Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cells ; 9(11)2020 11 13.
Article in English | MEDLINE | ID: mdl-33202992

ABSTRACT

We have previously used a hepatotropic adeno-associated viral (AAV) vector with a modified human insulin gene to treat diabetic mice. The HLP (hybrid liver-specific promoter) used was constitutively active and non-responsive to glucose. In this study, we examined the effects of addition of glucose responsive elements (R3G) and incorporation of a 3' albumin enhancer (3'iALB) on insulin expression. In comparison with the original promoter, glucose responsiveness was only observed in the modified promoters in vitro with a 36 h lag time before the peak expression. A 50% decrease in the number of viral particles at 5 × 109 vector genome (vg)/mouse was required by AAV8-R3GHLP-hINSco to reduce the blood sugar level to near normoglycemia when compared to the original AAV8-HLP-hINSco that needed 1 × 1010 vg/mouse. The further inclusion of an 860 base-pairs 3'iALB enhancer component in the 3' untranslated region increased the in vitro gene expression significantly but this increase was not observed when the packaged virus was systemically injected in vivo. The addition of R3G to the HLP promoter in the AAV8-human insulin vector increased the insulin expression and secretion, thereby lowering the required dosage for basal insulin treatment. This in turn reduces the risk of liver toxicity and cost of vector production.


Subject(s)
Dependovirus/metabolism , Diabetes Mellitus, Experimental/therapy , Genetic Therapy , Hepatocytes/drug effects , Animals , Dependovirus/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Gene Expression/drug effects , Genetic Therapy/methods , Genetic Vectors/pharmacology , Glucose/metabolism , Hepatocytes/metabolism , Humans , Insulin/metabolism , Mice , Promoter Regions, Genetic/genetics , Transgenes/drug effects
2.
Gene Ther ; 26(1-2): 40-56, 2019 02.
Article in English | MEDLINE | ID: mdl-30514969

ABSTRACT

We report the restoration of euglycaemia in chemically induced diabetic C57BL/6 mice and spontaneously diabetic Non Obese Diabetic (NOD) mice by intravenous systemic administration of a single-stranded adeno-associated virus (ssAAV2/8) codon optimised (co) vector encoding furin cleavable human proinsulin under a liver-specific promoter. There were no immunological barriers to efficacy of insulin gene therapy in chemically induced C57BL/6 mice, which enjoyed long-lasting correction of hyperglycaemia after therapy, up to 250 days. Euglycaemia was also restored in spontaneously diabetic NOD mice, although these mice required a 7-10-fold higher dose of vector to achieve similar efficacy as the C57BL/6 mice and the immunodeficient NODscid mice. We detected CD8+ T cell reactivity to insulin and mild inflammatory infiltration in the livers of gene therapy recipient NOD mice, neither of which were observed in the treated C57BL/6 mice. Efficacy of the gene therapy in NOD mice was partially improved by targeting the immune system with anti-CD4 antibody treatment, while transfer of NOD mouse AAV2/8-reactive serum to recipients prevented successful restoration of euglycaemia in AAV2/8-HLP-hINSco-treated NODscid mice. Our data indicate that both immune cells and antibodies form a barrier to successful restoration of euglycaemia in autoimmune diabetic recipient mice with insulin gene therapy, but that this barrier can be overcome by increasing the dose of vector and by suppressing immune responses.


Subject(s)
Dependovirus/immunology , Diabetes Mellitus, Experimental/therapy , Genetic Therapy/adverse effects , Immunosuppression Therapy/methods , Insulin/immunology , Animals , CD4 Antigens/immunology , Dependovirus/genetics , Genetic Therapy/methods , HEK293 Cells , Humans , Insulin/genetics , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes/immunology
3.
J Gene Med ; 21(1): e3067, 2019 01.
Article in English | MEDLINE | ID: mdl-30592790

ABSTRACT

BACKGROUND: Diabetes mellitus is caused by a partial or complete lack of insulin production in the body. We have previously shown that a single injection of an adeno-associated virus serotype 8 (AAV8) vector carrying a modified and codon optimized human insulin gene induced hepatic production of insulin and corrected streptozotocin (STZ)-induced diabetes in mice for more than 1 year. Insulin production was constitutive, analogous to long-acting insulin therapy. METHODS: We have developed a single AAV8 vector with a Tet-Off regulatable system as a safety mechanism to turn off insulin secretion should hypoglycaemia develop in vector-treated diabetic mice. We first transfected HepG2 cells or freshly isolated rat hepatocytes in vitro with the Tet-Off system (pAAV-Tetoffbidir -Alb-luc) regulating a luciferase reporter gene. We subsequently incorporated a furin-cleavable codon-optimised human proinsulin cDNA into pAAV-Tetoffbidir backbone to form the doxycycline inducible pAAV-Tetoffbidir -Alb-hINSco. RESULTS: Using STZ-induced diabetic mice, we were able to switch off insulin secretion repeatedly with doxycycline administration, and showed full restoration of insulin secretion on withdrawing doxycycline. CONCLUSIONS: The present study provides proof of concept that, under circumstances when inappropriate basal insulin secretion is a safety concern, insulin secretion from AAV8 gene therapy can be turned off reversibly with doxycycline.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/therapy , Genetic Therapy , Genetic Vectors/genetics , Insulin/genetics , Liver/metabolism , Animals , Diabetes Mellitus, Experimental/diagnosis , Disease Models, Animal , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Genes, Reporter , Genetic Therapy/methods , Hepatocytes/metabolism , Humans , Mice , Molecular Imaging , Transfection
4.
J Gene Med ; 18(10): 312-321, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27572655

ABSTRACT

BACKGROUND: The lack of an ideal cell type that can be easily acquired, modified to produce insulin, and re-implanted has been a limitation for ex vivo insulin gene therapy. Canine diabetes is currently treated with human insulin and is a good model for human diabetes. Mesenchymal stromal cells (MSCs) are a promising candidate cell type for gene therapy. In the present study, we optimised insulin production using lentiviral transduced canine MSCs (cMSCs), aiming to evaluate their ability for use as surrogate beta cells. METHODS: Canine MSCs were derived from bone marrow and validated by measuring the expression of MSC lineage specific markers. Lentivirus vectors encoding the proinsulin gene (with or without a Kozak sequence) under the control of spleen focus forming virus, cytomegalovirus, elongation factor 1α and simian virus 40 promotors were generated and used to transduce primary cMSCs and a hepatocyte cell line. The insulin-producing capacity of transduced primary cMSCs was assessed by measuring the concentration of C-peptide produced. RESULTS: Primary cMSC could be readily expanded in culture and efficiently transduced using lentiviral vectors encoding proinsulin. Increasing the multiplicity of infection from 3 to 20 led to an increase in C-peptide secretion (from 1700 to 4000 pmol/l). The spleen focus forming virus promoter conferred the strongest transcriptional ability. CONCLUSIONS: The results of the present study suggest that optimised lentiviral transduction of the insulin gene into primary cMSCs renders these cells capable of secreting insulin over both the short- and long-term, in sufficient quantities in vitro to support their potential use in insulin gene therapy.


Subject(s)
Gene Expression , Insulin/genetics , Lentivirus/genetics , Mesenchymal Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Animals , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Dogs , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Proinsulin/genetics , Proinsulin/metabolism
6.
Curr Gene Ther ; 16(1): 65-72, 2016.
Article in English | MEDLINE | ID: mdl-26795016

ABSTRACT

We report the correction of hyperglycemia of STZ induced diabetic mice using one intravenous systemic administration of a single stranded serotype 8 pseudotyped adeno-associated virus (ssAAV2/8) vector encoding the human proinsulin gene under a constitutive liver specific promoter. In vivo dose titration experiments were carried out and we identified an optimal range that achieved maintenance of euglycaemia or a mild diabetic condition for at least 9 months and ongoing to beyond 1 year for some animals, accompanied by human C-peptide secretion and weight gain. Further DNA codon optimization of the insulin gene construct resulted in approximately 3-10 times more human C-peptide secreted in the blood of codon optimized treated animals thereby reducing the number of vector particles required to achieve the same extent of reduction in blood glucose levels as the non-codon optimized vector. The constitutive secretion of insulin achieved with a single administration of the vector could be of therapeutic value for some diabetic patients.


Subject(s)
Genetic Vectors/administration & dosage , Hyperglycemia/therapy , Insulin/genetics , Animals , C-Peptide/metabolism , Codon , Dependovirus/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/therapy , Humans , Hyperglycemia/genetics , Liver/metabolism , Mice, Inbred NOD , Pancreas/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics
8.
Cell Transplant ; 24(7): 1205-20, 2015.
Article in English | MEDLINE | ID: mdl-24667101

ABSTRACT

Cell therapy could potentially meet the need for pancreas and islet transplantations in diabetes mellitus that far exceeds the number of available donors. Bone marrow stromal cells are widely used in clinical trials mainly for their immunomodulatory effects with a record of safety. However, less focus has been paid to developing these cells for insulin secretion by transfection. Although murine models of diabetes have been extensively used in gene and cell therapy research, few studies have shown efficacy in large preclinical animal models. Here we report optimized conditions for ex vivo expansion and characterization of porcine bone marrow stromal cells and their permissive expression of a transfected insulin gene. Our data show that these cells resemble human bone marrow stromal cells in surface antigen expression, are homogeneous, and can be reproducibly isolated from outbred Yorkshire-Landrace pigs. Porcine bone marrow stromal cells were efficiently expanded in vitro to >10(10) cells from 20 ml of bone marrow and remained karyotypically normal during expansion. These cells were electroporated with an insulin expression plasmid vector with high efficiency and viability, and secreted human insulin and C-peptide indicating appropriate processing of proinsulin. We showed that autologous insulin-secreting bone marrow stromal cells implanted and engrafted in the liver of a streptozotocin-diabetic pig that modeled type 1 diabetes resulted in partial, but significant, improvement in hyperglycemia that could not be ascribed to regeneration of endogenous ß-cells. Glucose-stimulated insulin secretion in vivo from implanted cells in the treated pig was documented by a rise in serum human C-peptide levels during intravenous glucose tolerance tests. Compared to a sham-treated control pig, this resulted in significantly reduced fasting hyperglycemia, a slower rise in serum fructosamine, and prevented weight loss. Taken together, this study suggests that bone marrow stromal cells merit further development as autologous cell therapy for diabetes.


Subject(s)
Bone Marrow Cells/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Cell- and Tissue-Based Therapy , Diabetes Mellitus, Experimental , Disease Models, Animal , Humans , Mesenchymal Stem Cells/cytology , Swine
10.
Cell Transplant ; 22(1): 133-45, 2013.
Article in English | MEDLINE | ID: mdl-22710060

ABSTRACT

Harvesting, expansion, and directed differentiation of human bone marrow-derived mesenchymal stem cells (BM-MSCs) could provide an autologous source of surrogate ß-cells that would alleviate the limitations of availability and/or allogenic rejection following pancreatic or islet transplantation. Bone marrow cells were obtained from three adult type 2 diabetic volunteers and three nondiabetic donors. After 3 days in culture, adherent MSCs were expanded for two passages. At passage 3, differentiation was carried out in a three-staged procedure. Cells were cultured in a glucose-rich medium containing several activation and growth factors. Cells were evaluated in vitro by flow cytometry, immunolabeling, RT-PCR, and human insulin and c-peptide release in responses to increasing glucose concentrations. One thousand cell clusters were inserted under the renal capsule of diabetic nude mice followed by monitoring of their diabetic status. At the end of differentiation, ∼5-10% of cells were immunofluorescent for insulin, c-peptide or glucagon; insulin, and c-peptide were coexpressed. Nanogold immunolabeling for electron microscopy demonstrated the presence of c-peptide in the rough endoplasmic reticulum. Insulin-producing cells (IPCs) expressed transcription factors and genes of pancreatic hormones similar to those expressed by pancreatic islets. There was a stepwise increase in human insulin and c-peptide release by IPCs in response to increasing glucose concentrations. Transplantation of IPCs into nude diabetic mice resulted in control of their diabetic status for 3 months. The sera of IPC-transplanted mice contained human insulin and c-peptide but negligible levels of mouse insulin. When the IPC-bearing kidneys were removed, rapid return of diabetic state was noted. BM-MSCs from diabetic and nondiabetic human subjects could be differentiated without genetic manipulation to form IPCs that, when transplanted, could maintain euglycemia in diabetic mice for 3 months. Optimization of the culture conditions are required to improve the yield of IPCs and their functional performance.


Subject(s)
Bone Marrow Cells/metabolism , Diabetes Mellitus, Experimental/surgery , Insulin-Secreting Cells/transplantation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Adult , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Female , Gene Expression , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Nude , Middle Aged
11.
Clin Transpl ; : 111-2, 2013.
Article in English | MEDLINE | ID: mdl-25095498

ABSTRACT

Gene and stem cell therapy has been on the scientific agenda in many laboratories for more than 20 years. The literature is enormous, but practical applications have been few. Recently advances in stem cell biology and gene therapy are clarifying some of the issues. I have made a few observations concerning our own studies on bone marrow mesenchymal stem cells cultured to produce a small percentage of insulin-producing cells and human insulin gene engineered into Lenti and AA viruses. The aim of clinical application would still seem to be several years away, if all goes well. The first step will be to produce enough insulin-secreting cells to be of potential value to patients. The next crucial question will be how to persuade the cells to respond to blood glucose levels swiftly and appropriately. With both stem cell and gene therapy, another important factor will be to ensure that any positive results will continue long enough to be preferable to insulin injections.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Genetic Therapy/trends , Islets of Langerhans Transplantation/trends , Stem Cell Transplantation/trends , Humans
12.
Theor Med Bioeth ; 33(5): 359-76, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23054375

ABSTRACT

Peer review is a widely accepted instrument for raising the quality of science. Peer review limits the enormous unstructured influx of information and the sheer amount of dubious data, which in its absence would plunge science into chaos. In particular, peer review offers the benefit of eliminating papers that suffer from poor craftsmanship or methodological shortcomings, especially in the experimental sciences. However, we believe that peer review is not always appropriate for the evaluation of controversial hypothetical science. We argue that the process of peer review can be prone to bias towards ideas that affirm the prior convictions of reviewers and against innovation and radical new ideas. Innovative hypotheses are thus highly vulnerable to being "filtered out" or made to accord with conventional wisdom by the peer review process. Consequently, having introduced peer review, the Elsevier journal Medical Hypotheses may be unable to continue its tradition as a radical journal allowing discussion of improbable or unconventional ideas. Hence we conclude by asking the publisher to consider re-introducing the system of editorial review to Medical Hypotheses.


Subject(s)
Editorial Policies , Peer Review, Research , Periodicals as Topic , Research Report , Science , Selection Bias , Creativity , Humans , Observer Variation , Periodicals as Topic/ethics , Periodicals as Topic/standards , Periodicals as Topic/trends , Science/ethics , Science/standards , Science/trends
14.
Curr Opin Organ Transplant ; 16(4): 353-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21666472

ABSTRACT

PURPOSE OF REVIEW: To outline the rationale of powerful depleting induction therapy with alemtuzumab and minimal maintenance immunosuppression after organ transplantation. RECENT FINDINGS: The original observations in principle have been confirmed by many independent centres. SUMMARY: Follow-up of the 'prope tolerance' protocol has confirmed a low incidence of rejection, infection and post transplant lymphoproliferative disorder (PTLD). Especially, encouraging results were obtained in African-Americans. There were few side effects and the regimen was well tolerated by patients. Treg cells were observed in the circulation, which could be an important factor in the mechanisms of graft acceptance using a prope tolerance regimen. There was a considerable reduction in the costs of the transplantation procedure. It is suggested that this minimalisation of maintenance immunosuppression is the best therapy currently available that we can offer to our patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Organ Transplantation , Transplantation Tolerance/drug effects , Alemtuzumab , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/adverse effects , Graft Rejection/immunology , Humans , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents/adverse effects , Organ Transplantation/adverse effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Time Factors , Treatment Outcome
15.
Cell Transplant ; 20(11-12): 1827-41, 2011.
Article in English | MEDLINE | ID: mdl-21439131

ABSTRACT

In this study we describe the derivation and immunological characterization of a primary epithelial cell type from the human umbilical cord membrane. These cord lining epithelial cells (CLECs) expressed and/or secreted isoforms of the nonclassical human leukocyte antigen class I (HLA-1b) glycoproteins, HLA-G and E. Conditioned media from CLECs inhibited mitogen-stimulated T-lymphocyte responses, and in a mixed leukocyte reaction (MLR) assay, cocultured CLECs inhibited allogeneic responses with a concomitant reduction in proinflammatory cytokines. Using a transwell coculture system, it was demonstrated that these immunoregulatory effects were mediated by soluble factors secreted by CLECs, in a dose-dependent manner. Functional studies using HLA-G blocking antibody showed that the effects of CLEC-secreted products could be inhibited, thus demonstrating a significant and important role for soluble HLA-G. In vivo, we show that transplanted CLECs could be maintained for extended periods in immunocompetent mice where xenorejection rapidly destroyed primary keratinocytes, a control human epithelial cell type. Additionally, CLECs delayed the rejection of keratinocytes and extended their survival when cotransplanted, indicating an ability to protect adjacent human cell types that would otherwise be rejected if transplanted alone. We also show that CLECs transduced with a modified human proinsulin gene were transplanted intraperitoneally into streptozotocin (STZ)-induced diabetic mice, resulting in significantly lower levels of serum glucose compared to control mice. This study has characterized the immunological properties of CLECs and tested a potential therapeutic application in the treatment of a type 1 diabetes mouse model.


Subject(s)
Epithelial Cells/metabolism , Fetal Blood/cytology , Animals , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Experimental/therapy , Epithelial Cells/cytology , Epithelial Cells/transplantation , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , HLA-G Antigens/immunology , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/transplantation , Leukocytes, Mononuclear/cytology , Mice , Mice, SCID , Proinsulin/genetics , Proinsulin/metabolism , Transplantation, Heterologous , HLA-E Antigens
18.
Sci Prog ; 93(Pt 2): 141-50, 2010.
Article in English | MEDLINE | ID: mdl-20681319

ABSTRACT

Organ transplantation started in the mid-1950s with a kidney transplant between identical twins, demonstrating the surgical technique could provide successful therapy. The immunological barrier to be overcome, however proved to be far more difficult to deal with. The introduction of immunosuppressive agents produced some success but it was not until Cyclosporin became available in the 1980s that results became sufficiently good for widespread acceptance and rapid development of organ grafting. Now with more powerful and selective agents, although there is still much room for improvement in immunosuppression, one of the main problems in organ transplantation is a result of its success, namely a shortage of organ donors. In this review, I summarise these matters.


Subject(s)
Organ Transplantation/methods , Organ Transplantation/trends , Graft vs Host Disease/prevention & control , Humans , Immunosuppression Therapy , Organ Transplantation/ethics
19.
Nat Rev Endocrinol ; 6(3): 173-7, 2010 03.
Article in English | MEDLINE | ID: mdl-20173779

ABSTRACT

In this Perspectives article, we comment on the progress in experimental stem cell and gene therapies that might one day become a clinical reality for the treatment of patients with diabetes mellitus. Research on the ability of human embryonic stem cells to differentiate into islet cells has defined the developmental stages and transcription factors involved in this process. However, the clinical applications of human embryonic stem cells are limited by ethical concerns, as well as the potential for teratoma formation. As a consequence, alternative forms of stem cell therapies, such as induced pluripotent stem cells and bone marrow-derived mesenchymal stem cells, have become an area of intense study. Finally, gene therapy shows some promise for the generation of insulin-producing cells. Here, we discuss two of the most frequently used approaches: in vitro gene delivery into cells which are then transplanted into the recipient and direct delivery of genes in vivo.


Subject(s)
Diabetes Mellitus/therapy , Genetic Therapy/methods , Stem Cell Transplantation/methods , Animals , Humans
20.
Ann Acad Med Singap ; 38(4): 354-5, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19434339

ABSTRACT

In the past 50 years, organ transplantation has developed from an improbable laboratory exercise to a major therapeutic success. The surgical problems of organ grafting have, for the most part, been solved. Rejection of grafts is now partially understood and usually controllable by powerful immunosuppressive drugs. A steady improvement in patient outcome, especially following the introduction of cyclosporin as an immunosuppressive agent has resulted in a worldwide shortage of organs for transplantation. This has provoked serious ethical dilemmas in every country. These matters are summarised in the following text.


Subject(s)
Biomedical Research , Transplants , Cyclosporine/pharmacology , Graft Rejection/drug therapy , Graft Rejection/prevention & control , Humans , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Transplants/ethics
SELECTION OF CITATIONS
SEARCH DETAIL
...