Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters











Publication year range
1.
Diabetologia ; 45(9): 1292-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12242462

ABSTRACT

AIMS/HYPOTHESIS: Insulin receptor null mutant mice develop severe diabetes, ketoacidosis and liver steatosis and die within 1 week after birth. Since the liver plays an essential role in the control of glucose homeostasis, we examined in this work whether the metabolic disorders of insulin receptor-deficient mice could be improved upon restoration of hepatic glucose metabolism by transgenic constitutive overexpression of glucokinase selectively in the liver. METHODS: We first generated transgenic mice overexpressing rat glucokinase cDNA under control of the liver-specific phenylalanine hydroxylase gene promoter. These transgenic mice were crossed with heterozygous insulin-receptor-null mutants to produce homozygous insulin-receptor-null mice overexpressing glucokinase in the liver. RESULTS: The transgenic mice overexpressing glucokinase in the liver showed improved glucose tolerance and were mildly hypoglycaemic and hyperlipidaemic under starved conditions. The introduction of the glucokinase transgene in insulin receptor null mice did not prevent the development of glycosuria. However, ketoacidosis was delayed by more than 1 week and survival was prolonged to 10 to 16 days in 16% of the pups. In these longer surviving pups, serum glucose and triglyceride concentrations were lowered, hepatic glycogen stores were reconstituted and liver steatosis was absent as compared with the pups which had developed strong ketoacidosis and died earlier. CONCLUSIONS/INTERPRETATION: These results show that overexpression of hepatic glucokinase can compensate, in part, for the metabolic disorders developed by insulin receptor-deficient mice. This shows the importance of improving hepatic function in diabetes and must revive interest in enhancement of glucokinase activity as a therapeutic strategy for the treatment of diabetes.


Subject(s)
Blood Glucose/metabolism , Glucokinase/genetics , Receptor, Insulin/deficiency , Receptor, Insulin/physiology , Animals , Base Sequence , Cloning, Molecular , Crosses, Genetic , DNA, Complementary , Diabetic Ketoacidosis/genetics , Glucokinase/metabolism , Glucose Tolerance Test , Glycosuria/genetics , Homozygote , Humans , Liver/cytology , Liver/enzymology , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity , Phenylalanine Hydroxylase/genetics , Promoter Regions, Genetic , Rats , Receptor, Insulin/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
J Neuroendocrinol ; 14(9): 737-44, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213135

ABSTRACT

The vasopressin V3 receptor (V3) is specifically expressed in pituitary corticotropes and mediates the stimulatory effect of vasopressin on adrenocorticotropic hormone (ACTH) release. The V3 gene is overexpressed in corticotrope pituitary tumours compared to normal pituitaries. We hypothesized that V3 overexpression might induce changes in corticotrope function and alter the regulation of the hypothalamic-pituitary-adrenal axis. Thus, we generated transgenic mice (POMV3) expressing the human V3 receptor in the pituitary under the control of rat pro-opiomelanocortin (POMC) promoter sequences. The transgene was efficiently transcribed and vasopressin binding was increased in both corticotropes and melanotropes. In-vitro ACTH release and inositol phosphate formation were unchanged in POMV3 pituitaries, but the responses to vasopressin were significatively increased. In vivo, basal circulating concentrations of ACTH in POMV3 mice were similar to those of controls but corticosterone concentrations were moderately increased. In addition, the levels of POMC mRNA in the transgenic pituitaries were comparable to those of control mice. Finally, POMV3 mice responded with a similar maximal increase of ACTH and corticosterone to a 20-min acute restraint stress. Together, these results show that hypophyseal V3 overexpression led to increased basal concentrations of corticosterone and suggest that the negative glucocorticoid feedback may be altered at the pituitary level.


Subject(s)
Corticosterone/metabolism , Pituitary Gland/physiology , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Animals , Female , Gene Expression , Humans , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Mice, Transgenic , Pituitary-Adrenal System/physiology , Promoter Regions, Genetic/physiology , RNA, Messenger/analysis , Stress, Physiological/physiopathology
3.
Endocrinology ; 143(4): 1530-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897712

ABSTRACT

The targeted disruption of the two nonallelic insulin genes in mouse was reported previously to result in intrauterine growth retardation, severe diabetes immediately after suckling, and death within 48 h of birth. We have further used these animals to investigate the morphology and cell biology of the endocrine pancreas in late gestation and at birth when insulin is absent throughout development. Pancreatic beta-cells were identified by detecting the activity of the LacZ gene inserted at the Ins2 locus. A significant increase in the mean area of the islets was found at embryonic d 18.5 (E18.5) and in the newborn in Ins1-/-, Ins2-/- animals compared with Ins1-/-, Ins2+/- and wild-type controls, whereas the blood glucose levels were unaltered. The individual size of the beta-cells in the insulin-deficient fetuses was similar to controls, suggesting that the relative increase in islet size was due to an increase in cell number. Immunohistochemistry for proliferating cell nuclear antigen within the pancreatic ductal epithelium showed no differences in labeling index between insulin-deficient and control mice, and no change in the number of beta-cells associated with ducts, but the relative size distribution of the islets was altered so that fewer islets under 5,000 microm(2) and more islets greater than 10,000 microm(2) were present in Ins1-/-, Ins2-/- animals. This suggests that the greater mean islet size seen in insulin-deficient animals represented an enlargement of formed islets and was not associated with an increase in islet neogenesis. The proportional contribution of alpha- and beta-cells to the islets was not altered. This was supported by an increase in the number of cells containing immunoreactive proliferating cell nuclear antigen in both islet alpha- and beta-cells at E18.5 in insulin-deficient mice, and a significantly lower incidence of apoptotic cells, as determined by molecular histochemistry using the terminal deoxynucleotidyl transferase-mediated deoxy-UTP nick end labeling reaction. The density of blood vessels within sections of whole pancreas, or within islets, was determined by immunohistochemistry for the endothelial cell marker CD31 and was found to be increased 2-fold in insulin-deficient mice compared with controls at E18.5. However, no changes were found in the steady-state expression of mRNAs encoding vascular endothelial growth factor, its receptor Flk-1, IGF-I or -II, the IGF-I and insulin receptors, or insulin receptor substrates-1 or -2 in pancreata from Ins1-/-, Ins2-/- mice compared with Ins1-/-, Ins2+/- controls. Thus, we conclude that the relative hyperplasia of the islets in late gestation in the insulin-deficient mice was due to an increased islet cell proliferation coupled with a reduced apoptosis, which may be related to an increased vascularization of the pancreas.


Subject(s)
Apoptosis/genetics , Apoptosis/physiology , Insulin/deficiency , Insulin/genetics , Islets of Langerhans/cytology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Animals , Body Weight , Cell Division/physiology , Embryonic and Fetal Development/genetics , Embryonic and Fetal Development/physiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Glucagon/metabolism , Hyperplasia/pathology , Immunohistochemistry , Insulin/metabolism , Insulin-Like Growth Factor II/biosynthesis , Islets of Langerhans/blood supply , Islets of Langerhans/growth & development , Lymphokines/biosynthesis , Lymphokines/genetics , Mice , Mice, Knockout , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Regional Blood Flow , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Diabetes ; 50 Suppl 1: S146-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272177

ABSTRACT

Insulin receptor (IR)-deficient pups rapidly become hyperglycemic and hyperinsulinemic and die of diabetic ketoacidosis within a few days. Immunocytochemical analysis of the endocrine pancreas revealed that IR deficiency did not alter islet morphology or the number of beta-, alpha-, delta-, and pancreatic polypeptide (PP) cells. The lack of IR did not result in major changes in the expression of islet hormone genes or of beta-cell-specific marker genes encoding pancreas duodenum homeobox-containing transcription factor-1 (PDX-1), glucokinase (GCK), and GLUT2, as shown by reverse transcriptase-polymerase chain reaction analysis. The serum glucagon levels in IR-deficient and nondiabetic littermates were comparable. Finally, total insulin content in the pancreas of IR-deficient pups was gradually depleted, indicating sustained insulin secretion, not compensated for by increased insulin biosynthesis. These findings are discussed in light of recent results suggesting a role of IR in beta-cell function.


Subject(s)
Homeodomain Proteins , Islets of Langerhans/metabolism , Receptor, Insulin/genetics , Animals , Animals, Newborn , Female , Gene Expression , Genotype , Glucagon/genetics , Glucagon/metabolism , Glucokinase/genetics , Glucose Transporter Type 2 , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/chemistry , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Monosaccharide Transport Proteins/genetics , Pancreatic Polypeptide/genetics , Pancreatic Polypeptide/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/deficiency , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/genetics , Somatostatin/metabolism , Trans-Activators/genetics
5.
Diabetes ; 50 Suppl 1: S150-3, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272179

ABSTRACT

Intrauterine growth retardation and postnatal acute diabetes result from insulin deficiency in double homozygous null mutants for Ins1 and Ins2 (Duvillié B, et al., Proc. Natl. Acad. Sci. USA 94:5137-5140, 1997). The characterization of single homozygous null mutants for Ins1 or Ins2 is described here. Neither kind of mutant mice was diabetic. Immunocytochemical analysis of the islets showed normal distribution of the endocrine cells producing insulin, glucagon, somatostatin, or pancreatic polypeptide. Analysis of the expression of the functional insulin gene in Ins1-/- or Ins2-/- mice revealed a dramatic increase of Ins1 transcripts in Ins2-/- mutants. This compensatory response was quantitatively reflected by total pancreatic insulin content similar for both types of mutants and wild-type mice. Moreover, both mutants had normal plasma insulin levels and normal glucose tolerance tests. The determination of beta-cell mass by morphometry indicated beta-cell hyperplasia in the mutant mice. The beta-cell mass in Ins2-/- mice was increased almost threefold, which accounts for the increase of Ins1 transcripts in Ins2-/-mutants. This study thus contributes to evaluate the potential of increasing the beta-cell mass to compensate for low insulin production.


Subject(s)
Insulin/genetics , Islets of Langerhans/metabolism , Animals , Blotting, Western , Cell Count , Female , Gene Expression , Glucagon/analysis , Hyperplasia/genetics , Hyperplasia/metabolism , Immunohistochemistry , Insulin/blood , Insulin/deficiency , Islets of Langerhans/chemistry , Islets of Langerhans/cytology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mutation , Pancreatic Polypeptide/analysis , Proinsulin/analysis , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/analysis
6.
FEBS Lett ; 488(3): 174-8, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11163767

ABSTRACT

We have derived skeletal muscle cell lines from wild-type (wt) and insulin receptor (IR) knockout mice to unravel the metabolic potential of IGF-1 receptor (IGF-1R). Both wt and IR(-/-) myoblasts differentiated into myotubes with similar patterns of expression of muscle-specific genes such as MyoD, myogenin and MLC1A indicating that IR is not required for this process. Binding of 125I-IGF-1 on wt and IR(-/-) myotubes was similar showing that IGF-1R was not upregulated in the absence of IR. Stimulation of IR(-/-) myotubes with IGF-1 (10(-10) to 10(-7) M) increased glucose uptake and incorporation into glycogen, induced IRS-1 phosphorylation and activated PI 3-kinase and MAP kinase, two enzymes of major signaling pathways. These effects were comparable to those obtained with wt myotubes using insulin or IGF-1 or with IR(-/-) myotubes using insulin at higher concentrations. This study provides a direct evidence that IGF-1R can represent an alternative receptor for metabolic signaling in muscle cells.


Subject(s)
Gene Deletion , Muscle, Skeletal/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/deficiency , Animals , Animals, Newborn , Binding Sites , Biomarkers , Cells, Cultured , Culture Media, Serum-Free , Deoxyglucose/metabolism , Glucose/metabolism , Glycogen/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor, Insulin/genetics , Signal Transduction/drug effects
7.
J Immunol ; 166(5): 3083-9, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207259

ABSTRACT

Neoplastic B cells from H chain disease patients express a truncated B cell receptor (BCR), comprising a membrane Ig that lacks part of its extracellular domain. It has been speculated that deletion of the Ag binding domain would confer a constitutive activity on the BCR, as it has been shown for oncogenic growth factor receptors. A V region-less BCR has constitutive activity, because in transgenic mice it causes inhibition of endogenous H chain gene rearrangements and relieves the requirement for surrogate L chain in pre-B cell development. However, it has been speculated that normal Ag receptors also display constitutive activity. Here we show that transgenic B cells expressing a membrane H chain disease protein on their surface are phenotypically and functionally similar to B cells developing in the presence of their cognate Ag and that cells with normal levels of mutant BCR are eliminated in spleen via a bcl-2 sensitive pathway while progressing toward the mature stage. In contrast, cells with lower levels of mutant receptors develop as mature B cells. These findings support the view that the truncated BCR has a constitutive activity that mimics ligand binding, in analogy to what has been shown for oncogenic growth factor receptors.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Immune Tolerance/genetics , Receptors, Antigen, B-Cell/biosynthesis , Receptors, Antigen, B-Cell/genetics , Transgenes/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Clonal Anergy/genetics , Crosses, Genetic , Gene Rearrangement, B-Lymphocyte, Light Chain , Humans , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/genetics , Immunoglobulin mu-Chains/biosynthesis , Immunoglobulin mu-Chains/genetics , Immunophenotyping , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Antigen, B-Cell/deficiency , T-Lymphocytes/immunology
8.
J Immunol ; 164(4): 1681-8, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657610

ABSTRACT

"Immune privilege" is defined as tissue resistance to aggression by specifically activated lymphocytes, and involves the interaction between Fas expressed on infiltrating cells and Fas ligand (FasL) constitutively expressed on the target tissue. To test whether ectopic expression of FasL on thyrocytes could prevent autoimmune aggression of the thyroid by activated lymphoid cells, three lines of transgenic mice expressing low, intermediate, and high levels of functional FasL on thyroid follicular cells were generated. Experimental autoimmune thyroiditis was induced by immunization with mouse thyroglobulin. In all of the experiments, the effects were dependent on the level of FasL expression. Low and intermediate expression had no or only weak preventive effects, respectively, whereas high FasL expression strongly inhibited lymphocytic infiltration of the thyroid. Anti-mouse thyroglobulin-proliferative and cytotoxic T cell responses, as well as autoantibody production, were diminished in transgenic mice expressing high levels of FasL relative to controls. Furthermore, in these latter mice Th1 responses to mouse thyroglobulin were profoundly down-regulated, uncovering a new potential role for FasL in peripheral tolerance to organ-specific Ags. In sum, the prevention of experimental autoimmune thyroiditis by FasL on thyrocytes is dependent on the level of FasL expression.


Subject(s)
Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Thyroid Gland/immunology , Thyroid Gland/metabolism , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/prevention & control , fas Receptor/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Crosses, Genetic , Cytotoxicity, Immunologic/genetics , Fas Ligand Protein , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Injections, Intradermal , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Ligands , Lymph Nodes/immunology , Lymph Nodes/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Promoter Regions, Genetic/immunology , T-Lymphocytes, Cytotoxic/immunology , Thyroglobulin/administration & dosage , Thyroglobulin/genetics , Thyroglobulin/immunology , Thyroid Gland/cytology , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/pathology , Tumor Cells, Cultured
9.
J Biol Chem ; 275(5): 3021-4, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10652280

ABSTRACT

Ferritin molecules play an important role in the control of intracellular iron distribution and in the constitution of long term iron stores. In vitro studies on recombinant ferritin subunits have shown that the ferroxidase activity associated with the H subunit is necessary for iron uptake by the ferritin molecule, whereas the L subunit facilitates iron core formation inside the protein shell. However, plant and bacterial ferritins have only a single type of subunit which probably fulfills both functions. To assess the biological significance of the ferroxidase activity associated with the H subunit, we disrupted the H ferritin gene (Fth) in mice by homologous recombination. Fth(+/-) mice are healthy, fertile, and do not differ significantly from their control littermates. However, Fth(-/-) embryos die between 3.5 and 9.5 days of development, suggesting that there is no functional redundancy between the two ferritin subunits and that, in the absence of H subunits, L ferritin homopolymers are not able to maintain iron in a bioavailable and nontoxic form. The pattern of expression of the wild type Fth gene in 9.5-day embryos is suggestive of an important function of the H ferritin gene in the heart.


Subject(s)
Embryonic and Fetal Development/genetics , Ferritins/genetics , Gene Expression Regulation, Developmental , Animals , Homozygote , Mice , Mice, Knockout
10.
J Sleep Res ; 8 Suppl 1: 65-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10389109

ABSTRACT

Recently, insulin synthesis and the presence of an insulin receptor have been demonstrated in the brain. Intracerebroventricular infusion of insulin causes a selective increase in the amount of slow-wave sleep. In the present study, the sleep-wake cycle of transgenic mice, with or without habenular neuronal expression of the human insulin gene, was studied to investigate the possible role of brain insulin as a sleep modulator. Slow-wave sleep duration was increased in those mice expressing human insulin in the habenula. However, it is possible that this effect was not due to expression of the insulin transgene, but to the genetic background of one of the parental strains (CBA) used for insertion of the transgene. Users of transgenic mice should be aware of this possibility and be cautious in interpreting results when hybrid embryos are used as transgene recipients.


Subject(s)
DNA Transposable Elements/genetics , Insulin/genetics , Sleep/genetics , Wakefulness/genetics , Animals , Electroencephalography , Gene Deletion , Humans , Male , Mice , Mice, Transgenic , RNA, Messenger/genetics , Transgenes/genetics
11.
Hum Mol Genet ; 8(7): 1291-301, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10369875

ABSTRACT

Factor VIII-associated gene B ( F8B ) is a small human gene of unknown function which is nested within the gene encoding coagulation factor VIII ( FVIII ) in chromosome band Xq28. The sequence of F8B includes the C2 cell adhesion motif of factor VIII, which has also been identified in numerous proteins known to play important roles during development. Here we have constructed both chimeric and transgenic mice expressing normal human F8B to investigate its possible developmental effects. The chimeras produced from embryonic stem cells transfected with normal F8B under control of a cytomegalovirus promoter and selected for neomycin resistance expressed readily detectable levels of F8B mRNA in multiple tissues. They showed growth retardation, microcephaly, reduced longevity and severe ocular defects, and although they were fertile, gave birth to no F8B heterozygous pups. Seven transgenic mouse lines, produced by injection of the transgene into fertilized oocytes, were viable and of normal size but expressed lower levels of F8B mRNA. Strikingly, they showed the same severe eye abnormalities as the chimeras. These defects included anterior segment dysgenesis, absent or abnormal lens, persistence of the primary vitreous, Harderian gland tumors and ectopic pigmented cells, suggesting that migration of neural crest cells might have been perturbed during eye development. In addition, dysplastic retinas and the absence of photoreceptors were observed, providing a mouse model for retinal degeneration.


Subject(s)
Eye Abnormalities/genetics , Factor VIII/genetics , Animals , Cell Adhesion , Chimera , Eye Abnormalities/pathology , Humans , Male , Mice , Mice, Transgenic , Protein Conformation
13.
Biochem J ; 335 ( Pt 2): 193-204, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9761714

ABSTRACT

The expression of a number of genes encoding key players in insulin signalling and action, including insulin, insulin receptor (IR), downstream signalling molecules such as insulin receptor substrate-1 (IRS-1) and IRS-2, glucose transporters (GLUT4, GLUT2) and important metabolic enzymes such as glucokinase, has now been altered in transgenic or knockout mice. Such mice presented with phenotypes ranging from mild defects, revealing complementarity between key molecules or pathways, to severe diabetes with ketoacidosis and early postnatal death. Insulin action could also be improved by overproduction of proteins acting at regulatory steps. The development of diabetes by combining mutations, which alone do not lead to major metabolic alterations, validated the 'diabetogenes' concept of non-insulin-dependent diabetes mellitus. Genes encoding insulin-like growth factors (IGF-I and IGF-II) and their type I receptor (IGF-IR) have also been disrupted. It appears that although IR and IGF-IR are both capable of metabolic and mitogenic signalling, they are not fully redundant. However, IR could replace IGF-IR if efficiently activated by IGF-II. Studies with cell lines lacking IR or IGF-IR lend support to such conclusions. Concerning the issues of specificity and redundancy, studies with cell lines derived from IRS-1-deficient mice showed that IRS-1 and IRS-2 are also not completely interchangeable.


Subject(s)
Insulin/metabolism , Mice, Knockout/genetics , Mice, Transgenic/genetics , Muscle Proteins , Receptor, Insulin/genetics , Signal Transduction , Animals , Genetic Engineering , Glucose Transporter Type 2 , Glucose Transporter Type 4 , Insulin/genetics , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, Insulin/metabolism , Somatomedins/genetics , Somatomedins/metabolism
14.
J Biol Chem ; 273(32): 20175-9, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685363

ABSTRACT

USF1 and USF2 are ubiquitous transcription factors of the basic helix-loop-helix leucine zipper family. They form homo- and heterodimers and recognize a CACGTG motif termed E box. In the liver, USF binding activity is mainly accounted for by the USF1/USF2 heterodimer, which binds in vitro the glucose/carbohydrate response elements (GlRE/ChoRE) of glucose-responsive genes. To assign a physiological role of USFs in vivo, we have undertaken the disruption of USF1 and USF2 genes in mice. We present here the generation of USF1-deficient mice. In the liver of these mice, we demonstrate that USF2 remaining dimers can compensate for glucose responsiveness, even though the level of total USF binding activity is reduced by half as compared with wild type mice. The residual USF1 binding activity was similarly reduced in the previously reported USF2 -/- mice in which an impaired glucose responsiveness was observed (Vallet, V. S., Henrion, A. A., Bucchini, D., Casado, M. , Raymondjean, M., Kahn, A., and Vaulont, S. (1997) J. Biol. Chem. 272, 21944-21949). Taken together, these results clearly suggest differential transactivating efficiencies of USF1 and USF2 in promoting the glucose response. Furthermore, they support the view that USF2 is the functional transactivator of the glucose-responsive complex.


Subject(s)
Gene Expression Regulation/genetics , Glucose/pharmacology , Liver/physiology , Transcription Factors/genetics , Animals , DNA-Binding Proteins/analysis , Dimerization , Mice , Mice, Knockout , Nuclear Proteins , Protein Conformation , Proteins/genetics , Pyruvate Kinase/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Upstream Stimulatory Factors
15.
Biochem Biophys Res Commun ; 246(2): 426-30, 1998 May 19.
Article in English | MEDLINE | ID: mdl-9610377

ABSTRACT

The ability of insulin and insulin-like growth factors (IGF-I and IGF-II) to induce differentiation of 3T3-F442A cells into adipocytes was examined at various hormone concentrations. Both insulin and the IGFs promoted differentiation at concentrations compatible with binding to their cognate receptors, suggesting that both insulin and IGF-I receptors are capable of promoting this differentiation. Adipocyte conversion of 3T3-F442A cells was completely blocked in the presence of LY294002, a specific inhibitor of PI 3-kinase, indicating that PI 3-kinase activity plays a crucial role in the initial signalling events that trigger this differentiation process.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Insulin/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Receptor, IGF Type 1/metabolism , 3T3 Cells , Adipocytes/cytology , Animals , Cell Differentiation/drug effects , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Mice , Morpholines/pharmacology , Signal Transduction
16.
Mol Cell Biochem ; 182(1-2): 161-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9609125

ABSTRACT

Transgenic and gene targeting approaches have now been applied to a number of genes in order to investigate the metabolic disorders that would result by manipulating insulin action or pancreatic beta-cell function in the mouse. The availability of such mutant mice will allow in the future to develop animal models in which the pathophysiologies resulting from polygenic defects might be reconstituted and studied in detail. Such animal models hopefully will lead to better understanding of complex polygenic diseases such as non-insulin-dependent diabetes mellitus (NIDDM).


Subject(s)
Insulin/genetics , Insulin/physiology , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Animals , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic
17.
FEBS Lett ; 426(3): 381-5, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9600271

ABSTRACT

Cell systems derived from knockout mice for the insulin receptor (IR) or the IGF-1 receptor (IGF-1R) represent unique tools for dissecting complex interplay in the actions of insulin and insulin-like growth factors through their cognate versus non-cognate receptor. In this study, we used a fibroblast cell line derived from IR-deficient mice to investigate metabolic and mitogenic effects of IGF-1 and insulin. IGF-1 was able to stimulate glucose uptake, glucose incorporation into glycogen and thymidine incorporation in such cells. Phosphatidylinositol 3-kinase and mitogen-activated protein kinase, two enzymes of major metabolic-mitogenic signaling pathways, were activated upon stimulating these cells with IGF-1. All these effects were also achieved when IR-deficient cells were stimulated with insulin. Thus, IGF-1R can represent an alternative receptor through which insulin might exert some of its effects.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Receptor, Insulin/deficiency , Animals , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Knockout , Mitogens/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Thymidine/metabolism
18.
Genomics ; 47(1): 52-7, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9465295

ABSTRACT

The mouse gene encoding preproinsulin 2 (Ins2) is located on the distal end of chromosome 7 in a region of several hundred kilobases that contains several imprinted genes. The exclusive expression of the Ins2 paternal allele in the visceral yolk sac during the last part of gestation indicates that Ins2 also is imprinted. However, in other tissues in which Ins2 is expressed, both alleles are active at all developmental stages. Taking advantage of two mouse strains carrying different null mutations introduced at the Ins2 locus via homologous recombination in ES cells, we examined whether genes inserted at the Ins2 locus become imprinted and have the same restricted pattern of monoallelic expression. In the first null allele, Ins2 was replaced by LacZ, under the control of the endogenous Ins2 promoter, and a Neo cassette with its own promoter was inserted 3' to LacZ (Zneo allele). In the second null allele, Ins2 and its promoter were replaced by the same Neo cassette (Neo allele). Expression of the maternally and paternally inherited genes was monitored by RT-PCR performed on various reciprocal crosses involving the two mutants and the wildtype alleles. In (Zneo x wildtype) F1 embryos, the pattern of LacZ expression was similar to that of Ins2; i.e., LacZ is expressed in the yolk sac only when paternally inherited, while its expression in the embryo proper is independent of its paternal or maternal origin. For both of the mutant alleles, Neo was transcribed only when paternally inherited, in the yolk sac as well as in the embryo. Unexpectedly, we found that LacZ transcription on the maternal chromosome varied depending on the nature of the allele on the paternal chromosome. While fully expressed in the embryo when the paternal chromosome carries the wildtype allele, the maternally inherited LacZ is extinguished when the paternal allele is the Neo allele. The major conclusion from our results is that individual genes introduced into an imprinted chromosomal domain can become imprinted, indicating the influence of long-range cis-acting effects. In addition, our data suggest that the two parental alleles may "communicate" with each other and influence the transcription at the locus.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Imprinting , Proinsulin/genetics , Protein Precursors/genetics , Regulatory Sequences, Nucleic Acid , Animals , Crosses, Genetic , Embryo, Mammalian/physiology , Heterozygote , Insulin , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Mutation , Polymerase Chain Reaction , Recombination, Genetic , Transcription, Genetic , beta-Galactosidase/genetics
19.
J Biol Chem ; 272(35): 21944-9, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9268329

ABSTRACT

Upstream stimulatory factors (USF) 1 and 2 belong to the Myc family of transcription factors characterized by a basic/helix loop helix/leucine zipper domain responsible for dimerization and DNA binding. These ubiquitous factors form homo- and heterodimers and recognize in vitro a CACGTG core sequence termed E box. Through binding to E boxes of target genes, USF factors have been demonstrated to activate gene transcription and to enhance expression of some genes in response to various stimuli. In particular, in the liver USF1 and USF2 have been shown to bind in vitro glucose/carbohydrate response elements of glycolytic and lipogenic genes and have been proposed, from ex vivo experiments, to be involved in their transcriptional activation by glucose. However, the direct involvement of these factors in gene expression and nutrient gene regulation in vivo has not yet been demonstrated. Therefore, to gain insight into the specific role of USF1 and USF2 in vivo, and in particular to determine whether the USF products are required for the response of genes to glucose, we have created, by homologous recombination, USF2 -/- mice. In this paper, we provide the first evidence that USF2 proteins are required in vivo for a normal transcriptional response of L-type pyruvate kinase and Spot 14 genes to glucose in the liver.


Subject(s)
Gene Expression , Glucose/metabolism , Helix-Loop-Helix Motifs , Liver/metabolism , Transcription Factors/physiology , Animals , DNA-Binding Proteins/physiology , Dietary Carbohydrates/pharmacology , Dimerization , Gene Targeting , Liver/embryology , Mice , Mice, Knockout , Nuclear Proteins , Promoter Regions, Genetic , Proteins/genetics , Pyruvate Kinase/genetics , RNA, Messenger/metabolism , Upstream Stimulatory Factors
20.
Proc Natl Acad Sci U S A ; 94(10): 5137-40, 1997 May 13.
Article in English | MEDLINE | ID: mdl-9144203

ABSTRACT

Two mouse insulin genes, Ins1 and Ins2, were disrupted and lacZ was inserted at the Ins2 locus by gene targeting. Double nullizygous insulin-deficient pups were growth-retarded. They did not show any glycosuria at birth but soon after suckling developed diabetes mellitus with ketoacidosis and liver steatosis and died within 48 h. Interestingly, insulin deficiency did not preclude pancreas organogenesis and the appearance of the various cell types of the endocrine pancreas. The presence of lacZ expressing beta cells and glucagon-positive alpha cells was demonstrated by cytochemistry and immunocytochemistry. Reverse transcription-coupled PCR analysis showed that somatostatin and pancreatic polypeptide mRNAs were present, although at reduced levels, accounting for the presence also of delta and pancreatic polypeptide cells, respectively. Morphometric analysis revealed enlarged islets of Langherans in the pancreas from insulin-deficient pups, suggesting that insulin might function as a negative regulator of islet cell growth. Whether insulin controls the growth of specific islet cell types and the molecular basis for this action remain to be elucidated.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Growth Disorders/genetics , Insulin/deficiency , Animals , Animals, Newborn , Bone Development , DNA Primers , Death , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Growth Disorders/pathology , Growth Disorders/physiopathology , Heterozygote , Insulin/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Liver/pathology , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Polymerase Chain Reaction , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL