22 research outputs found

    Preclinical models of acute liver failure: a comprehensive review

    No full text
    Acute liver failure is marked by the rapid deterioration of liver function in a previously well patient over period of days to weeks. Though relatively rare, it is associated with high morbidity and mortality. This makes it a challenging disease to study clinically, necessitating reliance on preclinical models as means to explore pathophysiology and novel therapies. Preclinical models of acute liver failure are artificial by nature, and generally fall into one of three categories: surgical, pharmacologic or immunogenic. This article reviews preclinical models of acute liver failure and considers their relevance in modeling clinical disease

    Ferroptosis-inducing agents compromise in vitro human islet viability and function

    Get PDF
    Human islet transplantation has been hampered by donor cell death associated with the islet preparation procedure before transplantation. Regulated necrosis pathways are biochemically and morphologically distinct from apoptosis. Recently, ferroptosis was identified as a non-apoptotic form of iron-dependent regulated necrosis implicated in various pathological conditions. Mediators of islet oxidative stress, including glutathione peroxidase-4 (GPX4), have been identified as inhibitors of ferroptosis, and mechanisms that affect GPX4 function can impact islet function and viability. Ferroptosis has not been investigated directly in human islets, and its relevance in islet transplantation remains unknown. Herein, we sought to determine whether in vitro human islet viability and function is compromised in the presence of two distinct ferroptosis-inducing agents (FIA), erastin or RSL3, and whether these effects could be rescued with ferroptosis inhibitors, ferrostatin-1 (Fer-1), or desferrioxamine (DFO). Viability, as assessed by lactate dehydrogenase (LDH) release, revealed significant death in erastin- and RSL3-treated islets, 20.3% ± 3.8 and 24.4% ± 2.5, 24 h post culture, respectively. These effects were ameliorated in islets pre-treated with Fer-1 or the iron chelator, desferrioxamine (DFO). Stimulation index, a marker of islet function revealed a significant reduction in function in erastin-treated islets (control 1.97 ± 0.13 vs. 50 μM erastin 1.32 ± 0.1) (p < 0.05). Fer-1 and DFO pre-treatment alone did not augment islet viability or function. Pre-treatment of islets with erastin or Fer-1 did not impact in vivo engraftment in an immunodeficient mouse transplant model. Our data reveal that islets are indeed susceptible to ferroptosis in vitro, and induction of this novel cell death modality leads to compromised islet function, which can be recoverable in the presence of the ferroptosis inhibitors. The in vivo impact of this pathway in islet transplantation remains elusive given the constraints of our study, but warrants continued investigation

    AEB071 (sotrastaurin) does not exhibit toxic effects on human islets in vitro, nor after transplantation into immunodeficient mice

    No full text
    AEB071 (AEB, sotrastaurin), a specific inhibitor of protein kinase C, reduces T-lymphocyte activation and cytokine release. AEB delays islet allograft rejection in rats and prevents rejection when combined with cyclosporine. Since many immunosuppressive agents have toxic effects on the function of transplanted islets, we investigated whether this was also the case with AEB. Human islets were transplanted into Rag-knockout mice randomly assigned to vehicle control, AEB or sirolimus treatment groups. Non-fasting blood glucose levels, body weight and glucose tolerance was measured in recipients. In a separate experiment, human islets were cultured in the presence of AEB and assayed for glucose dependent insulin secretion and level of β-cell apoptosis. Eighty-six percent of the AEB-treated recipients achieved normoglycemia following transplant (compared with none in sirolimus-treated group, p < 0.05). AEB-treated recipients exhibited similar glucose homeostasis as vehicle-treated controls, which was better than in sirolimus-treated recipients. Human islets cultured with AEB showed similar rates of β-cell apoptosis (p = 0.98 by one-way ANOVA) and glucose stimulated insulin secretion (p = 0.15) as those cultured with vehicle. These results suggest that AEB is not associated with toxic effects on islet engraftment or function. AEB appears to be an appropriate immunosuppressive candidate for clinical trials in islet transplantation

    Glutathione ethyl ester supplementation during pancreatic islet isolation improves viability and transplant outcomes in a murine marginal islet mass model.

    Get PDF
    The success of pancreatic islet transplantation still faces many challenges, mainly related to cell damage during islet isolation and early post-transplant. The increased generation of reactive oxygen species (ROS) during islet isolation and the consumption of antioxidant defenses appear to be an important pathway related to islet damage.In the present study we evaluated whether supplementation of glutathione-ethyl-ester (GEE) during islet isolation could improve islet viability and transplant outcomes in a murine marginal islet mass model. We also cultured human islets for 24 hours in standard CMRL media with or without GEE supplementation. Supplementation of GEE decreased the content of ROS in isolated islets, leading to a decrease in apoptosis and maintenance of islet viability. A higher percentage of mice transplanted with a marginal mass of GEE treated islets became euglycemic after transplant. The supplementation of 20 mM GEE in cultured human islets significantly reduced the apoptosis rate in comparison to untreated islets.GEE supplementation was able to decrease the apoptosis rate and intracellular content of ROS in isolated islets and might be considered a potential intervention to improve islet viability during the isolation process and maintenance in culture before islet transplantation

    Lung-Derived Microscaffolds Facilitate Diabetes Reversal after Mouse and Human Intraperitoneal Islet Transplantation

    No full text
    <div><p>There is a need to develop three-dimensional structures that mimic the natural islet tissue microenvironment. Endocrine micro-pancreata (EMPs) made up of acellular organ-derived micro-scaffolds seeded with human islets have been shown to express high levels of key beta-cell specific genes and secrete quantities of insulin per cell similar to freshly isolated human islets in a glucose-regulated manner for more than three months <i>in vitro</i>. The aim of this study was to investigate the capacity of EMPs to restore euglycemia <i>in vivo</i> after transplantation of mouse or human islets in chemically diabetic mice. We proposed that the organ-derived EMPs would restore the extracellular components of the islet microenvironment, generating favorable conditions for islet function and survival. EMPs seeded with 500 mouse islets were implanted intraperitoneally into streptozotocin-induced diabetic mice and reverted diabetes in 67% of mice compared to 13% of controls (p = 0.018, n = 9 per group). Histological analysis of the explanted grafts 60 days post-transplantation stained positive for insulin and exhibited increased vascular density in a collagen-rich background. EMPs were also seeded with human islets and transplanted into the peritoneal cavity of immune-deficient diabetic mice at 250 islet equivalents (IEQ), 500 IEQ and 1000 IEQ. Escalating islet dose increased rates of normoglycemia (50% of the 500 IEQ group and 75% of the 1000 IEQ group, n = 3 per group). Human c-peptide levels were detected 90 days post-transplantation in a dose-response relationship. Herein, we report reversal of diabetes in mice by intraperitoneal transplantation of human islet seeded on EMPs with a human islet dose as low as 500 IEQ.</p></div

    Protein kinase C inhibitor, AEB-071, acts complementarily with cyclosporine to prevent islet rejection in rats

    No full text
    BACKGROUND: AEB-071 (AEB) is a specific inhibitor of protein kinase C, which prevents T-lymphocyte activation. The present study investigated the effect of AEB on rat islet allotransplantation alone or in combination with CTLA4-Ig, mycophenolate mofetil, or cyclosporine A (CsA). METHODS: A rodent allogeneic islet transplant model (Lewis to Wistar Furth) was used to investigate the efficacy of AEB as an immunosuppressive agent. Furthermore, the Lewis rat was used to screen for any AEB associated toxicities on glucose homeostasis in vivo. RESULTS: AEB alone (30 mg/kg per os [p.o.] two times per day [bid]) delayed rejection to a median survival time of 22 days (vs. 7 days in control vehicle-treated animals, P<0.05). When combined with CsA (5 mg/kg p.o. bid), AEB prolonged survival from 12 (CsA alone) to over 100 days in 80% of animals (P<0.05). No delay in allograft rejection (above that resulting from AEB alone) was observed when AEB was combined with a sub-therapeutic dose of CTLA4-Ig or mycophenolate mofetil, nor low dose of CsA. The frequency of allospecific interferon-gamma-secreting splenocytes, assessed ex vivo by enzyme-linked immunosorbent spot (ELISPOT) assay, was lower in AEB-treated recipients compared with controls (P<0.05). AEB treatment did not alter the intraperitoneal glucose tolerance, the glucose-dependent insulin release, or the insulin content of the native pancreas. CONCLUSIONS: These data suggest that AEB is an appropriate immunosuppressive agent for islet transplantation, as it can prolong islet graft survival alone or in combination with CsA, without toxicity on glucose metabolism

    Islet viability <i>in vitro</i>.

    No full text
    <p>A decrease in islet damage was observed in different assays after islet isolation supplemented with 10 mM GEE. (A) Membrane integrity evaluated with Syto Green/Ethidium Bromide (P<0.05). (B) Islet apoptosis was measured with TUNEL staining after islet isolation. (P<0.05). (C, D, E and F) Fractional beta-cell viability staining using Newport Green, 7-aminoactinomycin D (7-AAD) and tetramethylrhodamine ethyl ester (TMRE) (P<0.05).</p

    Human islets after 24 h culture on different GEE concentrations.

    No full text
    <p>Intracelullar ROS was evaluated using carboxy-H<sub>2</sub>DCFDA and flow cytometry. Membrane integrity was evaluated by Syto green/Ethidium bromide. Fractional beta-cell viability was assessed using Newport Green, 7-AAD and TMRE.</p>*<p><i>p</i><0.05;</p>**<p><i>p</i><0.01;</p>***<p><i>p</i><0.001 in comparison to control.</p
    corecore