Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1348, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355731

ABSTRACT

HIV-1 persistence during ART is due to the establishment of long-lived viral reservoirs in resting immune cells. Using an NHP model of barcoded SIVmac239 intravenous infection and therapeutic dosing of anti-TGFBR1 inhibitor galunisertib (LY2157299), we confirm the latency reversal properties of in vivo TGF-ß blockade, decrease viral reservoirs and stimulate immune responses. Treatment of eight female, SIV-infected macaques on ART with four 2-weeks cycles of galunisertib leads to viral reactivation as indicated by plasma viral load and immunoPET/CT with a 64Cu-DOTA-F(ab')2-p7D3-probe. Post-galunisertib, lymph nodes, gut and PBMC exhibit lower cell-associated (CA-)SIV DNA and lower intact pro-virus (PBMC). Galunisertib does not lead to systemic increase in inflammatory cytokines. High-dimensional cytometry, bulk, and single-cell (sc)RNAseq reveal a galunisertib-driven shift toward an effector phenotype in T and NK cells characterized by a progressive downregulation in TCF1. In summary, we demonstrate that galunisertib, a clinical stage TGF-ß inhibitor, reverses SIV latency and decreases SIV reservoirs by driving T cells toward an effector phenotype, enhancing immune responses in vivo in absence of toxicity.


Subject(s)
Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Female , Animals , Transforming Growth Factor beta , Virus Replication , Leukocytes, Mononuclear , CD4-Positive T-Lymphocytes , Viral Load
2.
bioRxiv ; 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38014094

ABSTRACT

HIV-1 persistence during ART is due to the establishment of long-lived viral reservoirs in resting immune cells. Using an NHP model of barcoded SIVmac239 intravenous infection and therapeutic dosing of the anti-TGFBR1 inhibitor galunisertib (LY2157299), we confirmed the latency reversal properties of in vivo TGF-ß blockade, decreased viral reservoirs and stimulated immune responses. Eight SIV-infected macaques on suppressive ART were treated with 4 2-week cycles of galunisertib. ART was discontinued 3 weeks after the last dose, and macaques euthanized 6 weeks after ART-interruption(ATI). One macaque did not rebound, while the remaining rebounded between week 2 and 6 post-ATI. Galunisertib led to viral reactivation as indicated by plasma viral load and immunoPET/CT with the 64Cu-DOTA-F(ab')2-p7D3-probe. Half to 1 Log decrease in cell-associated (CA-)SIV DNA was detected in lymph nodes, gut and PBMC, while intact pro-virus in PBMC decreased by 3-fold. No systemic increase in inflammatory cytokines was observed. High-dimensions cytometry, bulk and single-cell RNAseq revealed a shift toward an effector phenotype in T and NK cells. In summary, we demonstrated that galunisertib, a clinical stage TGFß inhibitor, reverses SIV latency and decreases SIV reservoirs by driving T cells toward an effector phenotype, enhancing immune responses in vivo in absence of toxicity.

3.
Microsyst Nanoeng ; 8: 36, 2022.
Article in English | MEDLINE | ID: mdl-35450328

ABSTRACT

The patient population suffering from pancreatic ductal adenocarcinoma (PDAC) presents, as a whole, with a high degree of molecular tumor heterogeneity. The heterogeneity of PDAC tumor composition has complicated treatment and stalled success in clinical trials. Current in vitro techniques insufficiently replicate the intricate stromal components of PDAC tumor microenvironments (TMEs) and fail to model a given tumor's unique genetic phenotype. The development of patient-derived organoids (PDOs) has opened the door for improved personalized medicine since PDOs are derived directly from patient tumors, thus preserving the tumors' unique behaviors and genetic phenotypes. This study developed a tumor-chip device engineered to mimic the PDAC TME by incorporating PDOs and stromal cells, specifically pancreatic stellate cells and macrophages. Establishing PDOs in a multicellular microfluidic chip device prolongs cellular function and longevity and successfully establishes a complex organotypic tumor environment that incorporates desmoplastic stroma and immune cells. When primary cancer cells in monoculture were subjected to stroma-depleting agents, there was no effect on cancer cell viability. However, targeting stroma in our tumor-chip model resulted in a significant increase in the chemotherapy effect on cancer cells, thus validating the use of this tumor-chip device for drug testing.

5.
Mol Cell Biochem ; 477(5): 1339-1345, 2022 May.
Article in English | MEDLINE | ID: mdl-35138511

ABSTRACT

Opioids are the most popular drugs for both acute and chronic pain management. The G protein-coupled mu-opioid receptor (MOR) is the therapeutic target for most clinically used opioids, including morphine. A mounting number of publications suggest a relationship between the MOR and possible cancer progression and recurrence extending to managing chronic cancer pain. In this study, we studied the possible link between opioid use and pancreatic cancer (PC) progression. We found increased MOR expression in murine and human PC cell lines, human PC-derived organoids, and in the undifferentiated or poorly differentiated areas of surgically resected PC tissues. Direct stimulation of MOR by morphine (MOR agonist) caused a significant dose-dependent increase in proliferation, invasion, and levels of stemness markers in PC cells. In a co-culture system, MOR stimulation of macrophages also resulted in increased proliferation of PC cells. MOR overexpression increased proliferation and cancer stemness, whereas knock-down of MOR followed opposite results in the PC cells. Morphine induced chemoresistance to conventional chemotherapeutic agents used for PC treatment. Overall, our results suggest that MOR is expressed in pancreatic cancer and may be involved in tumor progression and chemoresistance.


Subject(s)
Morphine , Pancreatic Neoplasms , Receptors, Opioid, mu , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Cell Line , Humans , Mice , Morphine/adverse effects , Morphine/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/etiology , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism
6.
Cancers (Basel) ; 13(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34503294

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDAC) is an expeditiously fatal malignancy with a five-year survival rate of 6-8%. Conventional chemotherapeutics fail in many cases due to inadequate primary response and rapidly developing resistance. This treatment failure is particularly challenging in pancreatic cancer because of the high molecular heterogeneity across tumors. Additionally, a rich fibro-inflammatory component within the tumor microenvironment (TME) limits the delivery and effectiveness of anticancer drugs, further contributing to the lack of response or developing resistance to conventional approaches in this cancer. As a result, there is an urgent need to model pancreatic cancer ex vivo to discover effective drug regimens, including those targeting the components of the TME on an individualized basis. Patient-derived three-dimensional (3D) organoid technology has provided a unique opportunity to study patient-specific cancerous epithelium. Patient-derived organoids cultured with the TME components can more accurately reflect the in vivo tumor environment. Here we present the advances in organoid technology and multicellular platforms that could allow for the development of "organ-on-a-chip" approaches to recapitulate the complex cellular interactions in PDAC tumors. We highlight the current advances of the organ-on-a-chip-based cancer models and discuss their potential for the preclinical selection of individualized treatment in PDAC.

7.
Biomedicines ; 9(7)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201419

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is an extremely lethal malignancy arising from the pancreas. The treatment of PDA is complicated by ineffective treatments and a lack of biomarkers predictive of treatment success. We have designed a patient-derived organoid (PDO) based high-throughput drug screening assay to model treatment response to a variety of conventional and investigational treatments for PDA. Consecutive patients undergoing endoscopic ultrasound-guided fine-needle biopsy for tissue diagnosis of PDA at Rush University Medical Center were offered to participate in the study. Biopsies were immediately processed to develop organoids. Fifteen PDOs were screened for sensitivity to 18 compounds, including conventional PDA chemotherapies and FDA-approved investigational targeted therapies in cancer using Cell-titer GLO 3D (Promega) cell viability assay. The area under the curve (AUC) was calculated and normalized to the maximum area under the curve to generate a normalized AUC between 0 and 1. Molecular profiling of PDOs was conducted using RNA-seq. Human PDA transcriptomic was extracted from The Cancer Genome Atlas (TCGA). The drug response curves were reproducible. We observed variation in response to conventional therapies overall as well as among individual patients. There were distinct transcriptome signatures associated with response to the conventional chemotherapeutics in PDA. The transcriptomic profile of overall resistance to conventional therapies in our study was associated with poor survival in PDA patients in TCGA. Our pathway analysis for targeted drugs revealed a number of predictors of response associated with the mechanism of action of the tested drug. The multiplex organoid-based drug assay could be used in preclinical to inform patient stratification and therapeutic selection in PDA. When combined with omics data, ex vivo response to treatment could help identify gene signatures associated with response to novel therapies.

8.
J Mater Chem B ; 9(11): 2631-2640, 2021 03 21.
Article in English | MEDLINE | ID: mdl-33683280

ABSTRACT

Islet cell transplantation has been an effective method for the treatment of type 1 diabetes. The transplanted islets release insulin in response to changes in blood glucose levels. The clinical application of islet transplantation, however, has been hindered because of some critical problems including immune responses to grafted islets and side effects caused by overdosed immunosuppressive drugs. Herein, surface modification technology using poly(ethylene glycol) (PEG)-dendron was proposed to safeguard islets from the host immune system. PEG-dendron was synthesized by a divergent polymerization method and utilized to cover the islet antigen surface. Successful conjugation of PEG-dendron on the islet surface was achieved without affecting islet morphology, viability, and functionality at a concentration of 1.00%. Surface modification using PEG-dendron effectively prevented protein absorption and immune activation. Foremost, it improved the survival rate of islet grafts in vivo when combined with a low dose of immunosuppressive drugs. In conclusion, PEG-dendron is a potential candidate for the surface modification of pancreatic islets to mitigate immune responses after transplantation.


Subject(s)
Dendrimers/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Immunosuppressive Agents/pharmacology , Islets of Langerhans/drug effects , Polyethylene Glycols/pharmacology , Animals , Dendrimers/chemistry , Diabetes Mellitus, Experimental/immunology , Immunosuppressive Agents/chemistry , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley
9.
Biomaterials ; 214: 119224, 2019 09.
Article in English | MEDLINE | ID: mdl-31153093

ABSTRACT

Ferumoxytol is the only clinically available ultrasmall superparamagnetic iron oxide. However, the labeling efficacy of islet magnetic resonance imaging (MRI) using ferumoxytol is not suitable for use in clinical pancreatic islet transplantation (PIT). We evaluated the feasibility of pancreatic islet MRI using ferumoxytol through multi-layer surface modification. A four-layer nanoshield with poly (ethylene) glycol (PEG, 2 layers), ferumoxytol, and heparin was formed on the pancreatic islets. We compared pancreatic islet function, viability, and labeling efficacy of control, ferumoxytol alone-labeled, heparin-PEGylated, and ferumoxytol-heparin-PEGylated islets. With optimization of the ferumoxytol concentration during the ferumoxytol-heparin-PEGylation process, the labeling contrast in ex vivo MRI of ferumoxytol-heparin-PEGylated pancreatic islets was stronger than that of pancreatic islets labeled with ferumoxytol alone, without decreasing ex vivo islet viability or function. In a syngeneic mouse renal subcapsular PIT model, heparin-PEGylation and ferumoxytol-heparin-PEGylation delayed the revascularization of pancreatic islet grafts but did not impair glucose tolerance or revascularization of pancreatic islet grafts four weeks post-transplantation. Pancreatic islet visibility after labeling was also confirmed in a syngeneic mouse intraportal PIT model and in preliminary analysis of a non-human primate intraportal PIT model. In conclusion, multi-layer islet surface modification is a promising option for pancreatic islet MRI in intraportal PIT.


Subject(s)
Contrast Media/chemistry , Ferrosoferric Oxide/chemistry , Islets of Langerhans/diagnostic imaging , Magnetic Resonance Imaging/methods , Animals , Heparin/chemistry , Male , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry
10.
Bioconjug Chem ; 29(9): 2945-2953, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29985588

ABSTRACT

The microenvironment of pancreatic islets gets disrupted during enzyme digestion and causes islets to remain in a vulnerable state, leading to poor outcome in the initial days of transplantation. To avoid immune invasion while allowing the reconstruction of the microenvironment of the transplanted site, we propose immunoisolation polymers, which can nanoencapsulate islets quickly without cytotoxicity. Here, nonhuman primate (NHP) islets were nanoencapsulated with hyperbranched polyethylene glycol (hb-PEG) and heparin by layer-by-layer technology and transplanted into the kidney subcapsular space of diabetic C57BL/6 mice. An immunosuppressive drug protocol was applied to increase the survival time until the animals were sacrificed. The recipients of NHP islets exhibited high nonfasting blood glucose level (BGL) for 2-3 weeks, which was normalized afterward. Immunohistochemical (IHC) analysis revealed an immature vascular basement membrane and cell surface integrins directly associated with poor initial insulin production. The transplanted grafts regained their own microenvironment within a month without any outside stimuli. No lymphocyte infiltration was observed in the grafts at any time. Humoral and cell-mediated immune responses were prominently diminished by the hb-PEG/Heparin nanoencapsulated islets. Immunoisolation accompanied by an immunosuppressive drug protocol protects islets by helping them avoid immunogenesis while at the same time allowing them to reconstruct their microenvironment.


Subject(s)
Blood Glucose/metabolism , Cellular Microenvironment , Heparin/chemistry , Islets of Langerhans Transplantation/methods , Nanotechnology , Polyethylene Glycols/chemistry , Animals , Antibody Formation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Immunity, Cellular , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL
11.
Biomaterials ; 171: 164-177, 2018 07.
Article in English | MEDLINE | ID: mdl-29698867

ABSTRACT

Intraportal pancreatic islet transplantation incurs huge cell losses during its early stages due to instant blood-mediated inflammatory reactions (IBMIRs), which may also drive regulation of the adaptive immune system. Therefore, a method that evades IBMIR will improve clinical islet transplantation. We used a layer-by-layer approach to shield non-human primate (NHP) islets with polyethylene glycol (nano-shielded islets, NSIs) and polyethylene glycol plus heparin (heparin nano-shielded islets; HNSIs). Islets ranging from 10,000 to 20,000 IEQ/kg body weight were transplanted into 19 cynomolgus monkeys (n = 4, control; n = 5, NSI; and n = 10, HNSI). The mean C-peptide positive graft survival times were 68.5, 64 and 108 days for the control, NSI and HNSI groups, respectively (P = 0.012). HNSI also reduced the factors responsible for IBMIR in vitro. Based on these data, HNSIs in conjunction with clinically established immunosuppressive drug regimens will result in superior outcomes compared to those achieved with the current protocol for clinical islet transplantation.


Subject(s)
Heparin/chemistry , Islets of Langerhans/physiology , Nanoparticles/chemistry , Polymers/chemistry , Allografts/physiology , Animals , Graft Survival , Humans , Liver/pathology , Lymphocyte Subsets/metabolism , Macaca fascicularis , Polyethylene Glycols/chemistry
12.
J Control Release ; 258: 10-21, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28433740

ABSTRACT

Islet transplantation is as effective as but also less immunogenic than pancreas transplantation for the treatment of type 1 diabetes mellitus. However, as the complete elimination of immunogenicity still remains a major obstacle in islet transplantation, layer-by-layer encapsulation (LbL) of pancreatic islets using biocompatible polymers offers a rational approach to reducing host immune response towards transplanted islets. We investigated the effect of LbL of non-human primate (NHP) islets on reducing immunogenicity as a preclinical model since NHPs have close phylogenetic and immunological relationship with humans. LbL with three-layers of polyethylene glycol (PEG) molecules (SH-6-arm-PEG-NHS, 6-arm-PEG-catechol and linear PEG-SH) showed a uniform nano-shielding on islets without the loss of viability or function of islets. An immunosuppressive drug protocol was also combined to improve the survival rate of the transplanted islets in vivo. A xenorecipient (C57BL/6 mice) of LbL islet transplanted along with our immunosuppressive drug protocol showed 100% survival rate for 150days after transplantation. On the other hand, naked islet recipients showed poor survival time of 5.5±1.4days without drugs and 77.5±42days with the drug protocol. Immunohistochemistry of the transplanted grafts and serum cytokine concentration demonstrated less immunogenicity in the LbL islet transplanted recipients compared with the naked islet ones.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Polyethylene Glycols/chemistry , Animals , Catechols/chemistry , Cell Separation , Cells, Immobilized/cytology , Graft Survival/drug effects , Humans , Macaca fascicularis , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Transplantation, Heterologous
13.
Biomaterials ; 84: 144-156, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26828680

ABSTRACT

This study suggests a novel method of stabilizing fragile porcine islets to prevent the dissociation after isolation and reducing immune cell invasion in a combination therapy of 'surface camouflaging' and immunosuppressive drugs (FK506, Rapamycin, MR-1, anti-CD19 mAb, and Clodrosome(®)) to effectively alleviate overall immune reactions against xenotransplanted porcine islets. The surface camouflage of pancreatic islets using biocompatible materials improved stabilization of pancreatic islet and prevented the infiltration of immune cells. Firstly, the surface of porcine islets was camouflaged by SH-6-arm-PEG-lipid and gelatin-catechol (artificial extracellular matrix) in order to stabilize the fragile isolated islets. Secondly, three different PEG layers (6-arm-PEG-SH, 6-arm-PEG-catechol, and linear PEG-SH) were chemically conjugated onto the surface of the stabilized porcine islets. Both artificial extracellular matrix (artificial ECM) and PEGylation effectively covered the surface of porcine islets without increasing the size of the whole islet. In addition, the viability and functionality of the islets were not affected by this multi-layer surface modification. The multi-layer modification significantly reduced the attachment of human serum albumin, fibronectin, and immunoglobulin G in comparison to the control collagen surface. The combination effect of multi-layer PEGylation and cocktailed immunosuppressive drugs on the survival time of the transplanted islets was assessed in a xenogeneic porcine-to-mouse model. The median survival time (MST) of 'artificial ECM + PEGylation' group was 4-fold increased compared to that of control group. In addition, the MST of 'artificial ECM + PEGylation + drug' group was 2.16-fold increased, compared to the 'control + drug' group. In conclusion, we proposed a novel porcine islet transplantation protocol using surface multi-layer modification and cocktailed immunosuppressive drugs, for stabilization and immunoprotection against xenogeneic immune reactions.


Subject(s)
Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/immunology , Polyethylene Glycols/chemistry , Adsorption , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Blood Glucose/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Humans , Islets of Langerhans/drug effects , Male , Mice, Inbred C57BL , Microscopy, Atomic Force , Rats, Sprague-Dawley , Sus scrofa , Tissue Survival/drug effects
14.
Tissue Eng Part A ; 21(5-6): 1024-35, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25344077

ABSTRACT

Although islet transplantation has been suggested as an alternative therapy for type 1 diabetes, there are efficiency concerns that are attributed to poor engraftment of transplanted islets. Hypoxic condition and delayed vasculogenesis induce necrosis and apoptosis of the transplanted islets. To overcome these limitations in islet transplantation, heterospheroids (HSs), which consist of rat islet cells (ICs) and human bone marrow-derived mesenchymal stem cells (hMSCs), were transplanted to the kidney and liver. The HSs cultured under the hypoxic condition system exhibited a significant increase in antiapoptotic gene expression in ICs. hMSCs in the HSs secreted angiogenic and antiapoptotic proteins. With the HS system, ICs and hMSCs were successfully located in the same area of the liver after transplantation of HSs through the portal vein, whereas the transplantation of islets and the dissociated hMSCs did not result in localization of transplanted ICs and hMSCs in the same area. HS transplantation resulted in an increase in angiogenesis at the transplantation area and a decrease in the apoptosis of transplanted ICs after transplantation into the kidney subcapsule compared with transplantation of islet cell clusters (ICCs). Insulin production levels of ICs were higher in the HS transplantation group compared with the ICC transplantation group. The HS system may be a more efficient transplantation method than the conventional methods for the treatment of type 1 diabetes.


Subject(s)
Apoptosis , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Spheroids, Cellular/cytology , Animals , Cell Aggregation , Cell Count , Cell Survival , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/metabolism , Kidney/cytology , Liver/cytology , Male , Portal Vein/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
15.
Pharm Res ; 31(9): 2453-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24633416

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of locally delivered pancreatic islet with liposomal clodronate (Clodrosome®) as an immunoprotection agent for the treatment of type 1 diabetes. METHOD: The bio-distribution of liposomal clodronate in matrigel was checked by imaging analyzer. To verify the therapeutic efficacy of locally delivered islet with liposomal clodronate using injectable hydrogel, four groups of islet transplanted mice (n = 6 in each group) were prepared: 1) the islet group, 2) the islet-Clodrosome group, 3) the islet-Matrigel group, and 4) the islet-Matrigel-Clodrosome group. Immune cell migration and activation, and pro-inflammatory cytokine secretion was evaluated by immunohistochemistry staining and ELISA assay. RESULTS: Cy5.5 labeled liposomes remained in the matrigel for over 7 days. The median survival time of transplanted islets (Islet-Matrigel-Clodrosome group) was significantly increased (>60 days), compared to other groups. Locally delivered liposomal clodronate in matrigel effectively inhibited the activation of macrophages, immune cell migration and activation, and pro-inflammatory cytokine secretion from macrophages. CONCLUSIONS: Locally co-delivered pancreatic islets and liposomal clodronate using injectable hydrogel effectively cured type 1 diabetes. Especially, the inhibition of macrophage attack in the early stage after local delivery of islets was very important for the successful long-term survival of delivered islets.


Subject(s)
Clodronic Acid/administration & dosage , Collagen/administration & dosage , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Laminin/administration & dosage , Proteoglycans/administration & dosage , Animals , Clodronic Acid/therapeutic use , Collagen/therapeutic use , Diabetes Mellitus, Type 1/immunology , Drug Combinations , Inflammation/immunology , Inflammation/prevention & control , Injections , Laminin/therapeutic use , Liposomes , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Proteoglycans/therapeutic use , Rats, Sprague-Dawley
16.
Biomaterials ; 34(8): 2098-106, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23261215

ABSTRACT

This study proposed a double-layer shielding method of using 8-arm-PEG-catechol (PEG(8)) and N-hydroxysuccinimidyl-linked unfractionated heparin (UFH-NHS) for the prevention of instant blood-mediated inflammatory reaction (IBMIR) and immune reactions against transplanted pancreatic islets. The surface of islet was evenly covered by PEG(8) and UFH-NHS. Both viability and functionality of islets were evaluated in vitro, and the anti-coagulation effect of conjugated heparin on the islet surface was also evaluated. The inhibition effects of PEG(8)/UFH double-layer shielding system on immune reactions and IBMIR induced by transplanted islets were evaluated in an allograft model. When pancreatic islets of Sprague-Dawley (SD) rats were transplanted in the liver of F344 rats, the mean survival time (MST) of PEG(8)/UFH double-layer shielded islets (6.8 ± 1.6 days) was statistically increased, compared to that of unmodified islets (3.6 ± 1.1 days). Furthermore, when 0.5 mg/kg of FK506 was daily administered, the MST of double-layer shielded islet (15.0 ± 2.1 days) was increased by two-fold, compared to that of unmodified islets treated with the same dose of FK506 (8.0 ± 2.4 days). Therefore, a newly developed strategy of combining the PEG(8)/UFH double-layer shielding system with FK506 would certainly be effective for preventing immune activation and IBMIR against allotransplanted islets.


Subject(s)
Catechols/chemistry , Graft Survival/drug effects , Heparin/chemistry , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation , Polyethylene Glycols/chemistry , Tacrolimus/pharmacology , Animals , Anticoagulants/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Fasting/blood , Fluorescein-5-isothiocyanate/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Succinimides/chemistry , Tacrolimus/chemistry , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...