1
|
Frontino G, Stancampiano MR, Aiuti A. Potentialities of Gene Therapy in Pediatric Endocrinology. Horm Res Paediatr 2021; 96:646-657. [PMID: 34801996 DOI: 10.1159/000520965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/09/2021] [Indexed: 11/19/2022] Open
Abstract
Gene therapy has become an appealing therapeutic option in many pediatric fields, including endocrinology. Unlike traditional drugs based on molecules that require repeated and frequent burdensome administrations, a single genetic therapeutic intervention may allow durable and curative clinical benefits. Although this highly innovative technology holds a great promise for the treatment of monogenic diseases, its clinical applications in the field of endocrinology have been so far challenging. In this review, we will discuss various ex vivo and in vivo approaches and potential applications of gene addition and gene editing approaches for treating hyperfunctional and hypofunctional endocrine diseases due to intrinsic defects or autoimmune origin. We will focus on the recent advances in gene therapy approaches aimed at treating type 1 diabetes and monogenic forms of endocrinopathies such as growth hormone deficiency, congenital adrenal hyperplasia, diabetes insipidus, IPEX, as well as their trends and future directions.
Collapse
Affiliation(s)
- Giulio Frontino
- Department of Pediatrics, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Pediatrics, Pediatric Immunohematology Unit, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
2
|
Akil AAS, Yassin E, Al-Maraghi A, Aliyev E, Al-Malki K, Fakhro KA. Diagnosis and treatment of type 1 diabetes at the dawn of the personalized medicine era. J Transl Med 2021; 19:137. [PMID: 33794915 PMCID: PMC8017850 DOI: 10.1186/s12967-021-02778-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Type 1 diabetes affects millions of people globally and requires careful management to avoid serious long-term complications, including heart and kidney disease, stroke, and loss of sight. The type 1 diabetes patient cohort is highly heterogeneous, with individuals presenting with disease at different stages and severities, arising from distinct etiologies, and overlaying varied genetic backgrounds. At present, the “one-size-fits-all” treatment for type 1 diabetes is exogenic insulin substitution therapy, but this approach fails to achieve optimal blood glucose control in many individuals. With advances in our understanding of early-stage diabetes development, diabetes stratification, and the role of genetics, type 1 diabetes is a promising candidate for a personalized medicine approach, which aims to apply “the right therapy at the right time, to the right patient”. In the case of type 1 diabetes, great efforts are now being focused on risk stratification for diabetes development to enable pre-clinical detection, and the application of treatments such as gene therapy, to prevent pancreatic destruction in a sub-set of patients. Alongside this, breakthroughs in stem cell therapies hold great promise for the regeneration of pancreatic tissues in some individuals. Here we review the recent initiatives in the field of personalized medicine for type 1 diabetes, including the latest discoveries in stem cell and gene therapy for the disease, and current obstacles that must be overcome before the dream of personalized medicine for all type 1 diabetes patients can be realized.
Collapse
Affiliation(s)
- Ammira Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| | - Esraa Yassin
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Aljazi Al-Maraghi
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Elbay Aliyev
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Khulod Al-Malki
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Khalid A Fakhro
- Department of Human Genetics-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.,Department of Genetic Medicine, Weill Cornell Medicine, P.O. Box 24144, Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| |
Collapse
|
3
|
Benskey MJ, Sandoval IM, Miller K, Sellnow RL, Gezer A, Kuhn NC, Vashon R, Manfredsson FP. Basic Concepts in Viral Vector-Mediated Gene Therapy. Methods Mol Biol 2019; 1937:3-26. [PMID: 30706387 DOI: 10.1007/978-1-4939-9065-8_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Today any researcher with the desire can easily purchase a viral vector. However, despite the availability of viral vectors themselves, the requisite knowledge that is absolutely essential to conducting a gene therapy experiment remains somewhat obscure and esoteric. To utilize viral vectors to their full potential, a large number of decisions must be made, in some instances prior to even obtaining the vector itself. For example, critical decisions include selection of the proper virus, selection of the proper expression cassette, whether to produce or purchase a viral vector, proper viral handling and storage, the most appropriate delivery method, selecting the proper controls, how to ensure your virus is expressing properly, and many other complex decisions that are essential to performing a successful gene therapy experiment. The need to make so many important decisions can be overwhelming and potentially prohibitive, especially to the novice gene therapist. In order to aid in this challenging process, here we provide an overview of basic gene therapy modalities and a decision tree that can be used to make oneself aware of the options available to the beginning gene therapist. This information can be used as a road map to help navigate the complex and perhaps confusing process of designing a successful gene therapy experiment.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Ivette M Sandoval
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Mercy Health Saint Mary's, Grand Rapids, MI, USA
| | - Kathryn Miller
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Rhyomi L Sellnow
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Aysegul Gezer
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Roslyn Vashon
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
- Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
4
|
Gheflat S, Sadeghi A, Bandehpour M, Ramezani K, Kazemi B. Designing an Engineered Construct Gene Sensitive to Carbohydrate In-vitro and Candidate for Human Insulin Gene Therapy In-vivo. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2019; 18:2111-2116. [PMID: 32184874 PMCID: PMC7059050 DOI: 10.22037/ijpr.2019.14650.12567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes is a common disorder worldwide, and exhaustive efforts have been made to cure this disease. Gene therapy has been considered as a potential curative method that has had more stability in comparison with other pharmaceutical methods. However, the application of gene therapy as a definitive treatment demands further investigation. This study is aimed to prepare a suitable high- performance vector for gene therapy in diabetes mellitus. The designed vector has had prominent characteristics, such as directed replacement, which makes it a suitable method for treating or preventing other genetic disorders. The whole rDNA sequence of the human genome was scanned. The 800 bp two homology arms were digested by EcoRI, synthesized and cloned into the pGEM-B1 plasmid (prokaryotic moiety). The carbohydrate sensitive promoter, L-pyruvate kinase, and insulin gene were sub-cloned between homologous arms (eukaryotic moiety). The PGEM-B1 plasmid was digested by EcoRI, and the eukaryotic fragments were purified and transfected into Hela cell and then cultured. Afterward, the 300 µg/mL of glucose were added to the culture medium. Insulin expression in the transfected cells with 200 and 400 ng of the construct in comparison with negative control was detected using western blot and ELISA methods. Results have shown insulin expression in different glucose concentrates.
Collapse
Affiliation(s)
- Shivasadat Gheflat
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abdolrahim Sadeghi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojgan Bandehpour
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Keyvan Ramezani
- Departement of Parasitology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Departement of Parasitology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Chellappan DK, Sivam NS, Teoh KX, Leong WP, Fui TZ, Chooi K, Khoo N, Yi FJ, Chellian J, Cheng LL, Dahiya R, Gupta G, Singhvi G, Nammi S, Hansbro PM, Dua K. Gene therapy and type 1 diabetes mellitus. Biomed Pharmacother 2018; 108:1188-1200. [PMID: 30372820 DOI: 10.1016/j.biopha.2018.09.138] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is an autoimmune disorder characterized by T cell-mediated self-destruction of insulin-secreting islet β cells. Management of T1DM is challenging and complicated especially with conventional medications. Gene therapy has emerged as one of the potential therapeutic alternatives to treat T1DM. This review primarily focuses on the current status and the future perspectives of gene therapy in the management of T1DM. A vast number of the studies which are reported on gene therapy for the management of T1DM are done in animal models and in preclinical studies. In addition, the safety of such therapies is yet to be established in humans. Currently, there are several gene level interventions that are being investigated, notably, overexpression of genes and proteins needed against T1DM, transplantation of cells that express the genes against T1DM, stem-cells mediated gene therapy, genetic vaccination, immunological precursor cell-mediated gene therapy and vectors. METHODS We searched the current literature through searchable online databases, journals and other library sources using relevant keywords and search parameters. Only relevant publications in English, between the years 2000 and 2018, with evidences and proper citations, were considered. The publications were then analyzed and segregated into several subtopics based on common words and content. A total of 126 studies were found suitable for this review. FINDINGS Generally, the pros and cons of each of the gene-based therapies have been discussed based on the results collected from the literature. However, there are certain interventions that require further detailed studies to ensure their effectiveness. We have also highlighted the future direction and perspectives in gene therapy, which, researchers could benefit from.
Collapse
Affiliation(s)
- Dinesh Kumar Chellappan
- Department of Life Sciences, International Medical University, Kuala Lumpur, 57000, Malaysia.
| | - Nandhini S Sivam
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Kai Xiang Teoh
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Wai Pan Leong
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Tai Zhen Fui
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Kien Chooi
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Nico Khoo
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Fam Jia Yi
- School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Lim Lay Cheng
- Department of Life Sciences, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Rajiv Dahiya
- Laboratory of Peptide Research and Development, School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017, Jaipur, India.
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology & Science (BITS), Pilani, Pilani Campus, 333031, Rajasthan, India
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, NSW, 2751, Australia; NICM Health Research Institute, Western Sydney University, NSW, 2751, Australia
| | - Philip Michael Hansbro
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW, 2007, Australia; School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia & Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW, 2007, Australia; School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia & Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia; School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| |
Collapse
|
6
|
Jaén ML, Vilà L, Elias I, Jimenez V, Rodó J, Maggioni L, Ruiz-de Gopegui R, Garcia M, Muñoz S, Callejas D, Ayuso E, Ferré T, Grifoll I, Andaluz A, Ruberte J, Haurigot V, Bosch F. Long-Term Efficacy and Safety of Insulin and Glucokinase Gene Therapy for Diabetes: 8-Year Follow-Up in Dogs. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017. [PMID: 28626777 PMCID: PMC5466581 DOI: 10.1016/j.omtm.2017.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Diabetes is a complex metabolic disease that exposes patients to the deleterious effects of hyperglycemia on various organs. Achievement of normoglycemia with exogenous insulin treatment requires the use of high doses of hormone, which increases the risk of life-threatening hypoglycemic episodes. We developed a gene therapy approach to control diabetic hyperglycemia based on co-expression of the insulin and glucokinase genes in skeletal muscle. Previous studies proved the feasibility of gene delivery to large diabetic animals with adeno-associated viral (AAV) vectors. Here, we report the long-term (∼8 years) follow-up after a single administration of therapeutic vectors to diabetic dogs. Successful, multi-year control of glycemia was achieved without the need of supplementation with exogenous insulin. Metabolic correction was demonstrated through normalization of serum levels of fructosamine, triglycerides, and cholesterol and remarkable improvement in the response to an oral glucose challenge. The persistence of vector genomes and therapeutic transgene expression years after vector delivery was documented in multiple samples from treated muscles, which showed normal morphology. Thus, this study demonstrates the long-term efficacy and safety of insulin and glucokinase gene transfer in large animals and especially the ability of the system to respond to the changes in metabolic needs as animals grow older.
Collapse
Affiliation(s)
- Maria Luisa Jaén
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Laia Vilà
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Ivet Elias
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Veronica Jimenez
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Jordi Rodó
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Luca Maggioni
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Rafael Ruiz-de Gopegui
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Miguel Garcia
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Sergio Muñoz
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - David Callejas
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Eduard Ayuso
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Tura Ferré
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Iris Grifoll
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Anna Andaluz
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jesus Ruberte
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| |
Collapse
|
7
|
Thulé PM, Lin Y, Jia D, Olson DE, Tang SC, Sambanis A. mRNA destabilization improves glycemic responsiveness of transcriptionally regulated hepatic insulin gene therapy in vitro and in vivo. J Gene Med 2017; 19. [PMID: 28181342 DOI: 10.1002/jgm.2946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Hepatic insulin gene therapy (HIGT) employing a glucose and insulin sensitive promoter to direct insulin transcription can lower blood sugars within 2 h of an intraperitoneal glucose challenge. However, post-challenge blood sugars frequently decline to below baseline. We hypothesize that this 'over-shoot' hypoglycemia results from sustained translation of long-lived transgene message, and that reducing pro-insulin message half-life will ameliorate post-challenge hypoglycemia. METHODS We compared pro-insulin message content and insulin secretion from primary rat hepatocytes expressing insulin from either a standard construct (2xfur), or a construct producing a destabilized pro-insulin message (InsTail), following exposure to stimulating or inhibitory conditions. RESULTS Hepatocytes transduced with a 2xfur construct accumulated pro-insulin message, and exhibited increased insulin secretion, under conditions that both inhibit or stimulate transcription. By contrast, pro-insulin message content remained stable in InsTail expressing cells, and insulin secretion increased less than 2xfur during prolonged stimulation. During transitions from stimulatory to inhibitory conditions, or vice versa, amounts of pro-insulin message changed more rapidly in InsTail expressing cells than 2xfur expressing cells. Importantly, insulin secretion increased during the transition from stimulation to inhibition in 2xfur expressing cells, although it remained unchanged in InsTail expressing cells. Use of the InsTail destabilized insulin message tended to more rapidly reduce glucose induced glycemic excursions, and limit post-load hypoglycemia in STZ-diabetic mice in vivo. CONCLUSIONS The data obtained in the present study suggest that combining transcriptional and post-transcriptional regulatory strategies may reduce undesirable glycemic excursion in models of HIGT.
Collapse
Affiliation(s)
- Peter M Thulé
- Atlanta VA Medical Center, Division of Endocrinology, Diabetes, & Lipids, Emory University School of Medicine, Decatur, Georgia, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Yulin Lin
- Atlanta VA Medical Center, Division of Endocrinology, Diabetes, & Lipids, Emory University School of Medicine, Decatur, Georgia, USA
| | - Dingwu Jia
- Atlanta VA Medical Center, Division of Endocrinology, Diabetes, & Lipids, Emory University School of Medicine, Decatur, Georgia, USA
| | - Darin E Olson
- Atlanta VA Medical Center, Division of Endocrinology, Diabetes, & Lipids, Emory University School of Medicine, Decatur, Georgia, USA
| | - Shiue-Cheng Tang
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Department of Medical Science, National Tsing Hua University, Taiwan, USA
| | - Athanassios Sambanis
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Qu S, Zhang T, Dong HH. Effect of hepatic insulin expression on lipid metabolism in diabetic mice. J Diabetes 2016; 8:314-23. [PMID: 25851734 DOI: 10.1111/1753-0407.12293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/27/2015] [Accepted: 03/21/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Hypertriglyceridemia is a common lipid disorder that is characterized by elevated plasma levels of triglyceride (TG)-rich particles, such as very low-density lipoprotein (VLDL), in poorly controlled diabetes. The aim of the present study was to determine the potential therapeutic effect of hepatic insulin production on hypertriglyceridemia in mice. METHODS Mice were induced diabetic and hypertriglyceridemic by streptozotocin (STZ) treatment. Using an adenovirus-mediated gene transfer approach, we delivered rat preproinsulin cDNA into the liver of diabetic mice and then determined plasma TG metabolism. To investigate the mechanism by which hepatic insulin improves TG metabolism, we determined hepatic expression of apolipoprotein C-III (ApoC-III), a structural moiety and functional inhibitor of VLDL-TG catabolism. RESULTS Plasma VLDL-TG levels were markedly elevated in STZ-treated mice, and were accompanied by hyperglycemia and hypertriglyceridemia. These metabolic abnormalities were restored to near normal following hepatic insulin production in insulin vector-treated diabetic mice. In contrast, hypertriglyceridemia and hyperglycemia persisted in control vector-treated diabetic animals. Hepatic ApoC-III expression became deregulated secondary to insulin deficiency, contributing to impaired TG metabolism in diabetic mice. Hepatic insulin production suppressed excessive hepatic ApoC-III production to basal levels. CONCLUSION Hepatic insulin production is efficacious in correcting hypertriglyceridemia associated with insulin deficiency in diabetic mice.
Collapse
Affiliation(s)
- Shen Qu
- Department of Endocrinology & Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ting Zhang
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - H Henry Dong
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Hong YH, Betik AC, Premilovac D, Dwyer RM, Keske MA, Rattigan S, McConell GK. No effect of NOS inhibition on skeletal muscle glucose uptake during in situ hindlimb contraction in healthy and diabetic Sprague-Dawley rats. Am J Physiol Regul Integr Comp Physiol 2015; 308:R862-71. [DOI: 10.1152/ajpregu.00412.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 03/10/2015] [Indexed: 11/22/2022]
Abstract
Nitric oxide (NO) has been shown to be involved in skeletal muscle glucose uptake during contraction/exercise, especially in individuals with Type 2 diabetes (T2D). To examine the potential mechanisms, we examined the effect of local NO synthase (NOS) inhibition on muscle glucose uptake and muscle capillary blood flow during contraction in healthy and T2D rats. T2D was induced in Sprague-Dawley rats using a combined high-fat diet (23% fat wt/wt for 4 wk) and low-dose streptozotocin injections (35 mg/kg). Anesthetized animals had one hindlimb stimulated to contract in situ for 30 min (2 Hz, 0.1 ms, 35 V) with the contralateral hindlimb rested. After 10 min, the NOS inhibitor, NG-nitro-l-arginine methyl ester (l-NAME; 5 μM) or saline was continuously infused into the femoral artery of the contracting hindlimb until the end of contraction. Surprisingly, there was no increase in skeletal muscle NOS activity during contraction in either group. Local NOS inhibition had no effect on systemic blood pressure or muscle contraction force, but it did cause a significant attenuation of the increase in femoral artery blood flow in control and T2D rats. However, NOS inhibition did not attenuate the increase in muscle capillary recruitment during contraction in these rats. Muscle glucose uptake during contraction was significantly higher in T2D rats compared with controls but, unlike our previous findings in hooded Wistar rats, NOS inhibition had no effect on glucose uptake during contraction. In conclusion, NOS inhibition did not affect muscle glucose uptake during contraction in control or T2D Sprague-Dawley rats, and this may have been because there was no increase in NOS activity during contraction.
Collapse
Affiliation(s)
- Yet Hoi Hong
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise and Active Living, Victoria University, Melbourne, Victoria, Australia
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Andrew C. Betik
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise and Active Living, Victoria University, Melbourne, Victoria, Australia
| | - Dino Premilovac
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia; and
| | - Renee M. Dwyer
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia; and
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Michelle A. Keske
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia; and
| | - Stephen Rattigan
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia; and
| | - Glenn K. McConell
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise and Active Living, Victoria University, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Handorf AM, Sollinger HW, Alam T. Genetic Engineering of Surrogate <i>β</i> Cells for Treatment of Type 1 Diabetes Mellitus. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/jdm.2015.54037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
|
12
|
Liu YY, Jia W, Wanke IE, Muruve DA, Xiao HP, Wong NCW. Glucose regulates secretion of exogenously expressed insulin from HepG2 cells in vitro and in a mouse model of diabetes mellitus in vivo. J Mol Endocrinol 2013; 50:337-46. [PMID: 23475748 DOI: 10.1530/jme-12-0239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glucose-controlled insulin secretion is a key component of its regulation. Here, we examined whether liver cell secretion of insulin derived from an engineered construct can be regulated by glucose. Adenovirus constructs were designed to express proinsulin or mature insulin containing the conditional binding domain (CBD). This motif binds GRP78 (HSPA5), an endoplasmic reticulum (ER) protein that enables the chimeric hormone to enter into and stay within the ER until glucose regulates its release from the organelle. Infected HepG2 cells expressed proinsulin mRNA and the protein containing the CBD. Immunocytochemistry studies suggested that GRP78 and proinsulin appeared together in the ER of the cell. The amount of hormone released from infected cells varied directly with the ambient concentration of glucose in the media. Glucose-regulated release of the hormone from infected cells was rapid and sustained. Removal of glucose from the cells decreased release of the hormone. In streptozotocin-induced diabetic mice, when infected with adenovirus expressing mature insulin, glucose levels declined. Our data show that glucose regulates release of exogenously expressed insulin from the ER of liver cells. This approach may be useful in devising new ways to treat diabetes mellitus.
Collapse
Affiliation(s)
- Y Y Liu
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | | | | | | | | | | |
Collapse
|
13
|
Jajarmi V, Bandehpour M, Kazemi B. Regulation of insulin biosynthesis in non-beta cells by a heat shock promoter. J Biosci Bioeng 2013; 116:147-51. [PMID: 23541501 DOI: 10.1016/j.jbiosc.2013.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/26/2013] [Accepted: 02/21/2013] [Indexed: 12/23/2022]
Abstract
Insulin production under the stringent control is the main issue in gene-based therapeutic strategies directed to type 1 diabetes. As a novel approach, inducible promoters may provide a promising tool for this purpose. In this study, we hypothesize that this control may be achieved via a promoter derived from the heat shock multigene family, Hsp70 A1A, which is inducible at 42°C. To yield mature insulin in transfected fibroblasts (3T3/NIH), a recombinant human insulin gene consisting of sequences corresponding to furin cleavable sites was fused to the promoter. Heat-stimulated cells initiated to release biologically active insulin within 30 min with a ten-fold increase after 24 h. The role of upstream regulatory elements of the promoter on its activity in heat stress conditions was examined. No significant difference between the activity of the minimal and full-length promoters was observed. This promoter exhibited low basal expression in non-inducing conditions. Results indicate that this promoter is responsive to a heat induction after approximately 30 min which causes an efficient insulin production over a relatively short period of time. These potential features of this promoter may provide an insight to control the insulin production in vivo upon an external and physical stimulation.
Collapse
Affiliation(s)
- Vahid Jajarmi
- Department of Medical Biotechnology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
14
|
Abstract
Current therapies for the treatment of type 1 diabetes include daily administration of exogenous insulin and, less frequently, whole-pancreas or islet transplantation. Insulin injections often result in inaccurate insulin doses, exposing the patient to hypo- and/or hyperglycemic episodes that lead to long-term complications. Islet transplantation is also limited by lack of high-quality islet donors, early graft failure, and chronic post-transplant immunosuppressive treatment. These barriers could be circumvented by designing a safe and efficient strategy to restore insulin production within the patient's body. Porcine islets have been considered as a possible alternative source of transplantable insulin-producing cells to replace human cadaveric islets. More recently, embryonic or induced pluripotent stem cells have also been examined for their ability to differentiate in vitro into pancreatic endocrine cells. Alternatively, it may be feasible to generate new β-cells by ectopic expression of key transcription factors in endogenous non-β-cells. Finally, engineering surrogate β-cells by in vivo delivery of the insulin gene to specific tissues is also being studied as a possible therapy for type 1 diabetes. In the present review, we discuss these different approaches to restore insulin production.
Collapse
Affiliation(s)
- Eva Tudurí
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
15
|
Han J, Kim EH, Choi W, Jun HS. Glucose-responsive artificial promoter-mediated insulin gene transfer improves glucose control in diabetic mice. World J Gastroenterol 2012; 18:6420-6426. [PMID: 23197887 PMCID: PMC3508636 DOI: 10.3748/wjg.v18.i44.6420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of insulin gene therapy using a glucose-responsive synthetic promoter in type 2 diabetic obese mice.
METHODS: We employed a recently developed novel insulin gene therapy strategy using a synthetic promoter that regulates insulin gene expression in the liver in response to blood glucose level changes. We intravenously administered a recombinant adenovirus expressing furin-cleavable rat insulin under the control of the synthetic promoter (rAd-SP-rINSfur) into diabetic Leprdb/db mice. A recombinant adenovirus expressing β-galactosidase under the cytomegalovirus promoter was used as a control (rAd-CMV-βgal). Blood glucose levels and body weights were monitored for 50 d. Glucose and insulin tolerance tests were performed. Immunohistochemical staining was performed to investigate islet morphology and insulin content.
RESULTS: Administration of rAd-SP-rINSfur lowered blood glucose levels and normoglycemia was maintained for 50 d, whereas the rAd-CMV-βgal control virus-injected mice remained hyperglycemic. Glucose tolerance tests showed that rAd-SP-rINSfur-treated mice cleared exogenous glucose from the blood more efficiently than control virus-injected mice at 4 wk [area under the curve (AUC): 21 508.80 ± 2248.18 vs 62 640.00 ± 5014.28, P < 0.01] and at 6 wk (AUC: 29 956.60 ± 1757.33 vs 60 016.60 ± 3794.47, P < 0.01). In addition, insulin sensitivity was also significantly improved in mice treated with rAd-SP-rINSfur compared with rAd-CMV-βgal-treated mice (AUC: 9150.17 ± 1007.78 vs 11 994.20 ± 474.40, P < 0.05). The islets from rAd-SP-rINSfur-injected mice appeared to be smaller and to contain a higher concentration of insulin than those from rAd-CMV-βgal-injected mice.
CONCLUSION: Based on these results, we suggest that insulin gene therapy might be one therapeutic option for remission of type 2 diabetes.
Collapse
|
16
|
Abstract
Despite the fact that insulin injection can protect diabetic patients from developing diabetes-related complications, recent meta-analyses indicate that rapid and long-acting insulin analogues only provide a limited benefit compared with conventional insulin regarding glycemic control. As insulin deficiency is the main sequel of type-1 diabetes (T1D), transfer of the insulin gene-by-gene therapy is becoming an attractive treatment modality even though T1D is not caused by a single genetic defect. In contrast to human insulin and insulin analogues, insulin gene therapy targets to supplement patients not only with insulin but also with C-peptide. So far, insulin gene therapy has had limited success because of delayed and/or transient gene expression. Sustained insulin gene expression is now feasible using current gene-therapy vectors providing patients with basal insulin coverage, but management of postprandial hyperglycaemia is still difficult to accomplish because of the inability to properly control insulin secretion. Enteroendocrine cells of the gastrointestinal track (K cells and L cells) may be ideal targets for insulin gene therapy, but cell-targeting difficulties have limited practical implementation of insulin gene therapy for diabetes treatment. Therefore, recent gene transfer technologies developed to generate authentic beta cells through transdifferentiation are also highlighted in this review.
Collapse
|
17
|
Ahmad Z, Rasouli M, Azman AZF, Omar AR. Evaluation of insulin expression and secretion in genetically engineered gut K and L-cells. BMC Biotechnol 2012; 12:64. [PMID: 22989329 PMCID: PMC3469342 DOI: 10.1186/1472-6750-12-64] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/17/2012] [Indexed: 12/25/2022] Open
Abstract
Background Gene therapy could provide an effective treatment of diabetes. Previous studies have investigated the potential for several cell and tissue types to produce mature and active insulin. Gut K and L-cells could be potential candidate hosts for gene therapy because of their special features. Results In this study, we isolated gut K and L-cells to compare the potential of both cell types to produce insulin when exposed to similar conditions. The isolated pure K and L-cells were transfected with recombinant plasmids encoding insulin and with specific promoters for K or L-cells. Insulin expression was studied in response to glucose or meat hydrolysate. We found that glucose and meat hydrolysate efficiently induced insulin secretion from K and L-cells. However, the effects of meat hydrolysate on insulin secretion were more potent in both cells compared with glucose. Results of enzyme-linked immunosorbent assays showed that L-cells secreted more insulin compared with K-cells regardless of the stimulator, although this difference was not statistically significant. Conclusion The responses of K and L-cells to stimulation with glucose or meat hydrolysate were generally comparable. Therefore, both K and L-cells show similar potential to be used as surrogate cells for insulin gene expression in vitro. The potential use of these cells for diabetic gene therapy warrants further investigation.
Collapse
Affiliation(s)
- Zalinah Ahmad
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Selangor 43400, Malaysia.
| | | | | | | |
Collapse
|
18
|
Gene regulation systems for gene therapy applications in the central nervous system. Neurol Res Int 2012; 2012:595410. [PMID: 22272373 PMCID: PMC3261487 DOI: 10.1155/2012/595410] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 09/23/2011] [Indexed: 01/02/2023] Open
Abstract
Substantial progress has been made in the development of novel gene therapy strategies for central nervous system (CNS) disorders in recent years. However, unregulated transgene expression is a significant issue limiting human applications due to the potential side effects from excessive levels of transgenic protein that indiscriminately affect both diseased and nondiseased cells. Gene regulation systems are a tool by which tight tissue-specific and temporal regulation of transgene expression may be achieved. This review covers the features of ideal regulatory systems and summarises the mechanics of current exogenous and endogenous gene regulation systems and their utility in the CNS.
Collapse
|
19
|
Remission of diabetes by insulin gene therapy using a hepatocyte-specific and glucose-responsive synthetic promoter. Mol Ther 2010; 19:470-8. [PMID: 21119621 DOI: 10.1038/mt.2010.255] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Efficient production of insulin in response to changes in glucose levels has been a major issue for insulin gene therapy to treat diabetes. To express target genes in response to glucose specifically in hepatocytes, we generated a synthetic promoter library containing hepatocyte nuclear factor-1, CAAT/enhancer-binding protein (C/EBP) response element, and glucose-response element. Combinations of these three cis-elements in 3-, 6-, or 9-element configurations were screened for transcriptional activity and then glucose responsiveness in vitro. The most effective promoter (SP23137) was selected for further study. Intravenous administration of a recombinant adenovirus expressing furin-cleavable rat insulin under control of the SP23137 promoter into streptozotocin (STZ)-induced diabetic mice resulted in normoglycemia, which was maintained for >30 days. Glucose tolerance tests showed that treated mice produced insulin in response to glucose and cleared exogenous glucose from the blood in a manner similar to nondiabetic control mice, although the clearance was somewhat delayed. Insulin expression was seen specifically in the liver and not in other organs. These observations indicate the potential of this synthetic, artificial promoter to regulate glucose-responsive insulin production and remit hyperglycemia, thus providing a new method of liver-directed insulin gene therapy for type 1 diabetes.
Collapse
|
20
|
Won JC, Rhee BD, Ko KS. Glucose-responsive gene expression system for gene therapy. Adv Drug Deliv Rev 2009; 61:633-40. [PMID: 19394377 DOI: 10.1016/j.addr.2009.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 03/25/2009] [Indexed: 12/30/2022]
Abstract
Regulation of gene expression by glucose is an important mechanism for mammals in adapting to their nutritional environment. Glucose, the primary fuel for most cells, modulates gene expression that is crucial in the cellular adaptation to glycemic variation. Transcription of the genes for insulin and glycolytic and lipogenic enzymes is stimulated by glucose in pancreatic beta-cells and liver. Recent findings further support the key role of the carbohydrate-responsive element binding protein in the regulation of glycolytic and lipogenic genes by glucose and dietary carbohydrates. Herein, we review the transcriptional regulation of glucose-responsive genes, and recent advances in the gene therapy using glucose-responsive gene expression for diabetes.
Collapse
Affiliation(s)
- Jong Chul Won
- Department of Internal Medicine, Sanggye Paik Hospital, Mitochondrial Research Group, Inje University College of Medicine, Seoul, Republic of Korea
| | | | | |
Collapse
|
21
|
Olson DE, Thulé PM. Gene transfer to induce insulin production for the treatment of diabetes mellitus. Expert Opin Drug Deliv 2008; 5:967-77. [DOI: 10.1517/17425247.5.9.967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Darin E Olson
- Assistant Professor of Internal Medicine Emory University School of Medicine, Atlanta VA Medical Center, Division of Endocrinology, Lipids & Metabolism, USA
| | - Peter M Thulé
- Associate Professor of Internal Medicine Emory University School of Medicine, Atlanta VA Medical Center, Division of Endocrinology, Lipids & Metabolism, USA ;
| |
Collapse
|
22
|
Tian J, Lei P, Laychock SG, Andreadis ST. Regulated Insulin Delivery From Human Epidermal Cells Reverses Hyperglycemia. Mol Ther 2008; 16:1146-53. [DOI: 10.1038/mt.2008.79] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
23
|
Olson DE, Campbell AG, Porter MH, Freeman KG, Kelso E, Flatt WP, Thulé PM. Hepatic insulin gene therapy normalizes diurnal fluctuation of oxidative metabolism in diabetic BB/Wor rats. Mol Ther 2008; 16:1235-42. [PMID: 18500248 DOI: 10.1038/mt.2008.97] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Previous studies of hepatic insulin gene therapy (HIGT) focused on glycemic effects of insulin produced from hepatocytes. In this study, we extend the observations of glycemic control with metabolically regulated HIGT to include systemic responses and whole-body metabolism. An insulin transgene was administered with an adenoviral vector [Ad/(GlRE)(3)BP1-2xfur] to livers of BB/Wor rats made diabetic with polyinosinic polycytidilic acid (poly-I:C) (HIGT group), and results compared with nondiabetic controls (non-DM), and diabetic rats receiving different doses of continuous-release insulin implants (DM-low BG and DM-high BG). Blood glucose and growth normalized in HIGT, with lower systemic insulin levels, elevated glucagon, and increased heat production compared with non-DM. Minimal regulation of systemic insulin levels were observed with HIGT, yet the animals maintained normal switching from carbohydrate to lipid metabolism determined by respiratory quotients (RQs), and tolerated 24-hour fasts without severe hypoglycemia. HIGT did not restore serum lipids as we observed increased triglycerides (TGs) and increased free fatty acids, but reduced weight of visceral fat pads despite normal total body fat content and retroperitoneal fat depots. HIGT favorably affects blood glucose, normalizes metabolic switching in diabetic rats, and reduces intra-abdominal fat deposition.
Collapse
Affiliation(s)
- Darin E Olson
- Research Service Line, Atlanta VA Medical Center, Decatur, Georgia 30033, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Muniappan L, Ozcan S. Induction of insulin secretion in engineered liver cells by nitric oxide. BMC PHYSIOLOGY 2007; 7:11. [PMID: 17941991 PMCID: PMC2121102 DOI: 10.1186/1472-6793-7-11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 10/17/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND Type 1 Diabetes Mellitus results from an autoimmune destruction of the pancreatic beta cells, which produce insulin. The lack of insulin leads to chronic hyperglycemia and secondary complications, such as cardiovascular disease. The currently approved clinical treatments for diabetes mellitus often fail to achieve sustained and optimal glycemic control. Therefore, there is a great interest in the development of surrogate beta cells as a treatment for type 1 diabetes. Normally, pancreatic beta cells produce and secrete insulin only in response to increased blood glucose levels. However in many cases, insulin secretion from non-beta cells engineered to produce insulin occurs in a glucose-independent manner. In the present study we engineered liver cells to produce and secrete insulin and insulin secretion can be stimulated via the nitric oxide pathway. RESULTS Expression of either human insulin or the beta cell specific transcription factors PDX-1, NeuroD1 and MafA in the Hepa1-6 cell line or primary liver cells via adenoviral gene transfer, results in production and secretion of insulin. Although, the secretion of insulin is not significantly increased in response to high glucose, treatment of these engineered liver cells with L-arginine stimulates insulin secretion up to three-fold. This L-arginine-mediated insulin release is dependent on the production of nitric oxide. CONCLUSION Liver cells can be engineered to produce insulin and insulin secretion can be induced by treatment with L-arginine via the production of nitric oxide.
Collapse
Affiliation(s)
- Latha Muniappan
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, 741 South Limestone, BBSRB, Lexington, KY 40536, USA.
| | | |
Collapse
|
25
|
Lei P, Ogunade A, Kirkwood KL, Laychock SG, Andreadis ST. Efficient Production of Bioactive Insulin from Human Epidermal Keratinocytes and Tissue-Engineered Skin Substitutes: Implications for Treatment of Diabetes. ACTA ACUST UNITED AC 2007; 13:2119-31. [PMID: 17518716 DOI: 10.1089/ten.2006.0210] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite many years of research, daily insulin injections remain the gold standard for diabetes treatment. Gene therapy may provide an alternative strategy by imparting the ability to secrete insulin from an ectopic site. The epidermis is a self-renewing tissue that is easily accessible and can provide large numbers of autologous cells to generate insulin-secreting skin substitutes. Here we used a recombinant retrovirus to modify human epidermal keratinocytes with a gene encoding for human proinsulin containing the furin recognition sequences at the A-C and B-C junctions. Keratinocytes were able to process proinsulin and secrete active insulin that promoted glucose uptake. Primary epidermal cells produced higher amounts of insulin than cell lines, suggesting that insulin secretion may depend on the physiological state of the producer cells. Modified cells maintained the ability to stratify into 3-dimensional skin equivalents that expressed insulin at the basal and suprabasal layers. Modifications at the furin recognition sites did not improve proinsulin processing, but a single amino acid substitution in the proinsulin B chain enhanced C-peptide secretion from cultured cells and bioengineered skin substitutes 10- and 28-fold, respectively. These results suggest that gene-modified bioengineered skin may provide an alternative means of insulin delivery for treatment of diabetes.
Collapse
Affiliation(s)
- Pedro Lei
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, New York 14260, USA
| | | | | | | | | |
Collapse
|
26
|
Kozlowski M, Olson DE, Rubin J, Lyszkowicz D, Campbell A, Thulé PM. Adeno-associated viral delivery of a metabolically regulated insulin transgene to hepatocytes. Mol Cell Endocrinol 2007; 273:6-15. [PMID: 17553615 DOI: 10.1016/j.mce.2007.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 04/20/2007] [Indexed: 10/23/2022]
Abstract
Transduction with a liver specific, metabolically responsive insulin transgene produces near-normal blood sugars in STZ-diabetic rats. To overcome the limited duration of hepatic transgene expression induced by E1A-deleted adenoviral vectors, we evaluated recombinant adeno-associated virus (rAAV2) for cell type specificity and glucose responsiveness in vitro. Co-infection of AAV2 containing the glucose responsive, liver-specific (GlRE)(3)BP-1 promoter with an empty adenovirus enhanced transduction efficiency, and shortened the duration of transgene expression in HepG2 hepatoma cells, but not primary hepatocytes. However, in the context of rAAV2, (GlRE)(3)BP-1 promoter activity remained confined to cells of hepatocyte lineage, and retained glucose responsiveness. While isolated infection with an insulin expressing rAAV2 failed to attenuate blood sugars in diabetic mice, adenoviral co-administration with the same rAAV2 induced transient, near-normal random blood sugars in a diabetic animal. We conclude that rAAV2 can induce metabolically responsive insulin secretion from hepatocytes in vitro and in vivo. However, alternative AAV serotypes will likely be required to efficiently deliver therapeutic genes to the liver for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Miroslaw Kozlowski
- Department of Orthopedics, Veterans Affairs Medical Center and Emory University School of Medicine, Atlanta, GA 30033, USA
| | | | | | | | | | | |
Collapse
|
27
|
Han J, Lee HH, Kwon H, Shin S, Yoon JW, Jun HS. Engineered enteroendocrine cells secrete insulin in response to glucose and reverse hyperglycemia in diabetic mice. Mol Ther 2007; 15:1195-202. [PMID: 17299398 DOI: 10.1038/sj.mt.6300117] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Type 1 diabetes is a metabolic disorder caused by loss of insulin-producing pancreatic beta-cells. Expression of insulin in non-beta-cells to create beta-cell surrogates has been tried to treat type 1 diabetes. Enteroendocrine K cells have characteristics similar to pancreatic beta-cells, such as a glucose-sensing system and insulin-processing proteases. In this study, we genetically engineered an enteroendocrine cell line (STC-1) to express insulin under the control of the glucose-dependent insulinotropic polypeptide promoter. We screened clones and chose one, Gi-INS-7, based on its high production of insulin. Gi-INS-7 cells expressed glucose transporter 2 (GLUT2) and glucokinase (GK) and secreted insulin in response to elevated glucose levels in vitro. To determine whether Gi-INS-7 cells can control blood glucose levels in diabetic mice, we transplanted these cells under the kidney capsule of streptozotocin (STZ)-induced diabetic mice and found that blood glucose levels became normal within 2 weeks of transplantation. In addition, glucose tolerance tests in mice that became normoglycemic after transplantation with Gi-INS-7 cells showed that exogenous glucose was cleared appropriately. These results suggest that engineered K cells may be promising surrogate beta-cells for possible therapeutic use for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Jaeseok Han
- Julia McFarlane Diabetes Research Centre, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Chistiakov DA, Tyurina I. Current strategies and perspectives in insulin gene therapy for diabetes. Expert Rev Endocrinol Metab 2007; 2:27-34. [PMID: 30743746 DOI: 10.1586/17446651.2.1.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Insulin gene therapy is an approach that might overcome the weakness of islet cell therapy owing to its vulnerability to autoimmune attack. There are several mandatory conditions for successful insulin gene therapy. Efficient insulin gene therapy should have an effective insulin gene delivery mechanism, a system of regulation of the insulin biosynthesis that responds to glucose within extremely narrow physiological limits, a system of insulin processing into its active form and a choice of appropriate target cells, which possess biochemical characteristics similar to β cells, but are not targets for β-cell-specific self-reactivity. In this article, advantages and disadvantages of non-β-cell types that are most likely to be used for generating surrogate insulin-producing β cells are compared. Current achievements in insulin gene therapy are critically evaluated and future challenges are discussed.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Assistant Professor, University of Pittsburgh Medical Center, Department of Pathology, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Inna Tyurina
- b Executive Manager and Consultant, Public Relations and Consulting Group 'Imya', 8th Tekstilschikov Street 11, 109129, Moscow, Russia.
| |
Collapse
|
29
|
Fodor A, Harel C, Fodor L, Armoni M, Salmon P, Trono D, Karnieli E. Adult rat liver cells transdifferentiated with lentiviral IPF1 vectors reverse diabetes in mice: an ex vivo gene therapy approach. Diabetologia 2007; 50:121-30. [PMID: 17131142 DOI: 10.1007/s00125-006-0509-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 09/01/2006] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS We examined a clinical model of ex vivo transdifferentiation of primary adult hepatocytes to insulin-secreting cells for the treatment of type 1 diabetes. MATERIALS AND METHODS Isolated rat hepatocytes were transduced in primary culture with a human lentivirus containing pancreatic duodenal homeobox 1 (PDX1, now known as insulin promoter factor 1, homeodomain transcription factor [IPF1]). Insulin expression and secretion of the newly engineered cells were assessed in vitro by RT-PCR, in situ hybridisation, immunostaining and radioimmunoassay. PDX1-transduced hepatocytes were further studied in vivo by injecting them under the renal capsule of diabetic SCID mice. RESULTS Isolated rat hepatocytes were efficiently transduced with the lentiviral vector, as assessed by green fluorescent reporter gene expression. The transduced cells exhibited insulin at both mRNA (RT-PCR, in situ hybridisation) and protein levels (immunostaining and radioimmunoassay). Moreover, insulin secretion by the engineered cells was dependent on glucose and sulfonylurea. Other beta cell genes, including those encoding solute carrier family 2 (facilitated glucose transporter), member 2 (Slc2a2), glucokinase (Gck), ATP-binding cassette, sub-family C (CFTR/MRP), member 8 (Abcc8), the potassium inwardly-rectifying channel, subfamily J, member 11 (Kcnj11) and proprotein convertase subtilisin/kexin type 1 (Pcsk1) were also expressed. The PDX1-transduced hepatocytes expressed several pancreatic transcription factors related to early pancreatic endocrine development (endogenous Pdx1, neurogenic differentiation factor 1 [Neurod1], and NK6 transcription factor related, locus 1 [Nkx6-1]) as well as the late-stage pancreatic transcription factors (paired box gene 4 [Pax4], paired box gene 6 [Pax6], and v-maf musculoaponeurotic fibrosarcoma oncogene homolog A [Mafa]). Transplantation of 3 x 10(6) transdifferentiated liver cells under the renal capsule of seven streptozotocin-induced diabetic SCID mice resulted in significant reduction of non-fasting blood glucose levels from 30.7 +/- 1.3 to 8.7 +/- 3.7 mmol/l (mean +/- SEM, p = 0.01), in 6 to 8 weeks. Removal of the graft resulted in severe hyperglycaemia. CONCLUSIONS/INTERPRETATION Ex vivo lentiviral-mediated PDX1 expression in isolated adult liver cells represents a potential model for type 1 diabetes mellitus therapy.
Collapse
Affiliation(s)
- A Fodor
- The Institute of Endocrinology, Diabetes and Metabolism, Rambam Medical Center, P.O. Box 9602, Haifa, 31096, Israel
| | | | | | | | | | | | | |
Collapse
|
30
|
Lan MS, Wang HW, Chong J, Breslin MB. Coupling of glucose response element from L-type pyruvate kinase and G6Pase promoter enhances glucose responsive activity in hepatoma cells. Mol Cell Biochem 2006; 300:191-6. [PMID: 17160355 DOI: 10.1007/s11010-006-9383-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Accepted: 11/07/2006] [Indexed: 11/24/2022]
Abstract
Type 1 diabetes results from the autoimmune destruction of pancreatic beta-cells, which leads to severe insulin deficiency. Insulin gene therapy provides an attractive approach to cure diabetes. The critical factor for insulin gene therapy in surrogate cells is to select an appropriate site for insulin expression and a tissue-specific promoter that is responsive to both physiological glucose and insulin concentrations. A novel chimeric promoter, (GIRE)n-G6Pase, consisting of a 1.6 kb glucose 6-phosphatase (G6Pase) promoter and a segment of the regulatory element derived from the L-type pyruvate kinase (L-PK) promoter, was designed to provide strong and tight control of insulin expression in liver. One or three copies of GIRE were linked to the G6Pase promoter, which showed a stronger promoter activity than the G6Pase promoter alone. The chimeric promoter was inhibited by insulin in a dosage-dependent manner and activated by glucose, two features essential for glucose metabolism. The promoter activity is conserved between species and highly specific for liver cells. The construction of a chimeric promoter with stronger and more sensitive responsive activity to glucose and insulin in liver cells could further advance studies in insulin gene therapy.
Collapse
Affiliation(s)
- Michael S Lan
- The Research Institute for Children, Departments of Pediatrics and Genetics, Children's Hospital, Louisiana State University Health Sciences Center, 200 Henry Clay Avenue, New Orleans, LA 70118, USA.
| | | | | | | |
Collapse
|
31
|
Oh TK, Li MZ, Kim ST. Gene therapy for diabetes mellitus in rats by intramuscular injection of lentivirus containing insulin gene. Diabetes Res Clin Pract 2006; 71:233-40. [PMID: 16171885 DOI: 10.1016/j.diabres.2005.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Accepted: 08/01/2005] [Indexed: 12/30/2022]
Abstract
We assessed therapeutic potential of intramuscular insulin gene delivery in a diabetic murine model. The human proinsulin gene cDNA engineered with concensus furin cleavage sequences was inserted into an advanced lentiviral vector that contained CMV early promoter. After injection of concentrated lentiviral vector (3.5 microg p24 Gag antigen) carrying the insulin gene into the thigh muscle, treated rats demonstrated an increase in body weight, increased survivability, attenuated the hyperglycemic response as well as prevented the formation of ketoacidosis. For these reasons, the intraparenchymal injection of lentiviral vectors into the skeletal muscle to ectopically produce insulin may be an easy and therapeutic treatment modality for type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Tae Keun Oh
- Department of Internal Medicine, Chungbuk National University College of Medicine, 62 Kaeshin-Dong, Hungdok-Gu, Cheongju, Chungbuk 361-711, South Korea
| | | | | |
Collapse
|
32
|
Abstract
Diabetes remains a devastating disease, with tremendous cost in terms of human suffering and healthcare expenditures. A bioartificial pancreas has the potential as a promising approach to preventing or reversing complications associated with this disease. Bioartificial pancreatic constructs are based on encapsulation of islet cells with a semipermeable membrane so that cells can be protected from the host's immune system. Encapsulation of islet cells eliminates the requirement of immunosuppressive drugs, and offers a possible solution to the shortage of donors as it may allow the use of animal islets or insulin-producing cells engineered from stem cells. During the past 2 decades, several major approaches for immunoprotection of islets have been studied. The microencapsulation approach is quite promising because of its improved diffusion capacity, and technical ease of transplantation. It has the potential for providing an effective long-term treatment or cure of Type 1 diabetes.
Collapse
Affiliation(s)
- Seda Kizilel
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
33
|
Burkhardt BR, Parker MJ, Zhang YC, Song S, Wasserfall CH, Atkinson MA. Glucose transporter-2 (GLUT2) promoter mediated transgenic insulin production reduces hyperglycemia in diabetic mice. FEBS Lett 2005; 579:5759-64. [PMID: 16223491 DOI: 10.1016/j.febslet.2005.09.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 09/22/2005] [Accepted: 09/23/2005] [Indexed: 11/27/2022]
Abstract
Insulin production afforded by hepatic gene therapy (HGT) retains promise as a potential treatment for type 1 diabetes, but successful approaches have been limited. We employed a novel and previously untested promoter for this purpose, glucose transporter-2 (GLUT2) to drive insulin production via delivery by recombinant adeno-associated virus (rAAV). In vitro, the GLUT2 promoter was capable of robust glucose-responsive expression in transduced HepG2 human hepatoma cells. Therefore, rAAV constructs were designed to express the furin-cleavable human preproinsulin B10 gene, under the control of the murine GLUT2 promoter and packaged for delivery with rAAV expressing the type 5 capsid. Streptozotocin-induced diabetic mice were subjected to hepatic portal vein injection immediately followed by implantation of a sustained-release insulin pellet to allow time for transgenic expression. All mice injected with the rAAV5-GLUT2-fHPIB10 virus remained euglycemic for up to 35 days post-injection, with 50% euglycemic after 77 days post-injection. In contrast, mock-injected mice became hyperglycemic within 15 days post-injection following dissolution of the insulin pellet. Serum levels of both human insulin and C-peptide further confirmed successful transgenic delivery by the rAAV5-GLUT2-fHPIB10 virus. These findings indicate that the GLUT2 promoter may be a potential candidate for regulating transgenic insulin production for hepatic insulin gene therapy in the treatment of type I diabetes.
Collapse
Affiliation(s)
- Brant R Burkhardt
- Department of Pathology, University of Florida College of Medicine, Gainesville, 32610, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Kim YD, Park KG, Morishita R, Kaneda Y, Kim SY, Song DK, Kim HS, Nam CW, Lee HC, Lee KU, Park JY, Kim BW, Kim JG, Lee IK. Liver-directed gene therapy of diabetic rats using an HVJ-E vector containing EBV plasmids expressing insulin and GLUT 2 transporter. Gene Ther 2005; 13:216-24. [PMID: 16177820 DOI: 10.1038/sj.gt.3302644] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Insulin gene therapy in clinical medicine is currently hampered by the inability to regulate insulin secretion in a physiological manner, the inefficiency with which the gene is delivered, and the short duration of gene expression. To address these issues, we injected the liver of streptozotocin-induced diabetic rats with hemagglutinating virus of Japan-envelope (HVJ-E) vectors containing Epstein-Barr virus (EBV) plasmids encoding the genes for insulin and the GLUT 2 transporter. Efficient delivery of the genes was achieved with the HVJ-E vector, and the use of the EBV replicon vector led to prolonged hepatic gene expression. Blood glucose levels were normalized for at least 3 weeks as a result of the gene therapy. Cotransfection of GLUT 2 with insulin permitted the diabetic rats to regulate their blood glucose levels upon exogenous glucose loading in a physiologically appropriate manner and improved postprandial glucose levels. Moreover, cotransfection with insulin and GLUT 2 genes led to in vitro glucose-stimulated insulin secretion that involved the closure of K(ATP) channels. The present study represents a new way to efficiently deliver insulin gene in vivo that is regulated by ambient glucose level with prolonged gene expression. This may provide a basis to overcome limitations of insulin gene therapy in humans.
Collapse
Affiliation(s)
- Y D Kim
- Department of Internal Medicine & Institute for Medical Sciences, Keimyung University School of Medicine, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lu YC, Sternini C, Rozengurt E, Zhukova E. Release of transgenic human insulin from gastric g cells: a novel approach for the amelioration of diabetes. Endocrinology 2005; 146:2610-9. [PMID: 15731364 DOI: 10.1210/en.2004-1109] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We explored the hypothesis that meal-regulated release of insulin from gastric G cells can be used for gene therapy for diabetes. We generated transgenic mice in which the coding sequence of human insulin has been knocked into the mouse gastrin gene. Insulin was localized specifically to antral G cells of G-InsKi mice by double immunofluorescence staining using antibodies against insulin and gastrin. Insulin extracted from antral stomach of G-InsKi mice decreased blood glucose upon injection into streptozotocin-diabetic mice. Intragastric administration of peptone, a known potent luminal stimulant of gastrin secretion, induced an increase in circulating levels of transgenic human insulin from 10.7 +/- 2 to 23.3 +/- 4 pm in G-InsKi mice. Although G cell-produced insulin decreased blood glucose in G-InsKi mice, it did not cause toxic hypoglycemia. Proton pump inhibitors, pharmacological agents that increase gastrin output, caused a further increase in the circulating levels of gastric insulin (41.5 +/- 2 pm). G cell-produced insulin was released into circulation in response to the same meal-associated stimuli that control release of gastrin. The most striking aspect of the results presented here is that in the presence of the G-InsKi allele, Ins2(Akita/+) mice exhibited a marked prolongation of life span. These results imply that G cell-derived transgenic insulin is beneficial in the amelioration of diabetes. We suggest that an efficient G cells-based insulin gene therapy can relieve diabetic patients from daily insulin injections and protect them from complications of insulin insufficiency while avoiding episodes of toxic hypoglycemia.
Collapse
Affiliation(s)
- Yu-Chun Lu
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine at University of California, Los Angeles, California 90095-1786, USA
| | | | | | | |
Collapse
|
36
|
Cotugno G, Pollock R, Formisano P, Linher K, Beguinot F, Auricchio A. Pharmacological regulation of the insulin receptor signaling pathway mimics insulin action in cells transduced with viral vectors. Hum Gene Ther 2005; 15:1101-8. [PMID: 15610610 DOI: 10.1089/hum.2004.15.1101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus derives from either insulin deficiency (type I) or resistance (type II). Homozygous mutations in the insulin receptor (IR) gene cause the rare leprechaunism and Rabson-Mendenhall syndromes, severe forms of hyperinsulinemic insulin resistance for which no therapy is currently available. Systems have been developed that allow protein-protein interactions to be brought under the control of small-molecule dimerizer drugs. As a potential tool to rescue glucose homeostasis at will in both insulin and insulin receptor deficiencies, we developed a recombinant chimeric insulin receptor (LFv2IRE) that can be homodimerized and activated by the small-molecule dimerizer AP20187. In HepG2 cells transduced with adeno-associated viral (AAV) vectors encoding LFv2IRE, AP20187 induces LFv2IRE homodimerization and transphosphorylation minutes after drug administration, resulting in the phosphorylation of a canonical substrate of the insulin receptor tyrosine kinase, IRS-1. AP20187 activation of LFv2IRE is dependent on the dose of drug and the amount of chimeric receptor expressed in AAV-transduced cells. Finally, AP20187-dependent activation of LFv2IRE results in insulin-like effects, such as induction of glycogen synthase activity and cellular proliferation. In vivo LFv2IRE transduction of insulin target tissues followed by AP20187 dosing may represent a therapeutic strategy to be tested in animal models of insulin resistance due to insulin receptor deficiency or of type I diabetes. This system may also represent a useful tool to dissect in vivo the independent contribution of insulin target tissues to hormone action.
Collapse
Affiliation(s)
- Gabriella Cotugno
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Qian Q, Williams JP, Karounos DG, Ozcan S. Nitric oxide stimulates insulin release in liver cells expressing human insulin. Biochem Biophys Res Commun 2005; 329:1329-33. [PMID: 15766572 DOI: 10.1016/j.bbrc.2005.02.111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Indexed: 01/15/2023]
Abstract
The establishment of surrogate islet beta cells is important for the treatment of diabetes. Hepatocytes have a similar glucose sensing system as beta cells and have the potential to serve as surrogate beta cells. In this report, we demonstrate that infection of Hepa1-6 liver cells with a lentivirus expressing the human insulin cDNA results in expression and secretion of human insulin. Furthermore, we show that l-arginine at low levels of glucose significantly stimulates the release of insulin from these cells, compared to exposure to high concentration of glucose. The arginine-induced insulin release is via the production of nitric oxide, since treatment with N(G)-nitro-l-arginine, an inhibitor of nitric oxide synthase, blocks insulin secretion induced by l-arginine. These results indicate that nitric oxide plays a role in l-arginine-stimulated insulin release in hepatocytes expressing the human insulin gene, and provides a new strategy to induce insulin secretion from engineered non-beta cells.
Collapse
Affiliation(s)
- Qingwen Qian
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
38
|
Park YM, Woo S, Lee GT, Ko JY, Lee Y, Zhao ZS, Kim HJ, Ahn CW, Cha BS, Kim KS, Park CW, Lee HC. Safety and efficacy of adeno-associated viral vector-mediated insulin gene transfer via portal vein to the livers of streptozotocin-induced diabetic Sprague-Dawley rats. J Gene Med 2005; 7:621-9. [PMID: 15651056 DOI: 10.1002/jgm.708] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Previous studies demonstrating the efficacy of insulin gene therapy have mostly involved use of adenoviral vectors or naked DNA to deliver the insulin gene. However, this procedure may not guarantee long-term insulin production. To improve the performance, we prepared recombinant adeno-associated viral vectors (rAAV) harboring the gene encoding a furin-modified human insulin under the cytomegalovirus (CMV) promoter [rAAV-hPPI(F12)]. METHODS Streptozotocin (STZ)-induced diabetic Sprague-Dawley rats were used as a diabetic animal model. The levels of blood glucose, insulin, and HbA1c were measured to test the effect. An intraperitoneal glucose tolerance test was performed to test the capability of blood glucose disposal. Immunohistochemical staining and Northern blot analyses were performed to survey the expression pattern of the therapeutic insulin gene. RESULTS STZ-induced diabetic Sprague-Dawley rats infused via the portal vein with rAAV-hPPI(F12) produced human insulin and after a 6-h fast were normoglycemic for over 90 days post-treatment, whereas diabetic rats treated with recombinant adenoviral vector harboring the hPPI(F12) gene [rAV-hPPI(F12)] were normoglycemic only for days 3 to 13 post-treatment. Insulin mRNA was detected mainly in the liver of the rAAV-hPPI(F12)-treated diabetic rats. The glucose tolerance capability of the rAAV-hPPI(F12)-treated diabetic rats was comparable to that of non-diabetic rats, even without injection of recombinant insulin. Furthermore, blood HbA1c concentrations in rAAV-hPPI(F12)-treated diabetic rats were reduced to almost the normal level. Importantly, studies of rAV or rAAV vector-dependent side effects on the targeted liver strongly suggested that only rAAV treatment caused no side effects. CONCLUSIONS These results demonstrate that our rAAV-mediated in vivo insulin gene therapy provides safer maintenance of the insulin gene expression required for long-term and thus more effective blood glycemic control.
Collapse
Affiliation(s)
- Young Mi Park
- Brain Korea 21 Project for Medical Science, College of Medicine, Yonsei University, Seoul 120-752, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Gene therapy has been hyped as a possible 'cure' for diabetes mellitus in the near future ever since insulin was first cloned and expressed in cultured cells in the late 1970s. In the past decade, however, the bar for gene therapy for diabetes has been raised because of recent advances in the clinical management of diabetes. Although current treatment modalities fall far short of a cure, they produce greatly improved, if imperfect, glycemic control. In this context, we review the latest advances in in vivo gene therapy and conclude that the most widely applied strategy of insulin gene transfer does not measure up to the existing treatment options, whereas the recently proved concept of induced islet neogenesis has the potential of bettering the currently available therapy. Much work remains to be done, however, before this regimen can be taken from the bench to the bedside.
Collapse
Affiliation(s)
- Lawrence Chan
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Texas Medical Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
40
|
Olson DE, Paveglio SA, Huey PU, Porter MH, Thulé PM. Glucose-responsive hepatic insulin gene therapy of spontaneously diabetic BB/Wor rats. Hum Gene Ther 2004; 14:1401-13. [PMID: 14577921 DOI: 10.1089/104303403769211628] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatic insulin gene therapy (HIGT) ameliorates hyperglycemia in multiple rodent models of diabetes mellitus, with variable degrees of glucose control. We demonstrate here that adenoviral delivery of a glucose-regulated transgene into rat hepatocytes produces near-normal glycemia in spontaneously diabetic BB/Wor rats without administration of exogenous insulin. We compared growth, glycemia, counterregulatory hormones, and lipids in HIGT-treated diabetic rats to nondiabetic rats and diabetic rats treated with either insulin injections or sustained-release insulin pellets. HIGT-treated rats achieved near-normal blood glucose levels within 1 week and maintained glycemic control for up to 3 months. Rats treated with sustained release insulin implants had similar blood sugars, but more hypoglycemia and gained more weight than HIGT-treated rats. HIGT-treated rats normalized blood glucose within 2 hr after a glucose load, and tolerated a 24-hr fast without hypoglycemia. HIGT treatment suppressed ketogenesis similarly to peripheral insulin. However, glucagon levels and free fatty acids were increased in HIGT-treated rats compared to either nondiabetic controls or rats treated with exogenous insulin. In addition to extending successful application of HIGT to a rat model of autoimmune diabetes, these findings emphasize the relative contribution of hepatic insulin effect in the metabolic stabilization of diabetes mellitus.
Collapse
Affiliation(s)
- Darin E Olson
- Division of Endocrinology and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
41
|
Yasutomi K, Itokawa Y, Asada H, Kishida T, Cui FD, Ohashi S, Gojo S, Ueda Y, Kubo T, Yamagishi H, Imanishi J, Takeuchi T, Mazda O. Intravascular insulin gene delivery as potential therapeutic intervention in diabetes mellitus. Biochem Biophys Res Commun 2003; 310:897-903. [PMID: 14550289 DOI: 10.1016/j.bbrc.2003.09.103] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We assessed therapeutic potential of intravascular insulin gene delivery in a diabetic murine model. The rat proinsulin-1 gene cDNA engineered to harbor furin consensus cleavage sequences was inserted into EBV-based plasmid vectors that contained CAG promoter or multimerized rat insulin promoter (RIP). Normal or streptozotocin (STZ)-induced diabetic mice were given an injection of the plasmids via the tail vein under high pressure. Transfection of the CAG-proinsulin construct markedly improved hyperglycemia of diabetic mice, accompanied by a considerable increase in serum insulin concentrations. Although the RIP-plasmid failed to reduce fasting blood glucose, the glucose tolerance test and RT-PCR analysis revealed that insulin production was regulated in the liver in a blood glucose level-dependent manner. The present results suggest a potential therapeutic means of controlling DM.
Collapse
Affiliation(s)
- Kakei Yasutomi
- Department of Microbiology, Kyoto Prefectural University of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Insulin-dependent diabetes mellitus (IDDM) is an autoimmune disease resulting in destruction of the pancreatic beta-cells in the islets of Langerhans. Commonly employed treatment of IDDM requires periodic insulin therapy, which is not ideal because of its inability to prevent chronic complications such as nephropathy, neuropathy and retinopathy. Although pancreas or islet transplantation are effective treatments that can reverse metabolic abnormalities and prevent or minimize many of the chronic complications of IDDM, their usefulness is limited as a result of shortage of donor pancreas organs. Gene therapy as a novel field of medicine holds tremendous therapeutic potential for a variety of human diseases including IDDM. This review focuses on the liver-based gene therapy for generation of surrogate pancreatic beta-cells for insulin replacement because of the innate ability of hepatocytes to sense and metabolically respond to changes in glucose levels and their high capacity to synthesize and secrete proteins. Recent advances in the use of gene therapy to prevent or regenerate beta-cells from autoimmune destruction are also discussed.
Collapse
Affiliation(s)
- Philipp C Nett
- Department of Surgery, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | | | | |
Collapse
|
43
|
Ajamian F, Titok T, Suhorada E, Ruban T, Reeben M. Hepatic expression of the human insulin gene reduces glucose levels in vivo in diabetic mice model. DIABETES & METABOLISM 2003; 29:424-9. [PMID: 14526271 DOI: 10.1016/s1262-3636(07)70054-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES The objective of these studies was to evaluate human insulin gene expression following intraliver plasmid injection in diabetic mice as a potential approach to gene therapy for insulin-dependent diabetes mellitus. METHODS The fragment containing human proinsulin gene lacking its own promoter, was cloned into plasmids containing promoter and enhancer of cytomegalovirus or human hepatitis B virus. The resulting gene constructs were first tested in vitro using 3T3 fibroblast cell line and subsequently in vivo applying streptozotocin-induced diabetic mice. RESULTS We found significant reduction in glucose levels in both experimental systems, giving evidence that prolonged constitutive systemic secretion of bioactive human (pro)insulin has been attained in non-neuroendocrine cell line in vitro and in mice following intra-liver plasmid injection. CONCLUSION Our data demonstrate the reduction of glucose levels in vitro in 3T3 fibroblast cells and in vivo in diabetic mice after treatment with plasmids expressing proinsulin, giving evidence that those constructs may have certain usage also in human gene therapy of diabetes mellitus type 1.
Collapse
Affiliation(s)
- F Ajamian
- Department of Cell Mechanism Regulation, Institute of Molecular Biology and Genetics, Kiev, Ukraine
| | | | | | | | | |
Collapse
|
44
|
Bottino R, Lemarchand P, Trucco M, Giannoukakis N. Gene- and cell-based therapeutics for type I diabetes mellitus. Gene Ther 2003; 10:875-89. [PMID: 12732873 DOI: 10.1038/sj.gt.3302015] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Type 1 diabetes mellitus, an autoimmune disorder is an attractive candidate for gene and cell-based therapy. From the use of gene-engineered immune cells to induce hyporesponsiveness to autoantigens to islet and beta cell surrogate transplants expressing immunoregulatory genes to provide a local pocket of immune privilege, these strategies have demonstrated proof of concept to the point where translational studies can be initiated. Nonetheless, along with the proof of concept, a number of important issues have been raised by the choice of vector and expression system as well as the point of intervention; prophylactic or therapeutic. An assessment of the current state of the science and potential leads to the conclusion that some strategies are ready for safety trials while others require varying degrees of technical and conceptual refinement.
Collapse
Affiliation(s)
- R Bottino
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Gene therapy has been applied in a variety of experimental models of autoimmunity with some success. In this article, we outline recent developments in gene therapy vectors, discuss advantages and disadvantages of each, and highlight their recent applications in autoimmune models. We also consider progress in vector targeting and components for regulating transgene expression, which will both improve gene therapy safety and empower gene therapy to fullfil its potential as a therapeutic modality. In conclusion, we consider candidate vectors that satisfy requirements for application in the principal therapeutic strategies in which gene therapy will be applied to autoimmune conditions.
Collapse
Affiliation(s)
- D J Gould
- 1Bone & Joint Research Unit, Barts & The London, Queen Mary's Medical School, University of London, London, UK
| | | |
Collapse
|
46
|
Tang SC, Sambanis A. Development of genetically engineered human intestinal cells for regulated insulin secretion using rAAV-mediated gene transfer. Biochem Biophys Res Commun 2003; 303:645-52. [PMID: 12659868 DOI: 10.1016/s0006-291x(03)00399-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell-based therapies for treating insulin-dependent diabetes (IDD) can provide a more physiologic regulation of blood glucose levels in a less invasive fashion than daily insulin injections. Promising cells include intestinal enteroendocrine cells genetically engineered to secrete insulin in response to physiologic stimuli; responsiveness occurs at the exocytosis level to regulate the acute release of recombinant insulin. In this work, we established a human cellular model to demonstrate that meat hydrolysate can simultaneously stimulate glucagon-like peptide-1 (GLP-1, an enteroendocrine cell-derived incretin hormone) and recombinant insulin secretion from the engineered human NCI-H716 intestinal cell line. Cells were genetically modified using the recombinant adeno-associated virus (rAAV)-mediated insulin gene transfer. Recombinant cells were then differentiated to display endocrine features, in particular the formation of granule-like compartments. A fusion protein of insulin and enhanced green fluorescence protein (EGFP) was designed to reveal the compartments of localization of the fusion protein and assess its co-localization with endogenous GLP-1. Our work provides a unique human cellular model for regulated insulin release through genetic engineering of GLP-1-secreting intestinal cells, which is expected to be useful for cell-based therapies of IDD.
Collapse
Affiliation(s)
- Shiue-Cheng Tang
- School of Chemical Engineering, Georgia Tech-Emory Center for the Engineering of Living Tissues, and P.H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
47
|
Tang SC, Sambanis A. Preproinsulin mRNA engineering and its application to the regulation of insulin secretion from human hepatomas. FEBS Lett 2003; 537:193-7. [PMID: 12606056 DOI: 10.1016/s0014-5793(03)00121-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cell-based therapies for treating insulin-dependent diabetes (IDD) can provide a more physiologic regulation of blood glucose levels in a less invasive fashion than daily insulin injections. Promising cells include non-beta cells genetically engineered to secrete insulin in response to physiologic cues; responsiveness can be introduced at the transcriptional level to regulate preproinsulin (PPI) mRNA biosynthesis. However, these cells exhibit sluggish secretion dynamics, which is not appropriate for achieving euglycemia in higher animals and, eventually, humans. In this work, we have engineered the PPI mRNA so as to destabilize it through nonsense-mediated mRNA decay (NMD). When expressed under transcriptional regulation in HepG2 hepatomas, the engineered PPI mRNA level and of the insulin secretion rate declined faster upon switching off transcription, compared to the one-copy non-engineered control. Our work provides a simple and straightforward method to improve the dynamics of transcriptionally regulated insulin secretion, which can be a useful tool in developing cell-based therapies for IDD.
Collapse
Affiliation(s)
- Shiue-Cheng Tang
- School of Chemical Engineering, Georgia Tech-Emory Center for the Engineering of Living Tissues, Atlanta 30332, USA
| | | |
Collapse
|
48
|
Giannoukakis N, Robbins PD. Gene and cell therapies for diabetes mellitus: strategies and clinical potential. BioDrugs 2003; 16:149-73. [PMID: 12102644 DOI: 10.2165/00063030-200216030-00001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The last 5 years have witnessed an explosion in the use of genes and cells as biomedicines. While primarily aimed at cancer, gene engineering and cell therapy strategies have additionally been used for Mendelian, neurodegenerative and metabolic disorders. The main focus of gene and cell therapy strategies in metabolism has been diabetes mellitus. This disease is a disorder of glucose homeostasis, either due to the immune-mediated eradication of pancreatic beta cells in the islets of Langerhans (type 1 diabetes) or resulting from insulin resistance and obesity syndromes where the insulin-producing capability of the beta cell is ultimately exhausted in the face of insensitivity to the effects of insulin in the peripheral glucose-utilising tissues (type 2 diabetes). A significant number of animal studies have demonstrated the potential in restoring normoglycaemia by islet transplantation in the context of immunoregulation achieved by gene transfer of immunoregulatory genes to allo- and xenogeneic islets ex vivo. Additionally, gene and cell therapy has also been used to induce tolerance to auto- and alloantigens and to generate the tolerant state in autoimmune rodent animal models of type 1 diabetes or rodent recipients of allogeneic/xenogeneic islet transplants. The achievements of gene and cell therapy in type 2 diabetes are less evident, but seminal studies promise that this modality can be relevant to treat and perhaps prevent the underlying causes of the disease. Here we present an overview of the current status of gene and cell therapy for type 1 and 2 diabetes and we propose potential therapeutic options that could be clinically useful. For type 1 diabetes, transplantation of islets engineered to evade or suppress the recipient immune response is the most readily-available technology today. A number of gene delivery vectors encoding proteins that impair a variety of immune cells have already been examined and proven versatile. More challenging but, nonetheless, just over the horizon are attempts to promote tolerance to islet allografts. Type 2 diabetes will likely require a better understanding of the processes that determine insulin sensitivity in the periphery. Targeting tissues such as muscle and fat with vectors encoding genes whose products promote insulin sensitivity and glucose uptake is an approach that does not carry with it the side-effects often associated with pharmacologic agents currently in use. In the end, progress in vector design, elucidation of antigen-specific immunity and insulin sensitivity will provide the framework for gene drug use in the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.
| | | |
Collapse
|
49
|
Abstract
Encapsulation of insulin-producing cells in semipermeable membranes has the potential to provide an effective treatment for insulin-dependent diabetes with little or no immunosuppression of the host. Improvements in alginate, a marine polysaccharide commonly used for cell encapsulation, have revived interest in this material. However, serious obstacles, including a reliable cell source and a better understanding of immune acceptance issues, remain to be addressed before a clinically applicable therapeutic procedure based on encapsulated cells becomes available.
Collapse
Affiliation(s)
- Athanassios Sambanis
- School of Chemical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
50
|
Bottino R, Balamurugan AN, Giannoukakis N, Trucco M. Islet/pancreas transplantation: challenges for pediatrics. Pediatr Diabetes 2002; 3:210-23. [PMID: 15016150 DOI: 10.1034/j.1399-5448.2002.30408.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Beta cell replacement is a valid alternative to exogenous insulin injections to treat type 1 diabetic patients. The rate of success obtained after whole-pancreas transplantation, performed alone or in combination with kidney, and, as shown recently, by islet transplantation, justifies optimism and sets the stage for a larger clinical application of these approaches. Lifetime immunosuppression, however, required to protect the graft against recurrent autoimmune destruction and allorejection, raises serious doubts about the safety of its employment in children. While it is evident that children may be helped even more than adults by the possibility to correct diabetic metabolic disorders without exogenous insulin, and to lower in a more effective way the chance to develop secondary complications, the drawbacks of the currently used immunosuppressive drugs largely overcome the potential benefits. A great step forward for immediate applicability of transplantation to children involves the optimization of tolerogenic protocols and a better understanding of the concept of immune ignorance. Functional tolerance should be sufficient to entail the absence of immune reactivity against self- and graft antigens, while maintaining immune reactivity against other non-self, non-donor antigens. In addition, novel strategies aimed at utilizing surrogate beta cells obtained from non-islet cells, or by genetic manipulation of beta-cell precursors merit consideration as the use of xenogeneic donors. However, much work is still needed for their safe clinical implementation.
Collapse
Affiliation(s)
- Rita Bottino
- Diabetes Institute, Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|