Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Clin Exp Immunol ; 174(2): 302-17, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23865694

ABSTRACT

While much is known about tolerogenic dendritic cell effects on forkhead box protein 3 (FoxP3)⁺ regulatory T cells, virtually nothing is known about their effects on another arm of immunoregulation that is mediated by a subpopulation of immunosuppressive B cells. These cells suppress rheumatoid arthritis, lupus and inflammatory bowel disease in mice, and functional defects have been reported in human lupus. We show that co-stimulation-impaired tolerogenic dendritic cells that prevent and reverse type 1 diabetes mellitus induce the proliferation of human immunosuppressive B cells in vitro. We also show that the suppressive properties of these B cells concentrate inside the CD19⁺ CD24⁺ B cell population and more specifically inside the CD19⁺ CD24⁺ CD38⁺ regulatory B cell population. We discovered that B cell conversion into suppressive cells in vitro is partially dependent on dendritic cell production of retinoic acid and also that CD19⁺ CD24⁺ CD38⁺ B regulatory cells express retinoic acid receptors. Taken together, our data suggest a model whereby part of the immunosuppressive properties of human tolerogenic dendritic cells could be mediated by retinoic acid which, in addition to its known role in favouring T cell differentiation to FoxP3⁺ regulatory T cells, acts to convert B cells into immunosuppressive cells.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD19/metabolism , CD24 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Immunosuppression Therapy , T-Lymphocytes, Regulatory/immunology , Tretinoin/immunology , Tretinoin/metabolism
2.
Clin Exp Immunol ; 165(1): 60-76, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21413939

ABSTRACT

We have identified a novel interleukin (IL)-7-responsive T cell population [forkhead box P3 (FoxP3(+) ) CD4(+) CD25(+) CD127(+) ] that is comparably functionally suppressive to conventional FoxP3(+) CD4(+) CD25(+) regulatory T cells (T(regs) ). Although IL-2 is the most critical cytokine for thymic development of FoxP3(+) T(regs) , in the periphery other cytokines can be compensatory. CD25(+) CD127(+) T cells treated with IL-7 phenotypically 'matured' into the known 'classical' FoxP3(+) CD4(+) CD25(high) CD127(-) FoxP3(+) T(regs) . In freshly isolated splenocytes, the highest level of FoxP3 expression was found in CD127(+) CD25(+) T cells when compared with CD127(-) CD25(+) or CD127(+) CD25(-) cells. IL-7 treatment of CD4(+) CD25(+) T cells induced an increase in the accumulation of FoxP3 in the nucleus in vitro. IL-7-mediated CD25 cell surface up-regulation was accompanied by a concurrent down-regulation of CD127 in vitro. IL-7 treatment of the CD127(+) CD25(+) FoxP3(+) cells also resulted in up-regulation of cytotoxic T lymphocyte antigen 4 without any changes in CD45RA at the cell surface. Collectively, these data support emerging evidence that FoxP3(+) T cells expressing CD127 are comparably functionally suppressive to CD25(+) CD127(-) FoxP3(+) T cells. This IL-7-sensitive regulation of FoxP3(+) T(reg) phenotype could underlie one peripheral non-IL-2-dependent compensatory mechanism of T(reg) survival and functional activity, particularly for adaptive T(regs) in the control of autoimmunity or suppression of activated effector T cells.


Subject(s)
Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-7/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , Active Transport, Cell Nucleus , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/biosynthesis , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
4.
Gene Ther ; 11(20): 1506-14, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15229635

ABSTRACT

Islet transplantation is a viable long-term therapeutic alternative to daily insulin replacement for type I diabetes. The allogeneic nature of the transplants poses immunological challenges for routine clinical utility. Gene transfer of immunoregulatory molecules and those that improve insulin release kinetics provides rational approaches to facilitate allogeneic islet transplantation as a potential therapy. We have examined the efficacy of a soluble type 1 tumor necrosis factor receptor (TNFR) immunoglobulin-Fc fusion transgene (TNFR-Ig) to protect human islets from cytokine-induced apoptosis in culture, as well as in facilitating allogeneic islet transplants in diabetic mice. Cultured human islets were transduced with an adenoviral vector encoding human TNFR-Ig (Ad-TNFR-Ig). TNFR-Ig protein was secreted by cultured islets, as well as by transduced mouse islet transplants recovered from mouse recipients. Glucose-induced insulin release kinetics were comparable among untransduced, Ad-TNFR-Ig-infected human islets and vector-transduced islets exposed to cytokines. In parallel, Ad-TNFR-Ig-infected islets were protected from cytokine-induced apoptosis activation. Finally, diabetic mice transplanted with allogeneic islets expressing TNFR-Ig returned to and maintained normoglycemia significantly longer than untransduced islet recipients. These data support the potential utility of TNFR-Ig gene transfer to islets as a means of facilitating allogeneic islet transplantation.


Subject(s)
Diabetes Mellitus/therapy , Genetic Therapy/methods , Immunoglobulin Fc Fragments/genetics , Islets of Langerhans Transplantation , Receptors, Tumor Necrosis Factor/genetics , Transduction, Genetic/methods , Adenoviridae/genetics , Adenovirus E1 Proteins/genetics , Adenovirus E3 Proteins/genetics , Analysis of Variance , Apoptosis/drug effects , Cell Culture Techniques , Diabetes Mellitus/pathology , Diabetes Mellitus/surgery , Gene Deletion , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Graft Survival , Humans , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Recombinant Fusion Proteins/genetics , Transplantation, Homologous , Tumor Necrosis Factor-alpha/pharmacology
5.
Gene Ther ; 10(10): 875-89, 2003 May.
Article in English | MEDLINE | ID: mdl-12732873

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.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Genetic Therapy/methods , Islets of Langerhans Transplantation , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/transplantation , Autoimmunity , Bone Marrow Transplantation , Chemokines/genetics , Chemokines/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Islets of Langerhans Transplantation/immunology , Mice , Models, Animal , Proinsulin/genetics , Stem Cell Transplantation , T-Lymphocytes/immunology , Transplantation, Homologous
6.
Diabetes Nutr Metab ; 15(3): 173-203, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12173733

ABSTRACT

Although diabetes mellitus is a manageable disorder, the associated complications that result in significant morbidity and mortality worldwide necessitate novel approaches of pharmacologic, cell, and gene therapy for an eventual cure. A significant number of animal studies have demonstrated the potential of restoring normoglycemia by islet transplantation in the context of immunoregulation achieved by gene transfer of immunoregulatory genes to allo- and xenogeneic islets ex vivo. Examples include viral vector-mediated gene transfer of immunosuppressive cytokines, proteins that block co-stimulation and molecules that prevent apoptotic cell death. 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 mellitus (T1DM) or rodent recipients of allogeneic/xenogeneic islet transplants. Gene transfer of putative autoantigens is one example. The achievements of gene and cell therapy in Type 2 diabetes mellitus (T2DM) are less evident, but seminal studies promise that this modality can be relevant to treat and perhaps prevent the underlying causes of the disease including obesity and insulin resistance. Herein, we present an overview of the current status of drug, gene and cell therapy for T1DM and T2DM and we propose novel therapeutic options that could be clinically useful.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Animals , Apoptosis , Autoantigens/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Cytokines/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/prevention & control , Genetic Therapy , Humans , Hypoglycemic Agents/therapeutic use , Immune Tolerance/genetics , Insulin Resistance , Islets of Langerhans Transplantation , Obesity/prevention & control , Transfection
7.
J Biol Chem ; 275(47): 36509-13, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10967112

ABSTRACT

Interleukin-1beta (IL-1beta) is a pro-inflammatory cytokine that inhibits beta cell function and promotes Fas-triggered apoptosis. IL-1beta is thought to act early in the initiation of the autoimmune destruction of pancreatic beta cells in type I diabetes. IL-1beta promotes beta cell impairment, in part, by activating NF-kappaB transcription factor-dependent signaling pathways. We have examined whether beta cells could be protected from the effects of IL-1beta by overexpressing an inhibitor of NF-kappaB activity, IkappaB, by adenoviral gene transfer to intact human islets in culture. Infection of islets with an adenoviral vector encoding a non-phosphorylatable, non-degradable variant of IkappaBalpha resulted in normal insulin responses to glucose in the presence of IL-1beta. Furthermore, nitric oxide production was prevented and, more importantly, Fas-triggered apoptosis was inhibited following IkappaBalpha gene transfer. These results suggest that blocking the NF-kappaB pathway might prevent cytokine-induced beta cell impairment as a means of facilitating islet transplantation.


Subject(s)
DNA-Binding Proteins/genetics , I-kappa B Proteins , Interleukin-1/pharmacology , Islets of Langerhans/drug effects , NF-kappa B/antagonists & inhibitors , Adenoviridae , Cells, Cultured , DNA-Binding Proteins/pharmacology , Genetic Vectors , Humans , Insulinoma/genetics , Insulinoma/metabolism , Islets of Langerhans Transplantation , NF-KappaB Inhibitor alpha , Nitric Oxide/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction
8.
Mol Ther ; 1(5 Pt 1): 430-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10933964

ABSTRACT

Dendritic cells (DC) classically promote immune responses but can be manipulated to induce antigen-specific hyporesponsiveness in vitro. The expression of costimulatory molecules (CD40, CD86, CD80) at the DC cell surface correlates with their capacity to induce or suppress immune responses. Expression of these molecules is associated with NF-kB-dependent transcription of their genes. DC tolerogenicity has been associated with impaired NF-kB-dependent transcription of costimulatory genes as well as NF-kB translocation to the nucleus. In this report, we demonstrate that double-stranded oligodeoxyribonucleotides containing binding sites for NF-kB (NF-kB ODN) are efficiently incorporated by bone marrow-derived DC and specifically inhibit NF-kB-dependent transcription of a reporter gene. Moreover, exposure of DC to the oligonucleotide decoys inhibited lipopolysaccharide (LPS)-induced nitric oxide production, a marker of DC maturation. Treatment of bone marrow-derived DC progenitors with NF-kB ODN selectively suppressed the cell-surface expression of costimulatory molecules without interfering with MHC class I or class II expression. Furthermore, NF-kB ODN DC induced allogeneic donor-specific hyporesponsiveness in mixed leukocyte cultures, and this was associated with inhibition of Th1-type cytokine production. Finally, infusion of NF-kB ODN-modified bone marrow-derived DC into allogeneic recipients prior to heart transplantation resulted in significant prolongation of allograft survival in the absence of immunosuppression. Specific interference with NF-kB and other transcriptional pathways involved in immune stimulation in DC using ODN decoy approaches could be one means to promote tolerance induction in organ transplantation.


Subject(s)
Dendritic Cells/physiology , Graft Survival , Heart Transplantation/physiology , NF-kappa B/metabolism , Oligodeoxyribonucleotides/pharmacology , Animals , Antigens, CD/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/drug effects , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Nitric Oxide/biosynthesis , T-Lymphocytes/immunology , Transplantation, Homologous
9.
Gene Ther ; 7(23): 2015-22, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11175313

ABSTRACT

Interleukin-1beta is a potent pro-inflammatory cytokine that has been shown to inhibit islet beta cell function as well as to activate Fas-mediated apoptosis in a nitric oxide-dependent manner. Furthermore, this cytokine is effective in recruiting lymphocytes that mediate beta cell destruction in IDDM onset. The insulin-like growth factor I (IGF-I) has been shown to block IL-1beta actions in vitro. We hypothesized that gene transfer of the insulin-like growth factor I to intact human islets could prevent IL-1beta-induced beta cell dysfunction and sensitization to Fas-triggered apoptosis activation. Intact human islets were infected with adenoviral vectors encoding IGF-I as well as beta-galactosidase and enhanced green fluorescent protein as controls. Adenoviral gene transfer of human IGF-I prevented IL-1beta-mediated nitric oxide production from human islets in vitro as well as the suppression of beta cell function as determined by glucose-stimulated insulin production. Moreover, IGF-I gene transfer prevented IL-1beta-induced, Fas-mediated apoptosis. These results suggest that locally produced IGF-I from cultured islets may be beneficial in maintaining beta cell function and promoting islet survival before and following islet transplantation as a potential therapy for type I diabetes.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Genetic Therapy/methods , Insulin-Like Growth Factor I/genetics , Islets of Langerhans/metabolism , Transfection/methods , Adenoviridae/genetics , Analysis of Variance , Apoptosis , Cells, Cultured , Gene Expression , Genetic Vectors/pharmacology , Glucose/pharmacology , Green Fluorescent Proteins , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Like Growth Factor I/metabolism , Interleukin-1/physiology , Luminescent Proteins/genetics , Nitric Oxide/physiology , beta-Galactosidase/genetics
10.
Endocrinol Metab Clin North Am ; 28(4): 841-64, x, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10609123

ABSTRACT

Type 1 diabetes is the outcome of a progressive and selective destruction of insulin-producing cells in the pancreatic islets of Langerhans. The precise cause and mechanism(s) that trigger the insulin-producing cell destruction are still unclear, although it is well accepted that an autoimmune process plays a central role in diabetes development among genetically susceptible children. Additionally, certain viral infections, especially those caused by Coxsackievirus B, have been associated with the onset of type 1 diabetes. Possible gene therapy-based prevention and intervention strategies are discussed, based on the most accepted models of type 1 diabetes pathogenesis.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Humans , Lymphocyte Activation , T-Lymphocytes/immunology
11.
Diabetes ; 48(11): 2107-21, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535443

ABSTRACT

The autoimmune nature of insulin-dependent, or type 1, diabetes targets the beta-cells of the pancreas for destruction and results in a lifelong commitment to insulin replacement therapy. Although the number of formulations and dosing of insulin have become more sophisticated and more efficient in recent years, insulin therapy alone is unable to prevent nephropathy, retinopathy, or vascular and heart disease, which still occur in a large number of patients. Different approaches have been attempted to eliminate the requirement of exogenous insulin administration. Historically, these have included pancreatic and islet transplants, which were later combined with treatments intended to halt the destructive process directed against the islets. Despite significant advances made in all of these areas, each approach faces a hostile immunological response that frequently ends with the loss of the islets. Gene therapy-based approaches add a new dimension to the efforts aimed at specifically blocking the immunological attack against the islets in genetically at-risk individuals (autoimmunity) or the immunological response against transplanted allogeneic islets (rejection). This new technology may have an important role in the therapy and cure of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Genetic Therapy , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Diabetic Nephropathies/prevention & control , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Transplantation, Heterologous
12.
Gene Ther ; 6(9): 1499-511, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10490759

ABSTRACT

Facilitation of solid organ and cell transplantation depends on metabolic and immunologic factors that can be manipulated ex vivo and in vivo using gene transfer technology. Vectors have been developed which can optimally transfer relevant genes to various tissues and organs. Interventions aimed at promoting tissue preservation before transplantation, prevention of oxidative stress and immunological rejection have recently become attractive options using viral and nonviral gene delivery vehicles. Further understanding of the mechanisms involved in tolerance induction as well as the facilitation of xenogeneic engraftment have made possible a variety of avenues that can be exploited using gene transfer technology.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Graft Rejection/prevention & control , Transplantation Immunology , Animals , Apoptosis , Dendritic Cells , Gene Targeting/methods , Genetic Vectors/genetics , Humans , Immunosuppression Therapy , Reperfusion Injury/prevention & control , Transplantation, Heterologous
13.
Gene Ther ; 6(9): 1545-51, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10490763

ABSTRACT

The transfer of genes encoding immunomodulatory proteins to islets can be used to improve islet function, block apoptosis, and inhibit rejection following transplantation. Adenoviral vectors have been shown to infect intact human islets, but the immunogenicity and transient gene expression of the current adenoviral vectors may hinder their use clinically for islet transplantation. In this report, we compared an HIV-1-based lentiviral vector with the E1-deleted adenoviral vehicle of the Ad5 type for gene transfer to human islets in vitro. We demonstrate that at similar viral particle concentrations per islet that an HIV-based lentiviral vector is able to infect beta-cells within an intact human islet at an efficiency similar to an adenoviral vector. In addition, both the adenoviral and lentiviral vectors were able to express significant levels of soluble interleukin-1 receptor antagonist (IL-1Ra) protein following infection of intact islets. More importantly, there was no impairment of islet beta-cell function following adenoviral and lentiviral infection in responding to glucose stimulation. These results support the utility of replication-defective lentiviral vectors as efficient gene delivery vehicles to islets to faciliate transplantation of islets for therapy of type I diabetes.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , HIV-1/genetics , Islets of Langerhans/virology , Sialoglycoproteins/genetics , Adenoviridae/genetics , Apoptosis , Cells, Cultured , Defective Viruses , Flow Cytometry , Gene Expression , Glucose/pharmacology , Green Fluorescent Proteins , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Interleukin 1 Receptor Antagonist Protein , Islets of Langerhans/metabolism , Luminescent Proteins/genetics , Recombinant Proteins/metabolism , beta-Galactosidase/genetics
14.
Diabetes ; 48(9): 1730-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10480601

ABSTRACT

The beta-cells in the pancreatic islets of Langerhans are the targets of autoreactive T-cells and are destroyed in type 1 diabetes. Macrophage-derived interleukin-1beta (IL-1beta) is important in eliciting beta-cell dysfunction and initiating beta-cell damage in response to microenvironmental changes within islets. In particular, IL-1beta can impair glucose-stimulated insulin production in beta-cells in vitro and can sensitize them to Fas (CD95)/FasL-triggered apoptosis. In this report, we have examined the ability to block the detrimental effects of IL-1beta by genetically modifying islets by adenoviral gene transfer to express the IL-1 receptor antagonist protein. We demonstrate that adenoviral gene delivery of the cDNA encoding the interleukin-1 receptor antagonist protein (IL-1Ra) to cultured islets results in protection of human islets in vitro against IL-1beta-induced nitric oxide formation, impairment in glucose-stimulated insulin production, and Fas-triggered apoptosis activation. Our results further support the hypothesis that IL-1beta antagonism in in situ may prevent intra-islet proinsulitic inflammatory events and may allow for an in vivo gene therapy strategy to prevent insulitis and the consequent pathogenesis of diabetes.


Subject(s)
Adenoviridae/genetics , Apoptosis/physiology , Gene Transfer Techniques , Islets of Langerhans/pathology , Receptors, Interleukin-1/antagonists & inhibitors , Sialoglycoproteins/genetics , Cells, Cultured , Genetic Engineering , Glucose , Humans , Interleukin 1 Receptor Antagonist Protein , Islets of Langerhans Transplantation , Nitric Oxide/biosynthesis , Recombinant Proteins/genetics , Transplantation, Homologous , Virus Replication
15.
J Biol Chem ; 273(23): 14158-64, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9603916

ABSTRACT

The minisatellite DNA polymorphism consisting of a variable number of tandem repeats (VNTR) at the human INS (insulin gene) 5'-flanking region has demonstrated allelic effects on insulin gene transcription in vitro and has been associated with the level of insulin gene expression in vivo. We now show that this VNTR also has effects on the nearby insulin-like growth factor II gene (IGF2) in human placenta in vivo and in the HepG2 hepatoma cell line in vitro. We show that higher steady-state IGF2 mRNA levels are associated with shorter alleles (class I) than the longer class III alleles in term placentae. In vitro, reporter gene activity was greater from reporter gene constructs with IGF2 promoter 3 in the presence of class I alleles than from those with class III. Taken together with the documented transcriptional effects on the insulin gene, we propose that the VNTR may act as a long range control element affecting the expression of both INS and IGF2. The localization of a type 1 diabetes susceptibility locus (IDDM2) to the VNTR itself suggests that either or both of these genes may be involved in the biologic effects of IDDM2.


Subject(s)
Gene Expression Regulation/genetics , Insulin-Like Growth Factor II/genetics , Insulin/genetics , Minisatellite Repeats/genetics , Alleles , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Diabetes Mellitus, Type 1/genetics , Genes, Reporter/genetics , Genotype , Humans , Placenta/metabolism , Polymorphism, Genetic/genetics , Polymorphism, Restriction Fragment Length , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics
16.
Biochem Biophys Res Commun ; 220(3): 1014-9, 1996 Mar 27.
Article in English | MEDLINE | ID: mdl-8607783

ABSTRACT

In most non-neoplastic tissues studied to date, IGF2 is expressed only from the paternal allele and H19 is expressed only from the maternal allele. The choroid plexus, the only normal tissue to date where IGF2 is expressed from both parental alleles, does not express H19. We present an additional situation in which biallelic IGF2 expression is associated with the absence of H19 transcription in normal tissue: blood cells. In blood cells, functional IGF2 imprinting was found to be a polymorphic trait among individuals: expression was biallelic in 79 out of 85 individuals, but the remaining 6 expressed a single allele. Only the latter expressed H19. Finally, the familial clustering of functional IGF2 imprinting in blood cells suggests that the trait may be genotype-dependent.


Subject(s)
Blood Cells/metabolism , Genomic Imprinting , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Muscle Proteins/genetics , Placenta/metabolism , Polymorphism, Genetic , RNA, Untranslated , Alleles , Cesarean Section , Female , Fetal Blood , Gene Expression , Humans , Male , Pedigree , Pregnancy , RNA, Long Noncoding , Transcription, Genetic
17.
Diabetologia ; 38(6): 715-9, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7672495

ABSTRACT

Although association of insulin-dependent diabetes mellitus with a haplotype at a locus encompassing the genes for insulin and the insulin-like growth factor II has been well established, two major studies disagree as to whether linkage to this locus is confined to paternally inherited alleles, or is present in alleles transmitted from either parental sex. Towards resolving this discrepancy, we examined parent-of-origin specific association rather than linkage, using the haplotype relative risk method in a mixed Caucasian population. We find that the haplotype relative risk (HRR) conferred by paternal chromosomes was much higher (5.1, p < 0.01) than the corresponding maternal value (2.3, p = 0.07), which narrowly failed to reach statistical significance. Thus, although we cannot exclude an effect of the maternal allele, such an effect appears to be considerably weaker. We review evidence that parental imprinting is genotype-dependent, which may explain the different degrees to which the paternal effect is seen in different populations.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Insulin/genetics , Adult , Alleles , Child , Chromosome Mapping , Chromosomes, Human, Pair 11 , Female , Genetic Linkage , Genetic Markers , Genetic Predisposition to Disease , Genotype , Haplotypes , Heterozygote , Humans , Male , Odds Ratio , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length
18.
Dev Genet ; 17(3): 253-62, 1995.
Article in English | MEDLINE | ID: mdl-8565331

ABSTRACT

Parental genomic imprinting is the phenomenon in which the behavior of a gene is modified, depending on the sex of the transmitting parent [Peterson and Sapienza (1993): Annu Rev Genet 27:7-31]. Recent observations have revealed that the inheritance patterns, age-of-onset, severity, and etiology of certain human diseases can be explained by aberrations in the establishment or the maintenance of the imprint. Examples include the Prader-Willi, Angelman, and Beckwith-Wiedemann syndromes [Nicholls (1994): Am J Hum Genet 54:733-740], malignancy [Sapienza (1990): Biochim Biophys Acta 1072:51-61; Feinberg (1993): Nat Genet 4:110-113], and insulin-dependent diabetes mellitus (IDDM) [Julier et al. (1994) Nature 354:155-159; Bennett et al. (1995) Nat Genet 9:284-292]. We review the evidence that implicates an imprinted gene in the INS-IGF2 region of chromosome 11p15 in the etiology of IDDM (referred to as the IDDM2 locus) and show that in human fetal pancreas, INS is not imprinted, thus providing an argument against INS as the candidate gene. We also examine imprinting effects on the expression of IGF2 in components of the human immune system believed to be important in IDDM and show imprinted expression in fetal thymus as early as 15 weeks gestation. We demonstrate further that in the circulating mononuclear cells of two individuals, lectin-stimulated IGF2 transcription was biallelic, indicating relaxation of imprinting, whereas in one individual, transcription was monoallelic. Finally, we review the current available data supporting a role for insulin-like growth factor-II (IGF-II) in the immune system and, more specifically, discuss the evidence supporting a role for the IGFs in the prevention of apoptosis. These data have led us to formulate a novel hypothesis that could mechanistically explain the involvement of the IDDM2 locus in the pathogenesis of IDDM.


Subject(s)
Apoptosis/genetics , Diabetes Mellitus, Type 1/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Apoptosis/physiology , Cells, Cultured , Chromosomes, Human, Pair 11 , Diabetes Mellitus, Type 1/immunology , Humans , Insulin/genetics , Insulin-Like Growth Factor II/physiology , Pancreas/embryology , Pancreas/metabolism
19.
Nat Genet ; 4(1): 98-101, 1993 May.
Article in English | MEDLINE | ID: mdl-8099843

ABSTRACT

The mouse igf2 gene, coding for the insulin-like growth factor II (IGF-II) is parentally imprinted, only the gene copy derived from the father is expressed. To know whether IGF2, the human homologue, is also imprinted, we used an ApaI polymorphism at the 3' untranslated region in order to distinguish between mRNA derived from each copy of the gene in placentae from heterozygote human fetuses, studied after careful removal of the decidua. Six term and two pre-term placentae of heterozygotes were studied, and in each case the cDNA contained only one of the two alleles present in the genomic DNA. In three cases the mother was homozygous for the non-expressed allele, allowing assignment of paternal origin to the transcribed gene copy. We conclude that, as in the mouse, human IGF2 is parentally imprinted.


Subject(s)
Gene Expression Regulation , Insulin-Like Growth Factor II/genetics , Alleles , Animals , Base Sequence , Deoxyribonucleases, Type II Site-Specific , Gestational Age , Heterozygote , Humans , Mice/genetics , Molecular Sequence Data , Parents , Placenta/chemistry , Polymorphism, Restriction Fragment Length , Prader-Willi Syndrome/genetics , RNA, Messenger/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...