Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Diabetes Investig ; 15(6): 693-703, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38634411

ABSTRACT

AIMS/INTRODUCTION: To investigate the long-term efficacy of various encapsulated xenogeneic islet transplantation, and to explore the impact of different donor porcine genetic traits on islet transplantation outcomes. MATERIALS AND METHODS: Donor porcine islets were obtained from wild-type, α1,3-galactosyltransferase knockout (GTKO) and GTKO with overexpression of membrane cofactor protein genotype. Naked, alginate, alginate-chitosan (AC), alginate-perfluorodecalin (A-PFD) and AC-perfluorodecalin (AC-PFD) encapsulated porcine islets were transplanted into diabetic mice. RESULTS: In vitro assessments showed no differences in the viability and function of islets across encapsulation types and donor porcine islet genotypes. Xenogeneic encapsulated islet transplantation with AC-PFD capsules showed the most favorable long-term outcomes, maintaining normal blood glucose levels for 180 days. A-PFD capsules showed comparable results to AC-PFD capsules, followed by AC capsules and alginate capsules. Conversely, blood glucose levels in naked islet transplantation increased to >300 mg/dL within a week after transplantation. Naked islet transplantation outcomes showed no improvement based on donor islet genotype. However, alginate or AC capsules showed delayed increases in blood glucose levels for GTKO and GTKO with overexpression of membrane cofactor protein porcine islets compared with wild-type porcine islets. CONCLUSION: The AC-PFD capsule, designed to ameliorate both hypoxia and inflammation, showed the highest long-term efficacy in xenogeneic islet transplantation. Genetic modifications of porcine islets with GTKO or GTKO with overexpression of membrane cofactor protein did not influence naked islet transplantation outcomes, but did delay graft failure when encapsulated.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Transplantation, Heterologous , Islets of Langerhans Transplantation/methods , Animals , Swine , Mice , Transplantation, Heterologous/methods , Diabetes Mellitus, Experimental/therapy , Alginates , Galactosyltransferases/genetics , Graft Survival , Islets of Langerhans , Blood Glucose/analysis , Male , Genotype , Tissue Donors
2.
BMJ Open Diabetes Res Care ; 12(2)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38485229

ABSTRACT

INTRODUCTION: Static incubation (static glucose-stimulated insulin secretion, sGSIS) is a measure of islet secretory function. The Stimulation Index (SI; insulin produced in high glucose/insulin produced in low glucose) is currently used as a product release criterion of islet transplant potency. RESEARCH DESIGN AND METHODS: Our hypothesis was that the Delta, insulin secreted in high glucose minus insulin secreted in low glucose, would be more predictive. To evaluate this hypothesis, sGSIS was performed on 32 consecutive human islet preparations, immobilizing the islets in a slurry of Sepharose beads to minimize mechanical perturbation. Simultaneous full-mass subrenal capsular transplants were performed in chemically induced diabetic immunodeficient mice. Logistic regression analysis was used to determine optimal cut-points for diabetes reversal time and the Fisher Exact Test was used to assess the ability of the Delta and the SI to accurately classify transplant outcomes. Receiver operating characteristic curve analysis was performed on cut-point grouped data, assessing the predictive power and optimal cut-point for each sGSIS potency metric. Finally, standard Kaplan-Meier-type survival analysis was conducted. RESULTS: In the case of the sGSIS the Delta provided a superior islet potency metric relative to the SI.ConclusionsThe sGSIS Delta value is predicitive of time to diabetes reversal in the full mass human islet transplant bioassay.


Subject(s)
Diabetes Mellitus , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Mice , Animals , Insulin Secretion , Glucose/pharmacology , Glucose/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/physiology , Diabetes Mellitus/metabolism , Insulin/metabolism , Biological Assay
4.
Journal of Clinical Hepatology ; (12): 2506-2512, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-998322

ABSTRACT

Since the 1970s, patients with chronic pancreatitis (CP) have benefited from total pancreatectomy with autologous islet cell transplantation (TPAIT). With the continuous development of surgical techniques and perioperative management over the past few decades, there have been improvements in islet cell function, insulin independence rate, and the survival rate of patients. This article summarizes the preoperative indications for TPAIT, the development of surgical operations, postoperative management and monitoring, and prognosis, so as to help clinicians learn more about TPAIT.

5.
Journal of Chinese Physician ; (12): 321-324, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-992301

ABSTRACT

Type 1 diabetes mellitus (T1DM) is an organ-specific disease characterized by autoimmune damage to pancreatic β cells. Insulin therapy is the most basic and important treatment for T1DM, but insulin therapy cannot fundamentally terminate or improve the main cause of T1DM, namely the disorder of the immune regulation mechanism. With the advancement of science and technology, the continuous development of new insulin and hypoglycemic drugs has provided better means for glycemic control. Pancreas transplantation, islet transplantation, immunotherapy, and cell therapy have provided hope for the prevention or reversal of T1DM. It is of great significance to understand the current situation and future of new technologies for T1DM treatment for the research and management of T1DM patients.

6.
Endocrinol Metab (Seoul) ; 36(1): 146-156, 2021 02.
Article in English | MEDLINE | ID: mdl-33677936

ABSTRACT

BACKGROUND: The microencapsulation is an ideal solution to overcome immune rejection without immunosuppressive treatment. Poor biocompatibility and small molecular antigens secreted from encapsulated islets induce fibrosis infiltration. Therefore, the aims of this study were to improve the biocompatibility of microcapsules by dexamethasone coating and to verify its effect after xenogeneic transplantation in a streptozotocin-induced diabetes mice. METHODS: Dexamethasone 21-phosphate (Dexa) was dissolved in 1% chitosan and was cross-linked with the alginate microcapsule surface. Insulin secretion and viability assays were performed 14 days after microencapsulation. Dexa-containing chitosan-coated alginate (Dexa-chitosan) or alginate microencapsulated porcine islets were transplanted into diabetic mice. The fibrosis infiltration score was calculated from the harvested microcapsules. The harvested microcapsules were stained with trichrome and for insulin and macrophages. RESULTS: No significant differences in glucose-stimulated insulin secretion and islet viability were noted among naked, alginate, and Dexa-chitosan microencapsulated islets. After transplantation of microencapsulated porcine islets, nonfasting blood glucose were normalized in both the Dexa-chitosan and alginate groups until 231 days. The average glucose after transplantation were lower in the Dexa-chitosan group than the alginate group. Pericapsular fibrosis and inflammatory cell infiltration of microcapsules were significantly reduced in Dexa-chitosan compared with alginate microcapsules. Dithizone and insulin were positive in Dexa-chitosan capsules. Although fibrosis and macrophage infiltration was noted on the surface, some alginate microcapsules were stained with insulin. CONCLUSION: Dexa coating on microcapsules significantly suppressed the fibrotic reaction on the capsule surface after transplantation of xenogenic islets containing microcapsules without any harmful effects on the function and survival of the islets.


Subject(s)
Chitosan , Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Alginates/metabolism , Alginates/pharmacology , Animals , Capsules/metabolism , Capsules/pharmacology , Chitosan/metabolism , Chitosan/pharmacology , Dexamethasone/metabolism , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/surgery , Fibrosis , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Swine
7.
Stem Cells Transl Med ; 10(5): 674-680, 2021 05.
Article in English | MEDLINE | ID: mdl-33544449

ABSTRACT

Islet transplantation is an emerging treatment for type 1 diabetes which offers the prospect of physiological control of blood glucose and reductions in acute hypoglycaemic episodes. However, current protocols are limited by a rapid decline in islet functional viability during the isolation process, culture period, and post-transplantation. Much of this can be attributed to the deleterious effects of hypoxic and cytokine stressors on ß cells. One experimental strategy to improve the functional viability of islets is coculture or cotransplantation with mesenchymal stromal cells (MSCs). Numerous studies have shown that MSCs have the capacity to improve islet survival and insulin secretory function, and the mechanisms of these effects are becoming increasingly well understood. In this review, we will focus on recent studies demonstrating the capacity for MSCs to protect islets from hypoxia- and cytokine-induced stress. Islets exposed to acute hypoxia (1%-2% O2 ) or to inflammatory cytokines (including IFN-γ, TNF-α, and IL-B) in vitro undergo apoptosis and a rapid decline in glucose-stimulated insulin secretion. Coculture of islets with MSCs, or with MSC-conditioned medium, protects from these deleterious effects, primarily with secreted factors. These protective effects are distinct from the immunomodulatory and structural support MSCs provide when cotransplanted with islets. Recent studies suggest that MSCs may support secretory function by the physical transfer of functional mitochondria, particularly to metabolically compromised ß cells. Understanding how MSCs respond to stressed islets will facilitate the development of MSC secretome based, cell-free approaches to supporting islet graft function during transplantation by protecting or repairing ß cells.


Subject(s)
Islets of Langerhans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Coculture Techniques , Cytokines/metabolism , Humans , Hypoxia/metabolism , Insulin , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Mesenchymal Stem Cells/metabolism , Secretome
8.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-910577

ABSTRACT

Chronic pancreatitis is a progressive chronic inflammatory disease of caused by gene and other environmental factors, and clinical manifestation includes recurrent abdominal pain and dysfunction of exocrine and endocrine. For the chronic pancreatitis therapy, surgical treatment mainly aims at the intractable pain which is unresponsive to medical and endoscopic treatment, as well as complications of chronic pancreatitis. Total pancreatectomy with islet autotransplantation (TPIAT) gradually becomes a major therapeutic option for chronic pancreatitis surgical treatment, because it relieves the abdominal pain, reduces the opioid dependent, improves the quality of life, and increases the opportunity of insulin independent. In the past few years, a range of researches have been focusing on islet isolation, surgical approach, curative effect and postoperative complication, improvement of islet function after operation. The purpose of this article is to summarize the progression of TPIAT related research in the recent years.

9.
BMC Nephrol ; 21(1): 405, 2020 09 19.
Article in English | MEDLINE | ID: mdl-32950058

ABSTRACT

BACKGROUND: We here report on the first observation of a C3 mutation that is related to atypical hemolytic and uremic syndrome (aHUS), which occurred in a pancreatic islet transplant patient. Immunosuppressive treatments, such as calcineurin inhibitors, have been linked to undesirable effects like nephrotoxicity. CASE PRESENTATION: A 40-year-old man with brittle diabetes, who was included in the TRIMECO trial, became insulin-independent 2 months after pancreatic islet transplantation. About 15 months after islet transplantation, the patient exhibited acute kidney injury due to aHUS. Despite plasma exchange and eculizumab treatment, the patient developed end-stage renal disease. A genetic workup identified a missense variant (p.R592Q) in the C3 gene. In vitro, this C3 variant had defective Factor I proteolytic activity with membrane proteins as cofactor proteins, which was thus classified as pathogenic. About 1 year after the aHUS episode, kidney transplantation was carried out under the protection of the specific anti-C5 monoclonal antibody eculizumab. The patient had normal kidney function, with preserved pancreatic islet function 4 years later. CONCLUSIONS: Pancreatic islet transplantation could have triggered this aHUS episode, but this link needs to be clarified. Although prophylactic eculizumab maintains kidney allograft function, its efficacy still needs to be studied in larger populations.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement C3/genetics , Islets of Langerhans Transplantation , Mutation, Missense , Acute Kidney Injury/etiology , Adult , Diabetes Mellitus, Type 1/surgery , Humans , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Male
10.
Biochem Biophys Res Commun ; 528(4): 650-657, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32513541

ABSTRACT

INTRODUCTION: The extension of islet transplantation to a wider number of type 1 diabetes patients is compromised by severe adverse events related to the immunosuppressant therapy required for allogenic islet transplantation. In this context, microencapsulation offers the prospects of immunosuppressive-free therapy by physically isolating islets from the immune system. However, current biomaterials need to be optimized to: improve biocompatibility, guaranty the maintenance of graft viability and functionality, and prevent fibrosis overgrowth around the capsule in vivo. Accumulating evidence suggest that mesenchymal stem cells (MSCs) and anchor points consisting of tripeptides arg-gly-asp (RGD) have cytoprotective effects on pancreatic islets. Here, we investigated the effect of supplementing reference M-rich alginate microcapsules with MSCs and RGD-G rich alginate on bioprocessing as well as on human pancreatic islets viability and functionality. METHODS: We characterized the microcapsules components, and then for the new microcapsule composite product: we analyzed the empty capsules biocompatibility and then investigated the benefits of MSCs and RGD-G rich alginate on viability and functionality on the encapsulated human pancreatic islets in vitro. We performed viability tests by confocal microscopy and glucose stimulated insulin secretion (GSIS) test in vitro to assess the functionality of naked and encapsulated islets. RESULTS: Encapsulation in reference M-rich alginate capsules induced a reduction in viability and functionality compared to naked islets. This side-effect of encapsulation was in part counteracted by the presence of MSCs but the restoration was complete with the combination of both MSCs and the RGD-G rich alginate. CONCLUSIONS: The present findings show that bioprocessing a favorable composite environment inside the M-rich alginate capsule with both MSCs and RGD-G rich alginate improves human islets survival and functionality in vitro.


Subject(s)
Cell Survival/drug effects , Cells, Immobilized/cytology , Islets of Langerhans/cytology , Mesenchymal Stem Cells/cytology , Oligopeptides/pharmacology , Adult , Alginates/chemistry , Cells, Cultured , Cells, Immobilized/drug effects , Humans , Islets of Langerhans/drug effects , Mesenchymal Stem Cells/drug effects , Middle Aged
11.
Stem Cell Res Ther ; 10(1): 85, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30867050

ABSTRACT

BACKGROUND: Islets of Langerhans transplantation is a promising therapy for type 1 diabetes mellitus, but this technique is compromised by transplantation stresses including inflammation. In other tissues, co-transplantation with mesenchymal stem cells has been shown to reduce damage by improving anti-inflammatory and anti-oxidant defences. Therefore, we probed the protection afforded by bone marrow mesenchymal stem cells to islets under pro-inflammatory cytokine stress. METHODS: In order to evaluate the cytoprotective potential of mesenchymal stem cells on rat islets, co-cultures were exposed to the interleukin-1, tumour necrosis factor α and interferon γ cocktail for 24 h. Islet viability and functionality tests were performed. Reactive oxygen species and malondialdehyde were measured. Expression of stress-inducible genes acting as anti-oxidants and detoxifiers, such as superoxide dismutases 1 and 2, NAD(P)H quinone oxidoreductase 1, heme oxygenase-1 and ferritin H, was compared to non-stressed cells, and the corresponding proteins were measured. Data were analysed by a two-way ANOVA followed by a Holm-Sidak post hoc analysis. RESULTS: Exposure of rat islets to cytokines induces a reduction in islet viability and functionality concomitant with an oxidative status shift with an increase of cytosolic ROS production. Mesenchymal stem cells did not significantly increase rat islet viability under exposure to cytokines but protected islets from the loss of insulin secretion. A drastic reduction of the antioxidant factors heme oxygenase-1 and ferritin H protein levels was observed in islets exposed to the cytokine cocktail with a prevention of this effect by the presence of mesenchymal stem cells. CONCLUSIONS: Our data evidenced that MSCs are able to preserve islet insulin secretion through a modulation of the oxidative imbalance mediated by heme and iron via heme oxygenase-1 and ferritin in a context of cytokine exposure.


Subject(s)
Cytokines/pharmacology , Ferritins/biosynthesis , Heme Oxygenase (Decyclizing)/biosynthesis , Islets of Langerhans/metabolism , Mesenchymal Stem Cells/metabolism , Stress, Physiological/drug effects , Up-Regulation/drug effects , Animals , Coculture Techniques , Humans , Islets of Langerhans/cytology , Mesenchymal Stem Cells/cytology , Rats
12.
Cytotherapy ; 20(9): 1124-1142, 2018 09.
Article in English | MEDLINE | ID: mdl-30068495

ABSTRACT

BACKGROUND AIMS: Adipose tissue-derived mesenchymal stromal cells (AT-MSCs), widely known as multipotent progenitors, release several cytokines that support cell survival and repair. There are in vitro and in vivo studies reporting the regenerative role of AT-MSCs possibly mediated by their protective effects on functional islet cells as well as their capacity to differentiate into insulin-producing cells (IPCs). METHODS: On such a basis, our goal in the present study was to use three different models including direct and indirect co-cultures and islet-derived conditioned medium (CM) to differentiate AT-MSCs into IPCs and to illuminate the molecular mechanisms of the beneficial impact of AT-MSCs on pancreatic islet functionality. Furthermore, we combined in vitro co-culture of islets and AT-MSCs with in vivo assessment of islet graft function to assess whether co-transplantation of islets with AT-MSCs can reduce marginal mass required for successful islet transplantation and prolong graft function in a diabetic rat model. RESULTS: Our findings demonstrated that AT-MSCs are suitable for creating a microenvironment favorable for the repair and longevity of the pancreas ß cells through the improvement of islet survival and maintenance of cell morphology and insulin secretion due to their potent properties in differentiation. Most importantly, hybrid transplantation of islets with AT-MSCs significantly promoted survival, engraftment and insulin-producing function of the graft and reduced the islet mass required for reversal of diabetes. CONCLUSIONS: This strategy might be of therapeutic potential solving the problem of donor islet material loss that currently limits the application of allogeneic islet transplantation as a more widespread therapy for type 1 diabetes.


Subject(s)
Adipose Tissue/cytology , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Mesenchymal Stem Cell Transplantation/methods , Animals , Cell Differentiation , Coculture Techniques , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/therapy , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Rats, Wistar
13.
Diabetes Metab J ; 42(2): 164-168, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29676546

ABSTRACT

Over the past three decades, human pancreatic islet isolation and transplantation techniques have developed as a routine clinical procedure for selected patients with type 1 diabetes mellitus. However, due to the donor shortage and required chronic systemic immunosuppression, the widespread application of islet transplantation is limited. To overcome these limitations, providing a physical barrier to transplanted islet cells with encapsulating biomaterial has emerged as a promising approach to enhance engraftment and promote islet survival post-transplantation. Alginate has been considered to be a reliable biomaterial, as it enhances islet survival and does not hamper hormone secretion. Alginate-catechol (Al-CA) hydrogel was reported to provide high mechanical strength and chemical stability without deformation over a wide range of pH values. In this study, we, demonstrated, for the first time in the literature, that encapsulation of murine pancreatic islet cells with Al-CA hydrogel does not induce cytotoxicity ex vivo for an extended period; however, it does markedly abate glucose-stimulated insulin secretion. Catechol should not be considered as a constituent for alginate gelation for encapsulating islet cells in the application of islet transplantation.

14.
Horm Res Paediatr ; 90(4): 270-274, 2018.
Article in English | MEDLINE | ID: mdl-29669347

ABSTRACT

BACKGROUND: Cystic fibrosis-related diabetes (CFRD) is the most frequent extrapulmonary complication of cystic fibrosis (CF). METHODS: We report the first combined pancreatic islet-lung-liver transplantation in a 14-year-old adolescent. CFTR was analyzed by Sanger sequencing. Further genes were analyzed by high-throughput sequencing. RESULTS: The patient was diagnosed with CF at the age of 14 months. Nine years later, after diagnosis of CFRD, the patient's BMI and lung function began to decline. Bilateral lung transplantation with simultaneous liver transplantation was performed at the age of 14.5 years. The first islet transplantation (IT) was carried out 10 days later. Six months later, C-peptide secretion after arginine stimulation showed peak values of 371 pmol/L (vs. 569 pmol/L before IT) and insulin doses had slightly increased (1.40 vs. 1.11 units/kg/day before IT). A second IT was performed at the age of 15 years, a third at 16 years. Two years after the first IT, arginine-stimulated C-peptide secretion increased to 2,956 pmol/L and insulin doses could be reduced to 0.82 units/kg/day. HbA1c decreased from 7.3% (57.4 mmol/mol) to 5.9% (41.0 mmol/mol). CONCLUSION: IT following lung and liver transplantation, with injection of islets into a transplanted organ, is feasible. It improves C-peptide secretion, decreases insulin needs, and lowers HbA1c.


Subject(s)
C-Peptide/blood , Cystic Fibrosis , Diabetes Mellitus , Glycated Hemoglobin/metabolism , Insulin/administration & dosage , Islets of Langerhans Transplantation , Liver Transplantation , Lung Transplantation , Adolescent , Cystic Fibrosis/blood , Cystic Fibrosis/therapy , Diabetes Mellitus/blood , Humans , Male
15.
J Tissue Eng Regen Med ; 12(1): e171-e176, 2018 01.
Article in English | MEDLINE | ID: mdl-27687889

ABSTRACT

Rat and human beta cell proteomes were quantified by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS), searching for cell surface markers. In human beta cells, CD99 (cluster of differentiation 99) was ranked among the plasma membrane proteins that combine a high molar abundance with a relative degree of selectivity for the endocrine cells of the islets of Langerhans. Therefore, the applicability of CD99 as anchor for islet endocrine cell purification was investigated. The CD99 gene and protein expression were studied using microarray, LC-MS/MS, western blotting, flow cytometry and immunofluorescence, and a protocol was developed for magnetic bead-mediated beta cell enrichment from human pancreas digests using available anti-CD99 antibodies. In human, but not in rat, CD99 protein and mRNA were abundantly expressed by islet endocrine cells but undetectable in exocrine pancreas. The extracellular CD99 epitopes appeared to be trypsin-resistant, enabling the binding of anti-CD99 antibodies to an insulin+/TSQ+ cell subset and efficient coupling of magnetic beads for positive selection of CD99+ cells. A MACS-CD99 purification of human pancreas fractions with low endocrine purity consistently yielded a fourfold enrichment of insulin+/TSQ+ cells and formation of viable and functional endocrine aggregates after 24 h of culture. It is concluded that CD99 is a human beta cell surface marker that, by virtue of its high molar abundance and resistance to tryptic digestion, can be used as anchor for upscalable magnetic bead-mediated islet endocrine cell purification. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
12E7 Antigen/metabolism , Cell Separation/methods , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , 12E7 Antigen/chemistry , Animals , Humans , Protein Domains , Rats
16.
Article in English | WPRIM (Western Pacific) | ID: wpr-714101

ABSTRACT

Over the past three decades, human pancreatic islet isolation and transplantation techniques have developed as a routine clinical procedure for selected patients with type 1 diabetes mellitus. However, due to the donor shortage and required chronic systemic immunosuppression, the widespread application of islet transplantation is limited. To overcome these limitations, providing a physical barrier to transplanted islet cells with encapsulating biomaterial has emerged as a promising approach to enhance engraftment and promote islet survival post-transplantation. Alginate has been considered to be a reliable biomaterial, as it enhances islet survival and does not hamper hormone secretion. Alginate-catechol (Al-CA) hydrogel was reported to provide high mechanical strength and chemical stability without deformation over a wide range of pH values. In this study, we, demonstrated, for the first time in the literature, that encapsulation of murine pancreatic islet cells with Al-CA hydrogel does not induce cytotoxicity ex vivo for an extended period; however, it does markedly abate glucose-stimulated insulin secretion. Catechol should not be considered as a constituent for alginate gelation for encapsulating islet cells in the application of islet transplantation.


Subject(s)
Humans , Architectural Accessibility , Diabetes Mellitus, Type 1 , Hydrogels , Hydrogen-Ion Concentration , Immunosuppression Therapy , Insulin , Islets of Langerhans Transplantation , Islets of Langerhans , Temefos , Tissue Donors
17.
Endocrinol Metab (Seoul) ; 32(2): 190-194, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28685509

ABSTRACT

Impaired awareness of hypoglycemia has been found to be prevalent in 20% to 40% of people with type 1 diabetes. If a similar prevalence exists in Koreans with type 1 diabetes, at a minimum, thousands of people with type 1 diabetes suffer at least one unpredicted episode of severe hypoglycemia per year in Korea. For patients with problematic hypoglycemia, an evidence-based stepwise approach was suggested in 2015. The first step is structured education regarding multiple daily injections of an insulin analog, and the second step is adding a technological intervention, such as continuous subcutaneous insulin infusion or real-time continuous glucose monitoring. The next step is a sensor-augmented pump, preferably with a low glucose suspension feature or very frequent contact, and the final step is islet or pancreas transplantation. In Korea, however, none of these treatments are reimbursed by the National Health Insurance, and thus have not been widely implemented. The low prevalence of type 1 diabetes means that Korean physicians are relatively unfamiliar with the new technologies in this field. Therefore, the roles of new technologies and pancreas or islet transplantation in the treatment of problematic hypoglycemia need to be defined in the current clinical setting of Korea.

18.
Korean J Intern Med ; 32(1): 62-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28049286

ABSTRACT

Since the report of type 1 diabetes reversal in seven consecutive patients by the Edmonton protocol in 2000, pancreatic islet transplantation has been reappraised based on accumulated clinical evidence. Although initially expected to therapeutically target long-term insulin independence, islet transplantation is now indicated for more specific clinical benefits. With the long-awaited report of the first phase 3 clinical trial in 2016, allogeneic islet transplantation is now transitioning from an experimental to a proven therapy for type 1 diabetes with problematic hypoglycemia. Islet autotransplantation has already been therapeutically proven in chronic pancreatitis with severe abdominal pain refractory to conventional treatments, and it holds promise for preventing diabetes after partial pancreatectomy due to benign pancreatic tumors. Based on current evidence, this review focuses on islet transplantation as a realistic approach to treating diabetes.


Subject(s)
Diabetes Mellitus/surgery , Islets of Langerhans Transplantation , Pancreatectomy/adverse effects , Pancreatic Neoplasms/surgery , Animals , Biomarkers/blood , Blood Glucose/metabolism , Clinical Trials, Phase III as Topic , Diabetes Mellitus/blood , Diabetes Mellitus/etiology , Humans , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/methods , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome
19.
Article in English | WPRIM (Western Pacific) | ID: wpr-161478

ABSTRACT

Impaired awareness of hypoglycemia has been found to be prevalent in 20% to 40% of people with type 1 diabetes. If a similar prevalence exists in Koreans with type 1 diabetes, at a minimum, thousands of people with type 1 diabetes suffer at least one unpredicted episode of severe hypoglycemia per year in Korea. For patients with problematic hypoglycemia, an evidence-based stepwise approach was suggested in 2015. The first step is structured education regarding multiple daily injections of an insulin analog, and the second step is adding a technological intervention, such as continuous subcutaneous insulin infusion or real-time continuous glucose monitoring. The next step is a sensor-augmented pump, preferably with a low glucose suspension feature or very frequent contact, and the final step is islet or pancreas transplantation. In Korea, however, none of these treatments are reimbursed by the National Health Insurance, and thus have not been widely implemented. The low prevalence of type 1 diabetes means that Korean physicians are relatively unfamiliar with the new technologies in this field. Therefore, the roles of new technologies and pancreas or islet transplantation in the treatment of problematic hypoglycemia need to be defined in the current clinical setting of Korea.


Subject(s)
Humans , Education , Glucose , Hypoglycemia , Insulin , Islets of Langerhans Transplantation , Korea , National Health Programs , Pancreas , Pancreas Transplantation , Prevalence
20.
Article in English | WPRIM (Western Pacific) | ID: wpr-225713

ABSTRACT

Since the report of type 1 diabetes reversal in seven consecutive patients by the Edmonton protocol in 2000, pancreatic islet transplantation has been reappraised based on accumulated clinical evidence. Although initially expected to therapeutically target long-term insulin independence, islet transplantation is now indicated for more specific clinical benefits. With the long-awaited report of the first phase 3 clinical trial in 2016, allogeneic islet transplantation is now transitioning from an experimental to a proven therapy for type 1 diabetes with problematic hypoglycemia. Islet autotransplantation has already been therapeutically proven in chronic pancreatitis with severe abdominal pain refractory to conventional treatments, and it holds promise for preventing diabetes after partial pancreatectomy due to benign pancreatic tumors. Based on current evidence, this review focuses on islet transplantation as a realistic approach to treating diabetes.


Subject(s)
Humans , Abdominal Pain , Autografts , Hypoglycemia , Insulin , Islets of Langerhans , Islets of Langerhans Transplantation , Pancreatectomy , Pancreatitis, Chronic , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...