanda; Manoj Bhasin; Lauren Hoffman; Zhigang Fan; Andi Qipo; Hang Shi; Susan Bonner-Weir; Prabhakar Putheti; Nicolas Degauque; Towia A. Libermann; Hugh Auchincloss; Jeffrey S. Flier; Terry B. Strom

Invasive insulitis is a destructive T cell-dependent autoimmune process directed against insulin-producing β cells that is central to the pathogenesis of type 1 diabetes mellitus (T1DM) in humans and the clinically relevant nonobese diabetic (NOD) mouse model. Few therapies have succeeded in restoring long-term, drug-free euglycemia and immune tolerance to β cells in overtly diabetic NOD mice, and none have demonstrably enabled enlargement of the functional β cell mass. Recent studies have emphasized the impact of inflammatory cytokines on the commitment of antigen-activated T cells to various effector or regulatory T cell phenotypes and insulin resistance and defective insulin signaling. Hence, we tested the hypothesis that inflammatory mechanisms trigger insulitis, insulin resistance, faulty insulin signaling, and the loss of immune tolerance to islets. We demonstrate that treatment with α1-antitrypsin (AAT), an agent that dampens inflammation, does not directly inhibit T cell activation, ablates invasive insulitis, and restores euglycemia, immune tolerance to β cells, normal insulin signaling, and insulin responsiveness in NOD mice with recent-onset T1DM through favorable changes in the inflammation milieu. Indeed, the functional mass of β cells expands in AAT-treated diabetic NOD mice.


American Journal of Transplantation | 2003

The Effect of Low Versus High Dose of Streptozotocin in Cynomolgus Monkeys (Macaca Fascilularis)

Maria Koulmanda; Andi Qipo; S. Chebrolu; John J. O'Neil; Hugh Auchincloss; R. N. Smith

Streptozotocin (STZ) is often used to induce diabetes in animal models. However, morbidity associated with STZ and its ability to induce diabetes vary with different dosages among different animal species, including nonhuman primates. To find an optimal dose of STZ that would cause diabetes with minimal toxicity, we compared low and high doses of STZ. Male cynomolgus monkeys (3–6 years old) were given a single dose of 100 mg/kg (high dose, 4 animals) or 55 mg/kg (low dose, 20 animals) of STZ. Blood glucose levels, intravenous glucose tolerance test (IVGTT), pancreatic biopsies, liver function tests (LFTs), liver biopsies, kidney function tests, and kidney biopsies were performed periodically. Animals from both groups developed diabetes within 24 h after administration of STZ. Serum C‐peptide levels in both groups decreased from 2 to 8 ng/mL before STZ to between 0.01 and 0.6 ng/mL after STZ. Animals with the high dose of STZ developed transient vomiting within minutes after injection. During the first week after STZ injection, high‐dose animals developed elevated LFTs, BUN and creatinine. In contrast, low‐dose animals had normal liver and kidney function tests. Histological analysis showed that animals given the high dose of STZ developed marked steatosis of the liver and tubular injury in the kidneys, whereas animals given the low dose of STZ had normal‐looking liver and kidney histology. The pancreatic islets in both groups were indistinguishable by immunoperoxidase staining for insulin, and showed either no insulin‐positive cells or rare insulin‐positive cells. Glucagon staining was normal. Over time, low‐dose diabetic monkeys remained persistently hyperglycemic with negligible C‐peptide stimulation by intravenous glucose. We conclude that low‐dose STZ at 55 mg/mL successfully induces diabetes in cynomolgus monkeys with minimal liver and kidney toxicity.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Modification of adverse inflammation is required to cure new-onset type 1 diabetic hosts

Maria Koulmanda; Ejona Budo; Susan Bonner-Weir; Andi Qipo; Prabhakar Putheti; Nicolas Degauque; Hang Shi; Zhigang Fan; Jeffrey S. Flier; Hugh Auchincloss; Xin Xiao Zheng; Terry B. Strom

In nonobese diabetic (NOD) mice with overt new-onset type 1 diabetes mellitus (T1DM), short-term treatment with a “triple-therapy” regimen [rapamycin plus agonist IL-2-related and antagonist-type, mutant IL-15-related Ig fusion proteins (IL-2.Ig and mutIL-15.Ig)] halts autoimmune destruction of insulin-producing beta cells and restores both euglycemia and immune tolerance to beta cells. Increases in the mass of insulin-producing beta cells or circulating insulin levels were not linked to the restoration of euglycemia. Instead, the restoration of euglycemia was linked to relief from an inflammatory state that impaired the hosts response to insulin. Both restoration of immune tolerance to beta cells and relief from the adverse metabolic effects of an inflammatory state in insulin-sensitive tissues appear essential for permanent restoration of normoglycemia in this T1DM model. Thus, this triple-therapy regimen, possessing both tolerance-inducing and select antiinflammatory properties, may represent a prototype for therapies able to restore euglycemia and self-tolerance in T1DM.


Journal of Investigative Surgery | 2010

Prediction of Marginal Mass Required for Successful Islet Transplantation

Klearchos K. Papas; Clark K. Colton; Andi Qipo; Haiyan Wu; Rebecca A. Nelson; Bernhard J. Hering; Gordon C. Weir; Maria Koulmanda

ABSTRACT Islet quality assessment methods for predicting diabetes reversal (DR) following transplantation are needed. We investigated two islet parameters, oxygen consumption rate (OCR) and OCR per DNA content, to predict transplantation outcome and explored the impact of islet quality on marginal islet mass for DR. Outcomes in immunosuppressed diabetic mice were evaluated by transplanting mixtures of healthy and purposely damaged rat islets for systematic variation of OCR/DNA over a wide range. The probability of DR increased with increasing transplanted OCR and OCR/DNA. On coordinates of OCR versus OCR/DNA, data fell into regions in which DR occurred in all, some, or none of the animals with a sharp threshold of around 150-nmol/min mg DNA. A model incorporating both parameters predicted transplantation outcome with sensitivity and specificity of 93%% and 94%%, respectively. Marginal mass was not constant, depended on OCR/DNA, and increased from 2,800 to over 100,000 islet equivalents/kg body weight as OCR/DNA decreased. We conclude that measurements of OCR and OCR/DNA are useful for predicting transplantation outcome in this model system, and OCR/DNA can be used to estimate the marginal mass required for reversing diabetes. Because human clinical islet preparations in a previous study had OCR/DNA


American Journal of Transplantation | 2006

Prolonged Survival of Allogeneic Islets in Cynomolgus Monkeys After Short-Term Anti-CD154-Based Therapy: Nonimmunologic Graft Failure?

Maria Koulmanda; R. N. Smith; Andi Qipo; Gordon C. Weir; Hugh Auchincloss; Terry B. Strom

Conventional drug therapy and several anti‐CD154 mAb‐based regimens were tested in the nonhuman primate (NHP) islet allograft model and found to be inadequate because islets were lost to rejection. Short‐term therapy with an optimized donor‐specific transfusion (DST) + rapamycin (RPM) + anti‐CD154 mAb regimen enables immunosuppression drug‐free islet allograft function for months following cessation of therapy in the NHP islet allograft model. After a substantial period of drug‐free graft function, these allografts slowly and progressively lost function. Pathologic studies failed to identify islet allograft rejection as a destructive islet invasive lymphocytic infiltration of the allograft was not detected. To evaluate the mechanism, immunologic versus nonimmunologic, of the late islet allograft loss in hosts receiving the optimized therapeutic regimen, we performed experiments with islet autografts and studied islet function in NHPs with partial pancreatectomy. The results in both experiments utilizing autologous islet allografts and partially pancreatectomized hosts reinforce the view that the presence of a marginal islet mass leads to slowly progressive nonimmunological islet loss. Long‐term clinically successful islet cell transplantation cannot be realized in the absence of parallel improvements in tolerizing regimens and in the preparation of adequate numbers of islets.


Xenotransplantation | 2003

Pig islet xenografts are resistant to autoimmune destruction by non‐obese diabetic recipients after anti‐CD4 treatment

Maria Koulmanda; Andi Qipo; R. Neal Smith; Hugh Auchincloss

Abstract: In addition to providing a large source of donor tissue, xenogeneic islet transplantation might avoid recurrent autoimmunity in patients with type 1 diabetes. To examine this possibility further, xenogeneic pig islets were transplanted into recipient mice in the presence or absence of autoimmunity. Spontaneously, non‐obese diabetic (NOD) recipients rejected isografts rapidly whether or not the recipients were depleted of CD4+ T‐cells. Young NOD mice made diabetic with streptozotocin accepted islet isografts without immunosuppression, indicating that destructive autoimmunity did not develop in these recipients. Pig xenografts were rejected equally quickly in the two types of NOD recipients in the absence of immunosuppression and survived for up to 9 weeks in both types of NOD recipients after CD4 depletion. BALB/c mice often accepted pig xenografts indefinitely after anti‐CD4 antibody treatment. These results suggest that pig islets are resistant to recurrent autoimmunity when CD4+ T‐cells are depleted. The difficulty in obtaining indefinite islet xenograft survival in NOD recipients occurs independently from the development of destructive autoimmunity.


PLOS ONE | 2012

The Role of TNF-α in Mice with Type 1- and 2- Diabetes

Maria Koulmanda; Manoj Bhasin; Zuheir Awdeh; Andi Qipo; Zhigang Fan; Dusan Hanidziar; Prabhakar Putheti; Hang Shi; Eva Csizuadia; Towia A. Libermann; Terry B. Strom

Background Previously, we have demonstrated that short-term treatment of new onset diabetic Non-obese diabetic (NOD) mice, mice that are afflicted with both type 1 (T1D) and type 2 (T2D) diabetes with either Power Mix (PM) regimen or alpha1 antitrypsin (AAT) permanently restores euglycemia, immune tolerance to self-islets and normal insulin signaling. Methodology and Principal Findings To search for relevant therapeutic targets, we have applied genome wide transcriptional profiling and systems biology oriented bioinformatics analysis to examine the impact of the PM and AAT regimens upon pancreatic lymph node (PLN) and fat, a crucial tissue for insulin dependent glucose disposal, in new onset diabetic non-obese diabetic (NOD) mice. Systems biology analysis identified tumor necrosis factor alpha (TNF-α) as the top focus gene hub, as determined by the highest degree of connectivity, in both tissues. In PLNs and fat, TNF-α interacted with 53% and 32% of genes, respectively, associated with reversal of diabetes by previous treatments and was thereby selected as a therapeutic target. Short-term anti-TNF-α treatment ablated a T cell-rich islet-invasive and beta cell-destructive process, thereby enhancing beta cell viability. Indeed anti-TNF-α treatment induces immune tolerance selective to syngeneic beta cells. In addition to these curative effects on T1D anti-TNF-α treatment restored in vivo insulin signaling resulting in restoration of insulin sensitivity. Conclusions In short, our molecular analysis suggested that PM and AAT both may act in part by quenching a detrimental TNF-α dependent effect in both fat and PLNs. Indeed, short-term anti-TNF-α mAb treatment restored enduring euglycemia, self-tolerance, and normal insulin signaling.


Clinical and Experimental Immunology | 2003

Effects of streptozotocin on autoimmune diabetes in NOD mice

Maria Koulmanda; Andi Qipo; Hugh Auchincloss; R. N. Smith

Non‐obese diabetic (NOD) mice develop autoimmunity that destroys their native beta cells causing diabetes. Their autoimmunity will also destroy syngeneic transplanted islets and transfer both autoimmunity and diabetes via spleen cells to non‐diabetic mice. In this report, we studied the effects of streptozotocin (STZ) on the autoimmune diabetes in NOD mice. We transplanted NOD.SCID islets into three groups of NOD mice: (1) spontaneously diabetic NOD mice (NOD‐sp.); (2) prediabetic NOD mice made diabetic by streptozotocin (NOD‐stz); and (3) diabetic NOD mice also treated with streptozotocin (NOD‐sp./stz). In the first group, the transplants were rejected within 3 weeks. In the second and third groups, the transplants survived indefinitely. Alloxan, a drug similar to streptozotocin, did not have the same effect as streptozotocin. The ability of streptozotocin to prevent diabetes in young NOD mice was reversed by anti‐CD8 antibody treatment but not by anti‐CD4 treatment. Streptozotocin also made spleen cells from diabetic NOD mice less effective transferring diabetes. These results indicate that streptozotocin treatment both prevents and reverses the islet destructive autoimmunity in NOD mice. We postulate that the effects of streptozotocin treatment may be mediated in part by regulatory T cells.


American Journal of Transplantation | 2012

Prolonged Survival of Allogeneic Islets in Cynomolgus Monkeys After Short-Term Triple Therapy

Maria Koulmanda; Andi Qipo; Z. Fan; N. Smith; Hugh Auchincloss; Xin Xiao Zheng; Terry B. Strom

Preclinical studies in nonhuman primates (NHP) are particularly useful to evaluate the safety and efficacy of new therapeutic proteins developed for use in clinical transplantation. We hypothesized that a treatment that selectively destroys activated cytopathic donor reactive T cells while sparing resting and immunoregulatory T cells in a mouse model might also produce long‐term drug‐free engraftment and tolerance without the hazards of lymphopenia in the challenging nonhuman primate islet allograft model. Short‐term treatment with a regimen consisting of rapamycin, and IL‐2.Ig plus mutant antagonist‐type IL‐15.Ig cytolytic fusion proteins (triple therapy) posttransplantation results in prolonged, drug‐free engraftment of cynomolgus islet allografts. Moreover slow progressive loss of islet function in some recipients was not associated with obvious pathologic evidence of rejection.

Collaboration


Dive into the Andi Qipo's collaboration.

Top Co-Authors

Avatar

Maria Koulmanda

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terry B. Strom

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Zhigang Fan

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hang Shi

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Manoj Bhasin

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Prabhakar Putheti

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey S. Flier

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge