Network


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

Hotspot


Dive into the research topics where Rena Pawlick is active.

Publication


Featured researches published by Rena Pawlick.


American Journal of Transplantation | 2004

Multiple Combination Therapies Involving Blockade of ICOS/B7RP-1 Costimulation Facilitate Long-Term Islet Allograft Survival

Sulaiman A. Nanji; Wayne W. Hancock; Colin C. Anderson; Andrew B. Adams; Bin Luo; Colleen Schur; Rena Pawlick; Liqing Wang; Anthony J. Coyle; Christian P. Larsen; A. M. James Shapiro

In recent years a series of novel costimulatory molecules have been identified, including inducible costimulator (ICOS). In a fully major histocompatibility complex (MHC)‐mismatched mouse model of islet transplantation, we demonstrate that while monotherapy with CTLA4‐Ig, CD40 ligand monoclonal antibody (CD40L mAb) or rapamycin each improves islet allograft survival, graft rejection eventually develops. Immunohistologic analysis of rejected grafts revealed increased ICOS expression, suggesting a role for this costimulatory molecule as an alternate pathway for T‐cell activation. The combination of a blocking anti‐ICOS mAb with each of the above therapies resulted in significantly improved islet allograft survival, confirming the importance of ICOS signaling in islet allograft rejection. Mechanistic studies conducted in mice treated with anti‐ICOS mAb and rapamycin demonstrated a lack of donor‐specific immunological tolerance and an absence of regulatory T‐cell activity. However, a dramatic effect was seen on acute anti‐donor responses whereby anti‐ICOS mAb and rapamycin significantly reduced the initial expansion and function of alloreactive T cells. These data demonstrate that blockade of the ICOS/B7RP‐1 pathway has potential therapeutic benefit given its role in enhancing islet allograft survival and regulating acute alloresponses in vivo.


Transplant International | 2010

Liraglutide, a long-acting human glucagon-like peptide 1 analogue, improves human islet survival in culture

Christian Toso; Michael McCall; Juliet Emamaullee; Shaheed Merani; Joy Davis; Ryan Edgar; Rena Pawlick; Tatsuya Kin; Lotte Bjerre Knudsen; A. M. James Shapiro

The culture of human islets is associated with approximately 10–20% islet loss, occasionally preventing transplantation. Preconditioning of the islets to improve postculture yields would be of immediate benefit, with the potential to increase both the number of transplanted patients and their metabolic reserve. In this study, the effect of liraglutide, a long‐acting human glucagon‐like peptide 1 analogue, on cultured human islets was examined. Culture with liraglutide (1 μmol/l) was associated with a preservation of islet mass (significantly more islets at 24 and 48 h, compared to control; P ≤ 0.05 at 24 and 48 h) and with the presence of larger islets (P ≤ 0.05 at 48 h). These observations were supported by reduced apoptosis rates  after 24 h of treatment. We also demonstrated that human islet engraftment is improved in C57Bl/6‐RAG−/− mice treated with liraglutide 200 μg/kg sc twice daily (P ≤ 0.05), suggesting that liraglutide should be continued after transplantation. Overall, these data demonstrate the beneficial effect of liraglutide on cultured human islets, preserving islet mass. They support the design of clinical studies looking at the effect of liraglutide in clinical islet transplantation.


Diabetes | 2008

The caspase selective inhibitor EP1013 augments human islet graft function and longevity in marginal mass islet transplantation in mice.

Juliet Emamaullee; Joy Davis; Rena Pawlick; Christian Toso; Shaheed Merani; Sui-Xiong Cai; Ben Tseng; A. M. James Shapiro

OBJECTIVE—Clinical islet transplantation can provide insulin independence in patients with type 1 diabetes, but chronic graft failure has been observed. This has been attributed in part to loss of ≥60% of the transplanted islets in the peritransplant period, resulting in a marginal implant mass. Strategies designed to maximize survival of the initial islet mass are likely to have major impact in enhancing long-term clinical outcomes. EP1013 (N-benzyloxycabonyl-Val Asp-fluoromethyl ketone [zVD-FMK]), is a broad-spectrum caspase selective inhibitor with no observed toxicity in rodents. RESEARCH DESIGN AND METHODS—The therapeutic benefit of EP1013 was examined in a syngeneic rodent islet transplant model using deceased donor human islets to determine whether the amount of tissue required to restore euglycemia in diabetic animals could be reduced. RESULTS—EP1013 (combined pretransplant islet culture for 2 h and in vivo treatment for days 0–5 posttransplant) significantly improved marginal islet mass function following syngeneic islet transplantation in mice, even at lower doses, compared with previous studies using the pan-caspase inhibitor N-benzyloxycabonyl-Val Ala-Asp-fluoromethyl ketone (zVAD-FMK). EP1013 supplementation in vitro improved human islet yields following prolonged culture and reversed diabetes following implantation of a marginal human islet mass (80–90% reduction) into mice. CONCLUSIONS—Our data suggest that EP1013 therapy will markedly reduce the islet mass required in clinical islet transplantation, improving insulin independence rates following single-donor infusion.


American Journal of Transplantation | 2012

Anakinra Potentiates the Protective Effects of Etanercept in Transplantation of Marginal Mass Human Islets in Immunodeficient Mice

Michael McCall; Rena Pawlick; Tatsuya Kin; A. M. J. Shapiro

Anti‐inflammatory agents are used routinely in clinical islet transplantation in an attempt to promote islet engraftment. Infliximab, and more recently etanercept, is being used to neutralize tumor necrosis factor alpha, but this tenet is based on limited preclinical data. One group has promoted the potential of combined etanercept with an IL‐1 receptor antagonist, anakinra in a small clinical study, but without strong preclinical data to justify this approach. We therefore sought to evaluate the impact of combined anakinra and etanercept in a marginal islet mass transplant model using human islets in immunodeficient mice. The combination of anakinra and etanercept led to remarkable improvement in islet engraftment (control 36.4%; anakinra 53.9%; etanercept 45.45%; anakinra and etanercept 87.5% euglycemia, p < 0.05 by log‐rank) compared to single‐drug treated mice or controls. This translated into enhanced metabolic function (area under curve glucose tolerance), improved graft insulin content and marked reduction in beta‐cell specific apoptotis (0.67% anakinra + etanercept vs. 23.5% control, p < 0.001). These results therefore strongly justify the combined short‐term use of anakinra and etanercept in human islet transplantation.


Transplantation | 2010

Comparison of Human Islet Isolation Outcomes Using a New Mammalian Tissue-Free Enzyme Versus Collagenase NB-1

Doug OʼGorman; Tatsuya Kin; Sharleen Imes; Rena Pawlick; Peter A. Senior; A. M. James Shapiro

Background. After the discontinuation of the manufacturing Liberase HI because of a small potential for prion disease transmission, Roche Diagnostics (Indianapolis, IN) developed a new enzyme product (Liberase MTF [mammalian tissue free]), which is similar to Liberase HI with the exception that no mammalian tissue is used in the manufacture of the collagenase component. We report our experience using the MTF enzyme in clinical islet isolations compared with Serva NB-1 with modified enzyme delivery method. Methods. Islets were isolated from 41 pancreata using MTF enzyme (n=17) or NB-1 enzyme (n=24). NB-1 enzymes were delivered using a modified (nonsimultaneous) enzyme delivery method whereas isolations using MTF used the standard method of simultaneous collagenase and thermolysin perfusion. Islets were purified on a COBE 2991 Cell Blood Processor and subsequently cultured. Results. The average islet mass after purification was 392±36×103 islet equivalent (IE) for MTF versus 371±40×103 IE for Serva NB-1 (P=0.63). Post-IE/cm3 of tissue was 110±9×103 IE/cm3 and 91±11×103 IE/cm3 for MTF and NB-1, respectively (P=0.07). The isolation success rate (>400,000 IE) for MTF was 53% compared with 33% for Serva (P=0.33). Conclusion. We conclude that MTF may be successfully used for high-yield human islet isolation and clinical transplantation and provides similar quality islets to those derived using NB-1.


Transplant International | 2009

Effect of different induction strategies on effector, regulatory and memory lymphocyte sub-populations in clinical islet transplantation

Christian Toso; Ryan Edgar; Rena Pawlick; Juliet Emamaullee; Shaheed Merani; Parastoo Dinyari; Thomas F. Mueller; A. M. James Shapiro; Colin C. Anderson

This prospective study assessed lymphocyte subsets in the peripheral blood of 42 islet allograft recipients using flow cytometry from 2 weeks and up to 2 years post‐transplantation. Subjects received daclizumab (n = 16), Thymoglobulin (n = 12) or alemtuzumab (n = 14). Alemtuzumab was associated with an early (within 1 month) and transient (up to 6 months) increase in the frequency of CD3+ CD4+ Foxp3+ T cells, while daclizumab induced a near complete loss of these cells (P ≤ 0.001). The frequency of memory CD4+ T cells was increased following depleting immunosuppression induction with either Thymoglobulin or alemtuzumab (P ≤ 0.05), but remained unchanged while using daclizumab. Alemtuzumab induction resulted in a significant loss of memory B lymphocytes when compared with the other induction groups (P ≤ 0.001). While the clinical significance of these findings remains to be fully determined, the observed altered balance between effector, regulatory and memory cells suggests that the immune status of patients will be affected according to the induction strategy chosen.


Transplantation | 2015

Diabetes Is Reversed in a Murine Model by Marginal Mass Syngeneic Islet Transplantation Using a Subcutaneous Cell Pouch Device

Andrew R. Pepper; Rena Pawlick; Boris Gala-Lopez; Amanda MacGillivary; Delfina M. Mazzuca; D. J. G. White; Philip M. Toleikis; A. M. James Shapiro

Background Islet transplantation is a successful &bgr;-cell replacement therapy for selected patients with type 1 diabetes mellitus. Although high rates of early insulin independence are achieved routinely, long-term function wanes over time. Intraportal transplantation is associated with procedural risks, requires multiple donors, and does not afford routine biopsy. Stem cell technologies may require potential for retrievability, and graft removal by hepatectomy is impractical. There is a clear clinical need for an alternative, optimized transplantation site. The subcutaneous space is a potential substitute, but transplantation of islets into this site has routinely failed to reverse diabetes. However, an implanted device, which becomes prevascularized before transplantation, may alter this equation. Methods Syngeneic mouse islets were transplanted subcutaneously within Sernova Corps Cell Pouch (CP). All recipients were preimplanted with CPs 4 weeks before diabetes induction and transplantation. After transplantation, recipients were monitored for glycemic control and glucose tolerance. Results Mouse islets transplanted into the CP routinely restored glycemic control with modest delay and responded well to glucose challenge, comparable to renal subcapsular islet grafts, despite a marginal islet dose, and normoglycemia was maintained until graft explantation. In contrast, islets transplanted subcutaneously alone failed to engraft. Islets within CPs stained positively for insulin, glucagon, and microvessels. Conclusions The CP is biocompatible, forms an environment suitable for islet engraftment, and offers a potential alternative to the intraportal site for islet and future stem cell therapies.


Endocrinology | 2009

Porcine Marginal Mass Islet Autografts Resist Metabolic Failure Over Time and Are Enhanced by Early Treatment with Liraglutide

Juliet Emamaullee; Shaheed Merani; Christian Toso; Tatsuya Kin; Faisal Al-Saif; Wayne Truong; Rena Pawlick; Joy Davis; Ryan Edgar; Jennifer Lock; Susan Bonner-Weir; Lotte Bjerre Knudsen; A. M. James Shapiro

Although insulin independence is maintained in most islet recipients at 1 yr after transplant, extended follow-up has revealed that many patients will eventually require insulin therapy. Previous studies have shown that islet autografts are prone to chronic failure in large animals and humans, suggesting that nonimmunological events contribute to islet graft functional decay. Early intervention with therapies that promote graft stability should provide a measurable benefit over time. In this study, the efficacy of the long-acting glucagon-like peptide-1 analog liraglutide was explored in a porcine marginal mass islet autograft transplant model. Incubation with liraglutide enhanced porcine islet survival and function after prolonged culture. Most vehicle-treated (83%) and liraglutide-treated (80%) animals became insulin independent after islet autotransplantation. Although liraglutide therapy did not improve insulin independence rates or blood glucose levels after transplant, a significant increase in insulin secretion and acute-phase insulin response was observed in treated animals. Surprisingly, no evidence for deterioration of graft function was observed in any of the transplanted animals over more than 18 months of follow-up despite significant weight gain; in fact, an enhanced response to glucose developed over time even in control animals. Histological analysis showed that intraportally transplanted islets remained highly insulin positive, retained alpha-cells, and did not form amyloid deposits. This study demonstrates that marginal mass porcine islet autografts have stable long-term function, even in the presence of an increasing metabolic demand. These results are discrepant with previous large animal studies and suggest that porcine islets may be resistant to metabolic failure.


Artificial Organs | 2008

The Use of an Approved Biodegradable Polymer Scaffold as a Solid Support System for Improvement of Islet Engraftment

Tatsuya Kin; John J. O'neil; Rena Pawlick; Gregory S. Korbutt; A. M. James Shapiro; Jonathan R. T. Lakey

The aim of this study was to investigate whether the use of a medically approved biodegradable scaffold as a solid support system would enhance graft survival following transplantation into the omental pouch in a preclinical large animal model. Six beagle dogs underwent total pancreatectomy followed by islet autotransplantation into the omental pouch. Four dogs received islets seeded in a biodegradable polymer scaffold and two received free islets without a scaffold. All four animals that received islets in the scaffold became normoglycemic without exogeneous insulin injection. One dog, transplanted with the largest number of islets, maintained a normal metabolic state until the graft was removed at 5 months posttransplant. In two out of the three that received a marginal islet mass, insulin independence was sustained up to 2 months. In contrast, two dogs transplanted with a similar marginal mass without the scaffold never became normoglycemic. Histological examination of the grafts in the scaffold showed numerous well-granulated, insulin-containing cells as well as glucagon-positive cells. These results indicate that biodegradable scaffolds may enhance survival and function of islet grafts. Manipulation of the microenvironment of transplanted islets may constitute the basis for new approaches to enhance islet engraftment.


Surgery | 2011

The caspase inhibitor IDN-6556 (PF3491390) improves marginal mass engraftment after islet transplantation in mice

Michael McCall; Christian Toso; Juliet Emamaullee; Rena Pawlick; Ryan Edgar; Joy Davis; Allison H. Maciver; Tatsuya Kin; Robert H. Arch; A. M. James Shapiro

BACKGROUND Islet transplantation has become a viable option for selected type 1 diabetic patients; however, a significant portion need to return to exogenous insulin. The predominant factors include impaired islet engraftment and early islet loss. Caspase inhibition is a potent way to improve islet engraftment, but all tested compounds so far have not been clinically relevant. IDN-6556 (PF3491390) has already been used clinically and can be delivered orally with high portal vein concentrations. METHODS Mice were given a marginal mass islet graft of either mouse or human islets and treated with either IDN-6556 (10 or 20 mg/kg ip bid) or vehicle and followed for diabetes reversal. At 1 month post-transplant, mice were subjected to a glucose tolerance test and an assessment of graft mass. In separate experiments, human islets were cultured with IDN-6556 or vehicle to assess for islet survival and viability. RESULTS In both syngeneic mouse islets and human islets transplanted into immunodeficient mice, IDN-6556 (20 mg/kg) given for 7 days post-transplant led to a significantly enhanced rate of diabetes reversal as compared to vehicle. In addition, mice receiving caspase inhibitor displayed improved glucose tolerance and graft survival at the 1-month point. We also found protective effects in vitro for islet viability and marked reduction in apoptosis in vivo. CONCLUSION Taken together, these results demonstrate the effectiveness of caspase inhibition with IDN-6556 on islet transplantation and in particular islet engraftment and survival.

Collaboration


Dive into the Rena Pawlick's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge