Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mucosal Immunol ; 7(4): 753-62, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24736234

ABSTRACT

In this review, we examine the evidence that intestinal helminths can control harmful inflammatory responses and promote homeostasis by triggering systemic immune responses. Induction of separable components of immunity by helminths, which includes type 2 and immune regulatory responses, can both contribute toward the reduction in harmful type 1 immune responses that drive certain inflammatory diseases. Despite inducing type 2 responses, intestinal helminths may also downregulate harmful type 2 immune responses including allergic responses. We consider the possibility that intestinal helminth infection may indirectly affect inflammation by influencing the composition of the intestinal microbiome. Taken together, the studies reviewed herein suggest that intestinal helminth-induced responses have potent systemic effects on the immune system, raising the possibility that whole parasites or specific molecules produced by these metazoans may be an important resource for the development of future immunotherapies to control inflammatory diseases.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Host-Parasite Interactions/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Animals , Disease Models, Animal , Helminthiasis/complications , Helminthiasis/diagnosis , Helminthiasis/metabolism , Helminthiasis/microbiology , Helminthiasis/therapy , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/parasitology , Inflammation/pathology , Intestinal Diseases, Parasitic/complications , Intestinal Diseases, Parasitic/diagnosis , Intestinal Diseases, Parasitic/metabolism , Intestinal Diseases, Parasitic/microbiology , Intestinal Diseases, Parasitic/therapy , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Metabolic Syndrome/etiology , Microbiota
2.
Mucosal Immunol ; 6(2): 297-308, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806101

ABSTRACT

Helminth infection can prevent type 1 diabetes (T1D); however, the regulatory mechanisms inhibiting disease remain largely undefined. In these studies, nonobese diabetic (NOD) IL-4(-/-) mice were infected with the strictly enteric nematode parasite, Heligmosomoides polygyrus. Short-term infection, 5-7 weeks of age, inhibited T1D onset, as late as 40 weeks of age. CD4(+) T-cell STAT6 phosphorylation was inhibited, while suppressed signal transducer and activator of transcription 1 phosphorylation was sustained, as were increases in FOXP3(-), CD4(+) T-cell interleukin (IL)-10 production. Blockade of IL-10 signaling in NOD-IL-4(-/-), but not in NOD, mice during this short interval abrogated protective effects resulting in pancreatic ß-cell destruction and ultimately T1D. Transfer of CD4(+) T cells from H. polygyrus (Hp)-inoculated NOD IL-4(-/-) mice to NOD mice blocked the onset of T1D. These studies indicate that Hp infection induces non-T-regulatory cells to produce IL-10 independently of STAT6 signaling and that in this Th2-deficient environment IL-10 is essential for T1D inhibition.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Interleukin-10/immunology , Intestines/parasitology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Regulation , Insulin-Secreting Cells/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-4/deficiency , Islets of Langerhans/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Phenotype , Phosphorylation , Receptors, Interleukin-10/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism
3.
Diabetes ; 50(5): 1047-55, 2001 May.
Article in English | MEDLINE | ID: mdl-11334407

ABSTRACT

In addition to inhibiting matrix metalloproteinase-2 and matrix metalloproteinase-9 activity, recent studies suggest that tissue inhibitor of metalloproteinase (TIMP)-1 may inhibit apoptosis in various cell lines. To address this question in pancreatic islets and beta-cells, we treated rat pancreatic islets and INS-1 cells with a high-dose combination of the cytokines interleukin (IL)-1beta, tumor necrosis factor-alpha, and interferon-gamma with or without the addition of TIMP-1 and TIMP-2 protein. Using flow cytometry, we quantitated DNA fragmentation to assess cellular apoptosis and confirmed these observations with DNA laddering experiments. Next, we transfected the mouse TIMP-1 gene into INS-1 cells and performed Western immunoblotting to demonstrate expression of TIMP-1 protein. We treated TIMP-1-expressing INS-1 cells with high-dose cytokines and again used flow cytometry to assess DNA fragmentation. We also evaluated the effect of TIMP-1 on IL-1beta-induced inhibition of glucose-stimulated insulin secretion (GSIS) in freshly isolated rat pancreatic islets. Finally, we evaluated the effect of TIMP-1 on inducible nitric oxide synthase (iNOS) gene expression and nuclear factor (NF)-kappaB activity in INS-1 cells stimulated with high-dose cytokines. TIMP-1 but not TIMP-2 prevented cytokine-induced apoptosis and cytokine-mediated inhibition of GSIS in rat islets and beta-cells. TIMP-1 mediated these effects by inhibiting cytokine activation of NF-kappaB, but it did not affect nitric oxide production or iNOS gene expression. Therefore, TIMP-1 may be an ideal gene to prevent cytokine-mediated beta-cell destruction and dysfunction in models of type 1 diabetes and islet transplantation rejection.


Subject(s)
Apoptosis/drug effects , Cytokines/toxicity , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-1/physiology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Cytokines/antagonists & inhibitors , DNA Fragmentation/drug effects , Flow Cytometry , Gene Expression Regulation, Enzymologic/drug effects , Genes, Reporter , Glucose/pharmacology , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Interferon-gamma/toxicity , Interleukin-1/toxicity , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Luciferases/genetics , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Rats , Recombinant Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
4.
Gene Ther ; 7(18): 1553-61, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11021593

ABSTRACT

Insulin-dependent diabetes mellitus (IDDM) or type 1 diabetes is an autoimmune disease that results in destruction of the insulin-producing pancreatic islet beta cells. Several factors induce the invasion of immune cells into islets and trigger inflammation. Gene therapy approaches targeting the islet cells could be an effective treatment to prevent the onset or reverse type 1 diabetes. Allogeneic islet transplantation provides short-term treatment. However, genetically modified islets, which resist the host immune response, could provide long-term solutions. Adeno-associated virus (AAV) is emerging as a prominent vector system for delivering therapeutic genes for human gene therapy. AAV vector can transduce nondividing cells and provide long-term gene expression by integrating into host chromosome. Therefore, it is an appropriate vector system for islet cell gene therapy. To test the efficacy of AAV vector to transduce pancreatic endocrine cells, we constructed AAV vectors using plasmid pSub201. Wild-type AAV DNA analogue from plasmid psub201 was subcloned into a cloning plasmid pSP72 and AAV vectors were constructed by inserting the transgenes with heterologous promoter in place of AAV open reading frames (rep and cap). In this report we demonstrate the transduction of pancreatic islet cells with AAV vectors encoding bacterial -galactosidase enzyme or enhanced green fluorescent protein (EGFP) as reporter gene. Dispersed porcine and rat islet cells can be transduced by AAV vector, with an efficiency of 47% and 38%, respectively. In particular porcine islet insulin producing beta cells were transduced with an efficiency of 39%. Intact rat islet cells were transduced with an efficiency of 26% as estimated by FACS analysis following transduction with an AAV vector encoding EGFP. Transduction of intact rat islets with an AAV vector did not alter glucose-induced insulin secretion. AAV vector transduction was higher in transformed islet cell lines INS-1 and RIN m5F with an efficiency of 65% and 57%, respectively. These new results suggest that AAV vectors will provide an improved method of gene delivery to pancreatic islets and isolated pancreatic beta cells.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Type 1/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Islets of Langerhans/chemistry , Animals , Blotting, Southern , Blotting, Western , Cell Line , Cells, Cultured , Flow Cytometry , Gene Expression , Green Fluorescent Proteins , Histocytochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Rats , Swine , Transfection , beta-Galactosidase/analysis , beta-Galactosidase/genetics
5.
J Clin Invest ; 103(10): 1431-6, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10330425

ABSTRACT

Leukocyte 12-lipoxygenase (12-LO) gene expression in pancreatic beta cells is upregulated by cytotoxic cytokines like IL-1beta. Recent studies have demonstrated that 12-LO inhibitors can prevent glutamate-induced neuronal cell death when intracellular glutathione stores are depleted. Therefore, 12-LO pathway inhibition may prevent beta-cell cytotoxicity. To evaluate the role of 12-LO gene expression in immune-mediated islet destruction, we used 12-LO knockout (12-LO KO) mice. Male homozygous 12-LO KO mice and control C57BL/6 mice received 5 consecutive daily injections of low-dose streptozotocin to induce immune-mediated diabetes. Fasting serum glucose and insulin levels were measured at 7-day intervals, and the mice were followed up for 28 days. 12-LO KO mice were highly resistant to diabetes development compared with control mice and had higher serum insulin levels on day 28. Isolated pancreatic islets were treated with IL-1beta, TNF-alpha, and IFN-gamma for 18 hours. Glucose-stimulated insulin secretion in cytokine-treated islets from C57/BL6 mice decreased 54% from that of untreated islets. In marked contrast, the same cytokine mix led to only a 26% decrease in islets from 12-LO KO mice. Furthermore, cytokine-induced 12-hydroxyeicosatetraenoic acid (12-HETE) production was absent in 12-LO KO islets but present in C57/BL6 islets. Isolated peritoneal macrophages were stimulated for 48 hours with IFN-gamma + LPS and compared for nitrate/nitrite generation. 12-LO KO macrophages generated 50% less nitrate/nitrite when compared with C57BL/6 macrophages. In summary, elimination of leukocyte 12-LO in mice ameliorates low dose streptozotocin-induced diabetes by increasing islet resistance to cytokines and decreasing macrophage production of nitric oxide.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/genetics , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Recombinant Proteins , Superoxides/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Horm Res ; 52(2): 86-94, 1999.
Article in English | MEDLINE | ID: mdl-10681638

ABSTRACT

Several autoantibodies related to Type 1 diabetes mellitus and their corresponding autoantigens have been previously identified. While peptide antigens are more widely recognized, lipid antigens like sulfatides and gangliosides are also known epitopes for the diabetic humoral immune response. Islet cell antibodies (ICA) in Type 1 diabetes are heterogeneous immunoglobulins directed against selected antigens in the islets of Langerhans. Moreover, ICA may be the best predictive marker of disease in family members of patients with Type 1 diabetes. The aims of this study were: (1) to purify lipids from porcine pancreas that contain ICA epitopes; (2) to characterize these lipid antigens, and (3) to use the purified lipids in an assay to detect antibodies in patients with Type 1 diabetes. A unique family of 4 lysophospholipids, 1 fully characterized as lysophosphatidylmyoinositol, partially inhibited ICA staining, and therefore, were considered to be candidate antigens for an ICA immunoassay. Using a dot blot immunoassay, we detected antibodies directed against these phospholipids in 28 out of 46 (61%) diabetic sera, while detecting only 1 false positive out of 28 nondiabetic sera (3.6%; p < 0.0001 comparing diabetic vs. nondiabetic serum). Therefore, lysophospholipid immunoassay positivity is present in sera of Type 1 diabetic patients. Furthermore, we detected 15 out of 23 ICA-negative diabetic sera (65.2%), showing that our phospholipid immunoassay does not correlate with ICA positivity.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 1/immunology , Immunoblotting/methods , Lysophospholipids/immunology , Child , Diabetes Mellitus, Type 1/blood , Female , Gas Chromatography-Mass Spectrometry , Humans , Inositol/analysis , Islets of Langerhans/immunology , Male , Mass Screening , Mass Spectrometry
7.
Int J Mol Med ; 1(1): 265-72, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9852229

ABSTRACT

Leukocyte type 12-lipoxygenase (12-LO) catalyzes the conversion of arachidonic acid (AA; C20:4) to 12-hydroperoxyeicosatetraenoic acid (12-HPETE) and linoleic acid (LA; C18:2) to 13-hydroperoxyoctadecadienoic acid (13-HPODE). Previous studies have demonstrated that 12-LO, but not 5- or 15-lipoxygenase (5-LO, 15-LO respectively), is specifically expressed in pancreatic -cells and is involved in regulating glucose-stimulated insulin secretion. Lipoxygenase products also have been linked with inflammatory pathways in endothelial cells, kidney mesangial cells, inflammatory bowel disease, and corneal epithelial cells. Therefore, 12-LO may play a role in cytokine mediated inflammation in pancreatic beta-cells (i.e. beta -cell dysfunction and cytotoxicity). Cytokines such as IL-1 stimulate both de novo 12-LO protein synthesis and enzyme activity in pancreatic beta-cells. The products generated by 12-LO may ultimately be involved in cellular events that lead to lipid peroxidation. Hydroperoxide and free radical production in beta-cells can activate intracellular signaling pathways that lead to cell death or may directly damage mitochondrial and plasma membranes. Increased 12-LO expression has also been found in islets from prediabetic Zucker fatty rats, a model that demonstrates insulin secretory defects similar to human type 2 diabetes. In this review, we present an overview of the 12-LO pathway in regulating glucose-stimulated insulin secretion in beta-cells as well as more recent data which supports the hypothesis that the 12-LO pathway participates in cytokine mediated beta-cell dysfunction and cytotoxicity.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Islets of Langerhans/metabolism , Animals , Cytokines/toxicity , Humans , Insulin/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats
8.
Biochem Biophys Res Commun ; 230(2): 448-51, 1997 Jan 13.
Article in English | MEDLINE | ID: mdl-9016800

ABSTRACT

Cytokine induced pancreatic beta-cell destruction seen in Type 1 diabetes and islet graft rejection involves multiple intracellular signaling pathways that directly or indirectly lead to inflammatory damage or programmed cell death. IL-1beta has been shown to stimulate the 12-lipoxygenase pathway product 12-HETE, in RIN m5F cells; however, the precise role of 12-LO activation in mediating cytokine effects is not clear. Since the stress-activated protein kinase, JNK, has been linked to cytokine mediated inflammatory actions, we studied the effect of two LO products, 12-HETE and 15-HETE, on JNK activity. We demonstrate that 1 nM 12-HETE stimulates JNK activity, while 1 nM 15-HETE, the 15-lipoxygenase pathway product, does not. These results suggest 12-HETE is a novel upstream signal for IL-1beta induced JNK activation in RIN m5F cells.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Animals , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Cell Line , Enzyme Activation , Hydroxyeicosatetraenoic Acids/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans , JNK Mitogen-Activated Protein Kinases , Kinetics , Phosphorylation , Rats
9.
Endocrinology ; 137(11): 4871-7, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8895359

ABSTRACT

Interleukin-1 beta (IL-1 beta) causes rat islet cell dysfunction through mechanisms that involve inducible nitric oxide synthase (iNOS). However, IL-1 beta also activates several lipid pathways, including those generating phosphatidic acid (PA). Lisofylline (LSF), a water-soluble, nontoxic, selective inhibitor of the PA-1 alpha subspecies, which is stimulated by IL-1 beta and tumor necrosis factor-alpha, has been shown to prevent cytokine-induced cytotoxicity in in vivo animal models. To evaluate the effect of LSF on acute IL-1 beta-induced islet dysfunction, rat islets were exposed to IL-1 beta (0.1 ng/ml) with or without LSF (100 microM) for 24 h, followed by 25 mM glucose (G) stimulation, measurement of rat insulin by RIA, and calculation of the insulin secretion rate. In other experiments, rat islets were precultured for 48 h, then treated for 48 h in 25 mM G with or without IL-1 beta (0.1 ng/ml) and LSF (400 microM), and aliquots of medium were removed at 0, 24, and 48 h for measurement of rat insulin. In addition, islets were exposed to 25 mM G with or without IL-1 beta and LSF, lipids were then extracted, and PA subspecies were identified by TLC and mass spectroscopy, and quantitated using normal phase HPLC. Islets were also exposed to IL-1 beta with or without LSF, and Western immunoblots were performed to evaluate the effect of LSF on iNOS protein expression. IL-1 beta caused a 44% decrease in islet G-stimulated insulin secretion compared to that in untreated islets (P < 0.0005), which was totally reversed by LSF. In addition, IL-1 beta decreased the G-stimulated medium insulin content by 75% at 24 h (P = 0.0004) and 86% at 48 h compared to that in control islets (P < 0.0001). LSF-treated islets maintained 70% of medium insulin content at 24 h (P = 0.11) and 50% at 48 h (P < 0.0001) compared to control islets. HPLC quantitation of PA-1 alpha extracted from islets treated with IL-1 beta alone showed an approximately 15-fold increase over the PA-1 alpha content of islets treated with IL-1 beta and LSF. IL-1 beta-induced expression of iNOS was unchanged with the addition of LSF. These results suggest that LSF is effective in reducing IL-1 beta-induced islet dysfunction, thus supporting the role of lipid mediators such as PA in cytokine-induced islet toxicity.


Subject(s)
Insulin/metabolism , Interleukin-1/pharmacology , Islets of Langerhans/physiology , Pentoxifylline/analogs & derivatives , Phosphatidic Acids/biosynthesis , Adjuvants, Immunologic/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Drug Interactions , Glucose/pharmacology , Insulin Secretion , Interleukin-1/antagonists & inhibitors , Islets of Langerhans/drug effects , Male , Pentoxifylline/pharmacology , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/chemistry , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Fast Atom Bombardment
10.
Endocrinology ; 136(12): 5736-44, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7588331

ABSTRACT

The leukocyte type of 12-lipoxygenase (12-LO) may play a role in inflammatory reactions in many cell types through the conversion of arachidonic acid to proinflammatory eicosanoids that include 12-hydroperoxyeicosatetraenoic acid and 12-hydroeicosatetraenoic acid. Previous studies demonstrating the presence of a functional 12-LO pathway in rat and human pancreatic beta-cells plus the recent cloning of a rat leukocyte type of 12-LO allowed us to evaluate whether inflammatory cytokines such as interleukin-1 beta (IL-1 beta) can regulate the beta-cell 12-LO enzyme pathway, thus providing a potential link between the cytotoxic effects of cytokines on pancreatic beta-cells and the proinflammatory effects of 12-LO products. We demonstrate that IL-1 beta induces 12-LO protein and messenger RNA (mRNA) expression in RIN m5F cells and 12-LO mRNA expression in rat islets. RIN m5F cells treated for 16 h with IL-1 beta (25, 50, and 100 ng/liter) showed a maximal 2-fold increase in the expression of a leukocyte form of 12-LO demonstrated by Western blots. A concomitant increase in 12-LO mRNA expression was seen at this time point using a highly sensitive competitive polymerase chain reaction assay. The increase in mRNA and protein expression was preceded by increased 12-LO pathway activity measured by a RIA for 12-S-HETE. Separate experiments using purified Sprague-Dawley rat islets also showed increased expression of 12-LO mRNA and enzyme activity in response to IL-1 beta. These results demonstrate that IL-1 beta can up-regulate 12-LO expression and activity in rat beta-cells.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Interleukin-1/pharmacology , Islets of Langerhans/enzymology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Animals , Arachidonate 12-Lipoxygenase/genetics , Base Sequence , Cells, Cultured , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Immunohistochemistry , Male , Molecular Sequence Data , Nitric Oxide Synthase/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
11.
Diabetes ; 42(10): 1433-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8375582

ABSTRACT

Fathers with type I diabetes transmit diabetes to their offspring 2-3 times more frequently than mothers with type I diabetes. This phenomenon has provoked both genetic and nongenetic hypotheses, but the mechanism remains obscure. We find that mothers who develop diabetes before age 8 transmit diabetes at the same rate as diabetic fathers, and that the sex difference in diabetes transmission is explained by a decreased transmission rate in mothers who acquired diabetes after age 8. We constructed a data base containing 2156 nondiabetic and diabetic offspring of parents with type I diabetes. Families were selected from our main data base, which contains demographic information and diabetes autoantibody test results on > 8000 first-degree relatives of patients with type I diabetes and diabetic probands. Identification of offspring was made through diabetic parents who had participated in our autoantibody screening program at the Joslin Diabetes Center between 1983 and 1990. Questionnaires were sent to all other family members to determine the number of diabetic and nondiabetic offspring in each family. The 20-yr life-table risk of diabetes in offspring of diabetic fathers and mothers is 8.9 +/- 1.0 and 3.4 +/- 0.6%, respectively. For mothers acquiring diabetes before or after age 8, the risk of diabetes in offspring is 13.9 +/- 4.4 and 2.4 +/- 0.6% at 20 yr of age, respectively. Furthermore, we find that duration of diabetes in mothers before pregnancy has no effect on the risk of diabetes in their offspring.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Aging/metabolism , Androgens/metabolism , Diabetes Mellitus, Type 1/epidemiology , Sex Characteristics , Adolescent , Adrenal Cortex/metabolism , Adult , Age Factors , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Disease Susceptibility , Family Health , Female , Humans , Incidence , Infant , Infant, Newborn , Life Tables , Male , Maternal Age , Paternal Age , Risk Factors , Time Factors
12.
Diabetes Care ; 13(2): 111-8, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2190768

ABSTRACT

We intensively studied 5 islet cell-antibody-positive (ICA+) first-degree relatives of type I (insulin-dependent) diabetic patients before overt diabetes. In total, 55 intravenous glucose tolerance tests (IVGTTs) and 83 fasting plasma glucose determinations were made over a maximum 4-yr period before diabetes. The 5 prediabetic relatives (not diabetic when initially studied but subsequently progressed to overt diabetes) as a group showed a progressive rise in fasting glucose (r = 0.58, P less than 0.001, slope = 23.1 mg.dl-1.yr-1) and glucose at 60 min in IVGTT (r = 0.46, P = 0.01, slope = 47.5 mg.dl-1.yr-1) beginning 1.5 yr before diabetes. During the 4.0- to 1.5-yr period before overt diabetes, no change was observed in fasting glucose or glucose at 60 min on IVGTT (fasting glucose: r = 0.21, P = 0.18, slope = 2.1 mg.dl-1.yr-1; 60-min glucose: r = 0.08, P = 0.72, slope = 2.9 mg.dl-1.yr-1). The positive predictive value for a fasting glucose greater than 108 mg/dl to be within 1.5 yr of diabetes was 100% (11 of 11 values). The negative predictive value of a stimulated insulin (1-min + 3-min insulin - 2 X basal insulin) level greater than 24 microU/ml to be greater than 1.5 yr from diabetes was 90% (9 of 10 values) and 100% (10 of 10 values) at greater than 1 yr from overt diabetes.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Autoantibodies/analysis , Biomarkers/analysis , Diabetes Mellitus, Type 1/genetics , Prediabetic State/genetics , Adolescent , Adult , Blood Glucose/analysis , Child , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Diseases in Twins , Family , Female , Glucose Tolerance Test , Humans , Insulin , Islets of Langerhans/immunology , Male , Prediabetic State/diagnosis , Prediabetic State/immunology , Time Factors , Twins, Monozygotic
15.
Hastings Cent Rep ; 5(6): 8-9, 1975 Dec.
Article in English | MEDLINE | ID: mdl-126967
SELECTION OF CITATIONS
SEARCH DETAIL