1
|
Goyal P, Malviya R. Stem Cell Therapy for the Management of Type 1 Diabetes: Advances and Perspectives. Endocr Metab Immune Disord Drug Targets 2024; 24:549-561. [PMID: 37861029 DOI: 10.2174/0118715303256582230919093535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/20/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Due to insulin resistance and excessive blood sugar levels, type 1 diabetes mellitus (T1DM) is characterized by pancreatic cell loss. This condition affects young people at a higher rate than any other chronic autoimmune disease. Regardless of the method, exogenous insulin cannot substitute for insulin produced by a healthy pancreas. An emerging area of medicine is pancreatic and islet transplantation for type 1 diabetics to restore normal blood sugar regulation. However, there are still obstacles standing in the way of the widespread use of these therapies, including very low availability of pancreatic and islets supplied from human organ donors, challenging transplantation conditions, high expenses, and a lack of easily accessible methods. Efforts to improve Type 1 Diabetes treatment have been conducted in response to the disease's increasing prevalence. Type 1 diabetes may one day be treated with stem cell treatment. Stem cell therapy has proven to be an effective treatment for type 1 diabetes. Recent progress in stem cell-based diabetes treatment is summarised, and the authors show how to isolate insulin-producing cells (IPCs) from a variety of progenitor cells.
Collapse
Affiliation(s)
- Priyanshi Goyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
2
|
Schomburg L. Selenium Deficiency Due to Diet, Pregnancy, Severe Illness, or COVID-19-A Preventable Trigger for Autoimmune Disease. Int J Mol Sci 2021; 22:8532. [PMID: 34445238 PMCID: PMC8395178 DOI: 10.3390/ijms22168532] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
The trace element selenium (Se) is an essential part of the human diet; moreover, increased health risks have been observed with Se deficiency. A sufficiently high Se status is a prerequisite for adequate immune response, and preventable endemic diseases are known from areas with Se deficiency. Biomarkers of Se status decline strongly in pregnancy, severe illness, or COVID-19, reaching critically low concentrations. Notably, these conditions are associated with an increased risk for autoimmune disease (AID). Positive effects on the immune system are observed with Se supplementation in pregnancy, autoimmune thyroid disease, and recovery from severe illness. However, some studies reported null results; the database is small, and randomized trials are sparse. The current need for research on the link between AID and Se deficiency is particularly obvious for rheumatoid arthritis and type 1 diabetes mellitus. Despite these gaps in knowledge, it seems timely to realize that severe Se deficiency may trigger AID in susceptible subjects. Improved dietary choices or supplemental Se are efficient ways to avoid severe Se deficiency, thereby decreasing AID risk and improving disease course. A personalized approach is needed in clinics and during therapy, while population-wide measures should be considered for areas with habitual low Se intake. Finland has been adding Se to its food chain for more than 35 years-a wise and commendable decision, according to today's knowledge. It is unfortunate that the health risks of Se deficiency are often neglected, while possible side effects of Se supplementation are exaggerated, leading to disregard for this safe and promising preventive and adjuvant treatment options. This is especially true in the follow-up situations of pregnancy, severe illness, or COVID-19, where massive Se deficiencies have developed and are associated with AID risk, long-lasting health impairments, and slow recovery.
Collapse
Affiliation(s)
- Lutz Schomburg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institut für Experimentelle Endokrinologie, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Hessische Straße 3-4, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
3
|
Sun SY, Gao Y, Liu GJ, Li YK, Gao W, Ran XW. Efficacy and Safety of Stem Cell Therapy for T1DM: An Updated Systematic Review and Meta-Analysis. J Diabetes Res 2020; 2020:5740923. [PMID: 33102605 PMCID: PMC7569432 DOI: 10.1155/2020/5740923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/20/2020] [Accepted: 09/17/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The long-term insulin therapy for type 1 diabetes mellitus (T1DM) fails to achieve optimal glycemic control and avoid adverse events simultaneously. Stem cells have unique immunomodulatory capacities and have been considered as a promising interventional strategy for T1DM. Stem cell therapy in T1DM has been tried in many studies. However, the results were controversial. We thus performed a meta-analysis to update the efficacy and safety of stem cell therapy in patients with T1DM. METHODS We systematically searched the Medline, EMBASE, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, Web of Science, Wan Fang Data, China National Knowledge Infrastructure, VIP database, and the Chinese Biomedical Literature Database (SinoMed) for relevant studies published before March 19, 2019. The outcomes included parameters for glycemic control (i.e., glycosylated hemoglobin (HbA1c) levels and insulin dosages), β cell function (i.e., fasting C-peptide levels and area-under-curve of C-peptide concentration (AUCC)), and relative risk of adverse events. Statistical analysis was conducted by using RevMan 5.3 and Stata 12.0. RESULTS Five randomized controlled trials (RCTs) and eight nonrandomized concurrent control trials (NRCCTs) with a total of 396 individuals were finally included into the meta-analysis. Among RCTs, stem cell therapy could significantly reduce HbA1c levels (MD = -1.20, 95% CI -1.91 to -0.49, P = 0.0009) and increase fasting C-peptide levels (MD = 0.25, 95% CI 0.04 to 0.45, P = 0.02) and AUCC (SMD = 0.66, 95% CI 0.13 to 1.18, P = 0.01). Stem cell therapy could also reduce insulin dosages (SMD = -2.65, 95% CI -4.86 to -0.45, P = 0.02) at 6 months after treatment. NRCCTs also had consistent results. Furthermore, RCTs showed stem cell therapy did not increase relative risk of gastrointestinal symptom (RR = 0.69, 95% CI 0.14 to 3.28, P = 0.64) and infection (RR = 0.97, 95% CI 0.40 to 2.34, P = 0.95). However, NRCCTs showed stem cell therapy increased relative risk of gastrointestinal symptom (RR = 44.49, 95% CI 9.20 to 215.18, P < 0.00001). CONCLUSION Stem cell therapy for T1DM may improve glycemic control and β cell function without increasing the risk of serious adverse events. Stem cell therapy may also have a short-term (3-6 months) effect on reducing insulin dosages.
Collapse
Affiliation(s)
- Shi-Yi Sun
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital Sichuan University, Chengdu, Sichuan 610041, China
| | - Yun Gao
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital Sichuan University, Chengdu, Sichuan 610041, China
| | - Guan-Jian Liu
- Chinese Cochrane Centre, Chengdu, Sichuan 610041, China
| | - Yong-Kun Li
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Gao
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital Sichuan University, Chengdu, Sichuan 610041, China
| | - Xing-Wu Ran
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
4
|
Eter WA, Van der Kroon I, Andralojc K, Buitinga M, Willekens SMA, Frielink C, Bos D, Joosten L, Boerman OC, Brom M, Gotthardt M. Non-invasive in vivo determination of viable islet graft volume by 111In-exendin-3. Sci Rep 2017; 7:7232. [PMID: 28775287 PMCID: PMC5543140 DOI: 10.1038/s41598-017-07815-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/30/2017] [Indexed: 01/20/2023] Open
Abstract
Pancreatic islet transplantation is a promising therapy for patients with type 1 diabetes. However, the duration of long-term graft survival is limited due to inflammatory as well as non-inflammatory processes and routine clinical tests are not suitable to monitor islet survival. 111In-exendin-SPECT (single photon emission computed tomography) is a promising method to non-invasively image islets after transplantation and has the potential to help improve the clinical outcome. Whether 111In-exendin-SPECT allows detecting small differences in beta-cell mass (BCM) and measuring the actual volume of islets that were successfully engrafted has yet to be demonstrated. Here, we evaluated the performance of 111In-exendin-SPECT using an intramuscular islet transplantation model in C3H mice. In vivo imaging of animals transplanted with 50, 100, 200, 400 and 800 islets revealed an excellent linear correlation between SPECT quantification of 111In-exendin uptake and insulin-positive area of islet transplants, demonstrating that 111In-exendin-SPECT specifically and accurately measures BCM. The high sensitivity of the method allowed measuring small differences in graft volumes, including grafts that contained less than 50 islets. The presented method is reliable, convenient and holds great potential for non-invasive monitoring of BCM after islet transplantation in humans.
Collapse
Affiliation(s)
- Wael A Eter
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Inge Van der Kroon
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Karolina Andralojc
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mijke Buitinga
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Stefanie M A Willekens
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Cathelijne Frielink
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Desiree Bos
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Maarten Brom
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Penaforte-Saboia JG, Montenegro RM, Couri CE, Batista LA, Montenegro APDR, Fernandes VO, Akhtar H, Negrato CA, Malmegrim KCR, Moraes DA, Dias JBE, Simões BP, Gomes MB, Oliveira MC. Microvascular Complications in Type 1 Diabetes: A Comparative Analysis of Patients Treated with Autologous Nonmyeloablative Hematopoietic Stem-Cell Transplantation and Conventional Medical Therapy. Front Endocrinol (Lausanne) 2017; 8:331. [PMID: 29218029 PMCID: PMC5703738 DOI: 10.3389/fendo.2017.00331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To explore the impact on microvascular complications, long-term preservation of residual B-cell function and glycemic control of patients with type 1 diabetes treated with autologous nonmyeloablative hematopoietic stem-cell transplantation (AHST) compared with conventional medical therapy (CT). RESEARCH DESIGN AND METHODS Cross-sectional data of patients treated with AHST were compared with patients who received conventional therapy from the Brazilian Type 1 Diabetes Study Group, the largest multicenter observational study in type 1 diabetes mellitus in Brazil. Both groups of patients had diabetes for 8 years on average. An assessment comparison was made on the presence of microvascular complications, residual function of B cell, A1c, and insulin dose of the patients. RESULTS After a median of 8 years of diagnosis, none of the AHST-treated patients (n = 24) developed microvascular complications, while 21.5% (31/144) had at least one (p < 0.005) complication in the CT group (n = 144). Furthermore, no case of nephropathy was reported in the AHST group, while 13.8% of CT group (p < 0.005) developed nephropathy during the same period. With regard of residual B-cell function, the percentage of individuals with predicted higher C-peptide levels (IDAA1C ≤ 9) was about 10-fold higher in the AHST group compared with CT (75 vs. 8.3%) (p < 0.001) group. Among AHST patients, 54.1% (13/24) had the HbA1c < 7.0 compared with 13.1% in the CT (p < 0.001) group. CONCLUSION Patients with newly diagnosed type 1 diabetes treated with AHST presented lower prevalence of microvascular complications, higher residual B-cell function, and better glycemic control compared with the CT group.
Collapse
Affiliation(s)
| | - Renan M. Montenegro
- Post Graduate Program in Medical Sciences, Federal University of Ceará, Ceará, Brazil
- Federal University of Ceará, Ceará, Brazil
- *Correspondence: Renan M. Montenegro Jr.,
| | - Carlos E. Couri
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Livia A. Batista
- Post Graduate Program in Medical Sciences, Federal University of Ceará, Ceará, Brazil
| | | | | | - Hussain Akhtar
- Federal University of Ceará, Ceará, Brazil
- University of Oslo, UIO, Oslo, Noruega
| | | | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Aparecida Moraes
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana B. E. Dias
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Belinda P. Simões
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
6
|
Vanikar AV, Trivedi HL, Thakkar UG. Stem cell therapy emerging as the key player in treating type 1 diabetes mellitus. Cytotherapy 2016; 18:1077-86. [PMID: 27424148 DOI: 10.1016/j.jcyt.2016.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/24/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease causing progressive destruction of pancreatic β cells, ultimately resulting in loss of insulin secretion producing hyperglycemia usually affecting children. Replacement of damaged β cells by cell therapy can treat it. Currently available strategies are insulin replacement and islet/pancreas transplantation. Unfortunately these offer rescue for variable duration due to development of autoantibodies. For pancreas/islet transplantation a deceased donor is required and various shortfalls of treatment include quantum, cumbersome technique, immune rejection and limited availability of donors. Stem cell therapy with assistance of cellular reprogramming and β-cell regeneration can open up new therapeutic modalities. The present review describes the history and current knowledge of T1DM, evolution of cell therapies and different cellular therapies to cure this condition.
Collapse
Affiliation(s)
- Aruna V Vanikar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India.
| | - Hargovind L Trivedi
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Nephrology and Transplantation Medicine, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| | - Umang G Thakkar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| |
Collapse
|
7
|
Abstract
There is a clear need to develop strategies to induce tolerance without the need of chronic immunosuppression in transplant recipient and in patients with autoimmunity. Adoptive T regulatory cell (Treg) therapy offers the potential of long-lasting protection. However, based on results of clinical trials so far with ex vivo expanded autologous Tregs in type 1 diabetic (T1D) patients, it seems unlikely that single immunotherapy with Treg infusion without immunomodulation regimens that promote stable donor Treg engraftment and persistence would afford truly significant clinical benefit. Combination therapies could provide improved outcomes with consideration of the fundamental factors required for Treg generation, homeostasis, and function to promote long-term donor Treg persistence to provoke beneficial therapeutic outcomes.
Collapse
|
8
|
Askenasy N. Less Is More: The Detrimental Consequences of Immunosuppressive Therapy in the Treatment of Type-1 Diabetes. Int Rev Immunol 2015; 34:523-37. [DOI: 10.3109/08830185.2015.1010723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
Walter J, Harder O, Faendrich F, Schulze M. Generation of monocyte-derived insulin-producing cells from non-human primates according to an optimized protocol for the generation of PCMO-derived insulin-producing cells. J Clin Res Pediatr Endocrinol 2014; 6:93-9. [PMID: 24932602 PMCID: PMC4141582 DOI: 10.4274/jcrpe.1284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE The vision of potential autologous cell therapy for the cure of diabetes encourages ongoing research. According to a previously published protocol for the generation of insulin-producing cells from human monocytes, we analyzed whether the addition of growth factors could increase insulin production. This protocol was then transferred to a non-human primate model by using either blood- or spleen-derived monocytes. METHODS Human monocytes were treated to dedifferentiate into programmable cells of monocytic origin (PCMO). In addition to the published protocol, PCMOs were then treated with either activin A, betacellulin, exendin 3 or 4. Cells were characterized by protein expression of insulin, Pdx-1, C-peptide and Glut-2. After identifying the optimal protocol, monocytes from baboon blood were isolated and the procedure was repeated. Spleen monocytes following splenectomy of a live baboon were differentiated and analyzed in the same manner and calculated in number and volume. RESULTS Insulin content of human cells was highest when cells were treated with activin A and their insulin content was 13,000 µU/1 million cells. Insulin-producing cells form primate monocytes could successfully be generated despite using human growth factors and serum. Expression of insulin, Pdx-1, C-peptide and Glut-2 was comparable to that of human neo-islets. Total insulin content of activin A-treated baboon monocytes was 16,000 µU/1 million cells. CONCLUSION We were able to show that insulin-producing cells can be generated from baboon monocytes with human growth factors. The amount generated from one spleen could be enough to cure a baboon from experimentally induced diabetes in an autologous cell transplant setting.
Collapse
Affiliation(s)
- Jessica Walter
- University Hospital Essen, Department of General, Visceral and Transplantation Surgery, Hufelandstrasse 55, 45147 Essen, Germany. E-ma-il:
| | - Ole Harder
- Schleswig Holsetin University Hospital, Campus Kiel, Department of General, Thoracic, Transplantation and Pediatric Surgery, Kiel, Germany
| | - Fred Faendrich
- Schleswig Holsetin University Hospital, Campus Kiel, Department of General, Thoracic, Transplantation and Pediatric Surgery, Kiel, Germany
| | - Maren Schulze
- University Hospital Essen, Department of General, Visceral and Transplantation Surgery, Essen, Germany
,* Address for Correspondence: University Hospital Essen, Department of General, Visceral and Transplantation Surgery, Hufelandstrasse 55, 45147 Essen, Germany Phone: +4920172384003 E-mail:
| |
Collapse
|
10
|
Chhabra P, Brayman KL. Stem cell therapy to cure type 1 diabetes: from hype to hope. Stem Cells Transl Med 2013; 2:328-336. [PMID: 23572052 PMCID: PMC3667565 DOI: 10.5966/sctm.2012-0116] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 02/01/2013] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic, multifactorial autoimmune disease that involves the progressive destruction of pancreatic β-cells, ultimately resulting in the loss of insulin production and secretion. The goal of clinical intervention is to prevent or arrest the onset and progression of autoimmunity, reverse β-cell destruction, and restore glycometabolic and immune homeostasis. Despite promising outcomes observed with islet transplantation and advancements in immunomodulatory therapies, the need for an effective cell replacement strategy for curing T1D still persists. Stem cell therapy offers a solution to the cited challenges of islet transplantation. While the regenerative potential of stem cells can be harnessed to make available a self-replenishing supply of glucose-responsive insulin-producing cells, their immunomodulatory properties may potentially be used to prevent, arrest, or reverse autoimmunity, ameliorate innate/alloimmune graft rejection, and prevent recurrence of the disease. Herein, we discuss the therapeutic potential of stem cells derived from a variety of sources for the cure of T1D, for example, embryonic stem cells, induced pluripotent stem cells, bone marrow-derived hematopoietic stem cells, and multipotent mesenchymal stromal cells derived from bone marrow, umbilical cord blood, and adipose tissue. The benefits of combinatorial approaches designed to ensure the successful clinical translation of stem cell therapeutic strategies, such as approaches combining effective stem cell strategies with islet transplantation, immunomodulatory drug regimens, and/or novel bioengineering techniques, are also discussed. To conclude, the application of stem cell therapy in the cure for T1D appears extremely promising.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kenneth L. Brayman
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
11
|
Bevier WC, Trujillo AL, Primbs GB, Bradley MK, Jovanovič L. Oral anti-CD3 monoclonal antibody delays diabetes in non-obese diabetic (NOD) mice: effects on pregnancy and offspring--a preliminary report. Diabetes Metab Res Rev 2011; 27:480-7. [PMID: 21484981 DOI: 10.1002/dmrr.1204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The objective was to observe the effect of oral anti-CD3 monoclonal antibody (mAb) on non-obese diabetic mice, pregnancy, and offspring. METHODS Three protocols are classified as: (1) Twenty non-obese diabetic/ShiLtJ female mice were monitored for type 1 diabetes mellitus. If the blood glucose level was ≥ 250 mg/dL, the mice were treated for 8 days with either 50 or 100 µg oral anti-CD3 monoclonal antibody. If the diabetes resolved, the mice were bred. (2) F1 offspring were monitored for diabetes; 15 female mice became diabetic. Non-diabetic F1 female mice were divided into two groups [ten antibody (Ab) and ten control (C)] and bred. Ab female mice were given 100 µg monoclonal antibody before diabetes development and during pregnancy for 6 weeks. (3) Twenty-five F2 Ab and 23 F2 C mice were monitored. At 15-17 weeks, Ab mice, both female and male, were given 100 µg monoclonal antibody for 8 weeks. RESULTS (1) The diabetes in four female mice treated with 50 µg did not resolve; in three of the six diabetic female mice treated with 100 µg, the diabetes resolved and the mice were bred. The remaining ten non-diabetic female mice were given 100 µg oral monoclonal antibody and then bred. No diabetes developed during pregnancy; 13 litters were born. (2) Three diabetic Ab female mice sustained their pregnancies versus one diabetic C female mouse (p ≤ 0.05). (3) At 30 weeks, six Ab female and three Ab male mice and seven C female and three C male mice developed diabetes. The number of diabetic Ab and C mice was not different; diagnosis age was significantly different-Ab 23.3 ± 5.1 and C 18.8 ± 3.7 weeks (p ≤ 0.05). CONCLUSIONS In female non-obese diabetic mice, oral anti-CD3 monoclonal antibody was effective in reversing diabetes and allowing pregnancy and extending longevity, but it did not prevent diabetes in the offspring.
Collapse
Affiliation(s)
- Wendy C Bevier
- Sansum Diabetes Research Institute, Santa Barbara, CA 93105, USA.
| | | | | | | | | |
Collapse
|
12
|
Intramuscular delivery of a naked DNA plasmid encoding proinsulin and pancreatic regenerating III protein ameliorates type 1 diabetes mellitus. Pharmacol Res 2010; 63:320-7. [PMID: 21185938 DOI: 10.1016/j.phrs.2010.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 12/20/2010] [Indexed: 01/13/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by inflammation of pancreatic islets and destruction of β cells. Up to now, there is still no cure for this devastating disease and alternative approach should be developed. To explore a novel gene therapy strategy combining immunotherapy and β cell regeneration, we constructed a non-viral plasmid encoding proinsulin (PI) and pancreatic regenerating (Reg) III protein (pReg/PI). Therapeutic potentials of this plasmid for T1DM were investigated. Intramuscular delivery of pReg/PI resulted in a significant reduction in hyperglycemia and diabetes incidence, with an increased insulin contents in the serum of T1DM mice model induced by STZ. Treatment with pReg/PI also restored the balance of Th1/Th2 cytokines and expanded CD4(+)CD25(+)Foxp3(+) T regulatory cells, which may attribute to the establishment of self-immune tolerance. Additionally, in comparison to the mice treated with empty vector pBudCE4.1 (pBud), attenuated insulitis and apoptosis achieved by inhibiting activation of NF-κB in the pancreas of pReg/PI treated mice were observed. In summary, these results indicate that intramuscular delivery of pReg/PI distinctly ameliorated STZ-induced T1DM by reconstructing the immunological self-tolerance and promoting the regeneration of β cells, which might be served as a promising candidate for the gene therapy of T1DM.
Collapse
|