Adoptive immunotherapy with regulatory T cells (Tregs) is a promising treatment for allograft rejection and graft-versus-host disease (GVHD). Emerging data indicate that, compared with polyclonal Tregs, disease-relevant antigen-specific Tregs may have numerous advantages, such as a need for fewer cells and reduced risk of nonspecific immune suppression. Current methods to generate alloantigen-specific Tregs rely on expansion with allogeneic antigen-presenting cells, which requires access to donor and recipient cells and multiple MHC mismatches. The successful use of chimeric antigen receptors (CARs) for the generation of antigen-specific effector T cells suggests that a similar approach could be used to generate alloantigen-specific Tregs. Here, we have described the creation of an HLA-A2-specific CAR (A2-CAR) and its application in the generation of alloantigen-specific human Tregs. In vitro, A2-CAR-expressing Tregs maintained their expected phenotype and suppressive function before, during, and after A2-CAR-mediated stimulation. In mouse models, human A2-CAR-expressing Tregs were superior to Tregs expressing an irrelevant CAR at preventing xenogeneic GVHD caused by HLA-A2+ T cells. Together, our results demonstrate that use of CAR technology to generate potent, functional, and stable alloantigen-specific human Tregs markedly enhances their therapeutic potential in transplantation and sets the stage for using this approach for making antigen-specific Tregs for therapy of multiple diseases.
OBJECTIVE-Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of diabetes, but the roles of specific ER Ca 2ϩ release channels in the ER stress-associated apoptosis pathway remain unknown. Here, we examined the effects of stimulating or inhibiting the ER-resident inositol trisphosphate receptors (IP 3 Rs) and the ryanodine receptors (RyRs) on the induction of -cell ER stress and apoptosis.RESEARCH DESIGN AND METHODS-Kinetics of -cell death were tracked by imaging propidium iodide incorporation and caspase-3 activity in real time. ER stress and apoptosis were assessed by Western blot. Mitochondrial membrane potential was monitored by flow cytometry. Cytosolic Ca 2ϩ was imaged using fura-2, and genetically encoded fluorescence resonance energy transfer (FRET)-based probes were used to measure Ca 2ϩ in ER and mitochondria. RESULTS-NeitherRyR nor IP 3 R inhibition, alone or in combination, caused robust death within 24 h. In contrast, blocking sarco/endoplasmic reticulum ATPase (SERCA) pumps depleted ER Ca 2ϩ and induced marked phosphorylation of PKR-like ER kinase (PERK) and eukaryotic initiation factor-2␣ (eIF2␣), C/EBP homologous protein (CHOP)-associated ER stress, caspase-3 activation, and death. Notably, ER stress following SERCA inhibition was attenuated by blocking IP 3 Rs and RyRs. Conversely, stimulation of ER Ca 2ϩ release channels accelerated thapsigargin-induced ER depletion and apoptosis. SERCA block also activated caspase-9 and induced perturbations of the mitochondrial membrane potential, resulting eventually in the loss of mitochondrial polarization.CONCLUSIONS-This study demonstrates that the activity of ER Ca 2ϩ channels regulates the susceptibility of -cells to ER stress resulting from impaired SERCA function. Our results also suggest the involvement of mitochondria in -cell apoptosis associated with dysfunctional -cell ER Ca 2ϩ homeostasis and ER stress. Diabetes 58:422-432, 2009
Obesity is a principal risk factor for type 2 diabetes, and elevated fatty acids reduce -cell function and survival. An unbiased proteomic screen was used to identify targets of palmitate in -cell death. The most significantly altered protein in both human islets and MIN6 -cells treated with palmitate was carboxypeptidase E (CPE). Palmitate reduced CPE protein levels within 2 h, preceding endoplasmic reticulum (ER) stress and cell death, by a mechanism involving CPE translocation to Golgi and lysosomal degradation. Palmitate metabolism and Ca 2؉ flux were also required for CPE proteolysis and -cell death. Chronic palmitate exposure increased the ratio of proinsulin to insulin. CPE null islets had increased apoptosis in vivo and in vitro. Reducing CPE by Ϸ30% using shRNA also increased ER stress and apoptosis. Conversely, overexpression of CPE partially rescued -cells from palmitate-induced ER stress and apoptosis. Thus, carboxypeptidase E degradation contributes to palmitate-induced -cell ER stress and apoptosis. CPE is a major link between hyperlipidemia and -cell death pathways in diabetes.2D difference gel electrophoresis proteomics ͉ free fatty acids ͉ hyperproinsulinemia ͉ mechanisms of -cell lipotoxicity ͉ type 2 diabetes T here is a strong association between type 2 diabetes and obesity.High levels of circulating lipids, including free fatty acids, are a prominent clinical feature of type 2 diabetes and represent an important risk factor for this disease (1, 2). But exactly how elevated lipids might lead to diabetes remains unresolved. Fatty acids increase basal insulin secretion (3) and the relative levels of circulating proinsulin (4). Chronic exposure to the free fatty acid palmitate has been shown to impair glucose-stimulated insulin release (i.e., lipotoxicity) (5-10). -Cell apoptosis can be initiated by high levels of palmitate (6,7,(11)(12)(13)(14), which may account in part for alterations in insulin secretory function (13). A number of studies have established palmitate targets in the -cell, including lipid metabolism (15, 16), mitochondrial function (17-23), and prosurvival transcription factors such as Pdx1 (24,25). Recently, a role for endoplasmic reticulum (ER) stress in lipotoxicity has been demonstrated in multiple cell types, including -cells (11,26,27). The effects of palmitate on -cell survival are likely mediated by a number of mechanisms.In the present study, we conducted unbiased proteomic screens using human islets and MIN6 -cells to elucidate targets of palmitate. Carboxypeptidase E (CPE) was the most significantly changed protein in both screens. Mice lacking CPE develop hyperproinsulinemia and hyperglycemia (28), but the involvement of this protein in -cell apoptosis has not been reported. Palmitate caused the rapid intracellular redistribution and degradation of CPE via mechanisms that required palmitate metabolism, K ATP channel closure, Ca 2ϩ influx, and protease activity. We further showed that CPE levels control -cell ER stress and apoptosis. Thus, CPE is a cri...
Most of the adenosine triphosphate (ATP) synthesized during glucose metabolism is produced in the mitochondria through oxidative phosphorylation. This is a complex reaction powered by the proton gradient across the mitochondrial inner membrane, which is generated by mitochondrial respiration. A detailed model of this reaction, which includes dynamic equations for the key mitochondrial variables, was developed earlier by Magnus and Keizer. However, this model is extraordinarily complicated. We develop a simpler model that captures the behavior of the original model but is easier to use and to understand. We then use it to investigate the mitochondrial responses to glycolytic and calcium input. We use the model to explain experimental observations of the opposite effects of raising cytosolic Ca 2þ in low and high glucose, and to predict the effects of a mutation in the mitochondrial enzyme nicotinamide nucleotide transhydrogenase (Nnt) in pancreatic b-cells. r
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.