Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Cell Death Dis ; 7(11): e2461, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27831556

ABSTRACT

CD44, a large family of transmembrane glycoproteins, plays decisive roles in physiological and pathological conditions. CD44 isoforms are involved in several signaling pathways essential for life such as growth factor-induced signaling by EGF, HGF or VEGF. CD44 is also the main hyaluronan (HA) receptor and as such is involved in HA-dependent processes. To allow a genetic dissection of CD44 functions in homeostasis and disease, we generated a Cd44 floxed allele allowing tissue- and time-specific inactivation of all CD44 isoforms in vivo. As a proof of principle, we inactivated Cd44 in the skin epidermis using the K14Cre allele. Although the skin of such Cd44Δker mutants appeared morphologically normal, epidermal stiffness was reduced, wound healing delayed and TPA induced epidermal thickening decreased. These phenotypes might be caused by cell autonomous defects in differentiation and HA production as well as impaired adhesion and migration on HA by Cd44Δker keratinocytes. These findings support the usefulness of the conditional Cd44 allele in unraveling essential physiological and pathological functions of CD44 isoforms.


Subject(s)
Epidermis/metabolism , Gene Deletion , Hyaluronan Receptors/metabolism , Keratinocytes/metabolism , Stress, Mechanical , Animals , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Homeostasis/drug effects , Hyaluronic Acid/pharmacology , Keratinocytes/drug effects , Keratins/metabolism , Mice, Knockout , Organ Specificity/drug effects , Skin/metabolism , Wound Healing/drug effects
2.
Proc Natl Acad Sci U S A ; 107(8): 3406-11, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20160118

ABSTRACT

Interest in the development of new sources of transplantable materials for the treatment of injury or disease has led to the convergence of tissue engineering with stem cell technology. Bone and joint disorders are expected to benefit from this new technology because of the low self-regenerating capacity of bone matrix secreting cells. Herein, the differentiation of stem cells to bone cells using active multilayered capsules is presented. The capsules are composed of poly-L-glutamic acid and poly-L-lysine with active growth factors embedded into the multilayered film. The bone induction from these active capsules incubated with embryonic stem cells was demonstrated in vitro. Herein, we report the unique demonstration of a multilayered capsule-based delivery system for inducing bone formation in vivo. This strategy is an alternative approach for in vivo bone formation. Strategies using simple chemistry to control complex biological processes would be particularly powerful, as they make production of therapeutic materials simpler and more easily controlled.


Subject(s)
Embryonic Stem Cells/transplantation , Osteogenesis , Regeneration , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Capsules , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Mice , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Polyglutamic Acid/chemistry , Polylysine/chemistry , Tissue Engineering , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/pharmacology
3.
Cell Death Differ ; 13(4): 564-75, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16311515

ABSTRACT

Mouse embryonic stem (ES) cells remain pluripotent in vitro when grown in the presence of leukemia inhibitory factor (LIF). LIF starvation leads to apoptosis of some of the ES-derived differentiated cells, together with p38alpha mitogen-activated protein kinase (MAPK) activation. Apoptosis, but not morphological cell differentiation, is blocked by a p38 inhibitor, PD169316. To further understand the mechanism of action of this compound, we have identified its specific targets by microarray studies. We report on the global expression profiles of genes expressed at 3 days upon LIF withdrawal (d3) compared to pluripotent cells and of genes whose expression is modulated at d3 under anti-apoptotic conditions. We showed that at d3 without LIF cells express, earlier than anticipated, specialized cell markers and that when the apoptotic process was impaired, expression of differentiation markers was altered. In addition, functional tests revealed properties of anti-apoptotic proteins not to alter cell pluripotency and a novel role for metallothionein 1 gene, which prevents apoptosis of early differentiated cells.


Subject(s)
Apoptosis , Cell Differentiation , Stem Cells/cytology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line , Embryo, Mammalian/cytology , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Imidazoles/pharmacology , Interleukin-6/pharmacology , Leukemia Inhibitory Factor , Metallothionein/genetics , Metallothionein/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/enzymology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Stem Cells/drug effects , Stem Cells/enzymology , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Bioenerg Biomembr ; 35(1): 19-30, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12848338

ABSTRACT

Mice carrying a homozygous germ-line mutation in the nm23-M1 gene that eliminates its protein expression and drives expression of beta-galactosidase by nm23-M1 promoter have been generated. nm23-M1 gene inactivation is not teratogenic and the pups can grow to adult age without apparent health problems. However, they undergo a growth retardation and knocked out females cannot feed their pups. Both effects are background dependent. Beta-galactosidase mapping of nm23-M1 promoter activation during embryogenesis shows that the nm23-M1 gene is principally expressed in epithelial layer of tissues which require inductive epithelial-mesenchymal interactions for their formation. In conclusion, invalidated mice could be interesting models to analyze the role of nm23-M1 on signal transduction pathway regulation, or cancer induction and proliferation.


Subject(s)
Breast/metabolism , Fetal Growth Retardation/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Enzymologic/genetics , Models, Animal , Nucleoside-Diphosphate Kinase , Proteins/genetics , Proteins/metabolism , Animals , Animals, Newborn , Cloning, Molecular , Female , Fetal Growth Retardation/metabolism , Mice , Mice, Knockout/embryology , Mice, Knockout/growth & development , Mice, Knockout/metabolism , NM23 Nucleoside Diphosphate Kinases , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/genetics , Structure-Activity Relationship
5.
J Biol Chem ; 276(49): 46204-11, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11581263

ABSTRACT

Mouse embryonic stem (ES) cells remain "pluripotent" in vitro in the continuous presence of leukemia inhibitory factor (LIF). In the absence of LIF, ES cells are irreversibly committed to differentiate into various lineages. In this study we have set up an in vitro assay based on the anti-apoptotic activity of LIF to distinguish pluripotent from "differentiation-committed" ES cells. We have examined the phosphorylation profiles of known (STAT3 and ERKs) and identified new (ribosomal S6 kinases (RSKs) and cAMP-responsive element-binding protein (CREB)) LIF-regulated targets in ES and in ES-derived neuronal cells. We have demonstrated that although STAT3, a crucial player in the maintenance of ES cell pluripotency, is induced by LIF in all cell types tested, the LIF-dependent activation of RSKs is restricted to ES cells. We have shown that LIF-induced phosphorylation of RSKs in ES cells is dependent on ERKs, whereas STAT3 phosphorylation is not mediated by any known MAPK activities. Our results also demonstrate that the LIF-dependent phosphorylation of CREB is partially under the control of the RSK2 kinase.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Growth Inhibitors/metabolism , Interleukin-6 , Lymphokines/metabolism , Nuclear Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , CREB-Binding Protein , Cell Differentiation , Leukemia Inhibitory Factor , Mice , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/chemistry , Phosphorylation , STAT3 Transcription Factor , Stem Cells/cytology , Trans-Activators/chemistry
6.
EMBO J ; 20(15): 3967-74, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11483500

ABSTRACT

A group of specialized genes has been defined to govern the molecular mechanisms controlling the circadian clock in mammals. Their expression and the interactions among their products dictate circadian rhythmicity. Three genes homologous to Drosophila period exist in the mouse and are thought to be major players in the biological clock. Here we present the generation of mice in which the founding member of the family, Per1, has been inactivated by homologous recombination. These mice present rhythmicity in locomotor activity, but with a period almost 1 h shorter than wild-type littermates. Moreover, the expression of clock genes in peripheral tissues appears to be delayed in Per1 mutant animals. Importantly, light-induced phase shifting appears conserved. The oscillatory expression of clock genes and the induction of immediate-early genes in response to light in the master clock structure, the suprachiasmatic nucleus, are unaffected. Altogether, these data demonstrate that Per1 plays a distinct role within the Per family, as it may be involved predominantly in peripheral clocks and/or in the output pathways of the circadian clock.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Nuclear Proteins/physiology , Animals , Base Sequence , Biological Clocks/genetics , Cell Cycle Proteins , Circadian Rhythm/genetics , DNA, Complementary , Female , Gene Expression , Gene Targeting , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/genetics , Period Circadian Proteins , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger , Running
7.
Mol Cell ; 7(3): 509-15, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11463376

ABSTRACT

Metazoan genomes encode two related proteins, TBP and the TBP-like factor (TLF/TRF2), sharing a highly conserved saddle-like domain. TLF is highly expressed in a finely regulated pattern in the mouse testis during spermatogenesis. The murine TLF gene has been inactivated using homologous recombination. TLF-/- mice are viable, but mutant male mice are sterile due to a late, complete arrest of spermiogenesis. In mutant animals, spermatogonia and spermatocytes develop normally, but round spermatids undergo apoptosis at step 7. Although the expression of the transcriptional activator CREM and many other postmeiotic genes was unaltered in TLF null mice, several spermiogenesis genes transcribed in late round spermatids appeared to be under TLF control. Hence, TLF is not required for embryonic development in the mouse but is essential for spermiogenesis.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Gene Deletion , Spermatogenesis , Spermatozoa/cytology , Spermatozoa/metabolism , Transcription Factors/metabolism , Animals , Cell Extracts , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Nick-End Labeling , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Recombination, Genetic/genetics , Sequence Homology, Nucleic Acid , Spermatogenesis/genetics , Spermatogenesis/physiology , TATA Box Binding Protein-Like Proteins , Testis/cytology , Testis/embryology , Testis/metabolism , Transcription Factors/genetics
8.
J Biol Chem ; 276(6): 4261-9, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11053434

ABSTRACT

MLN64 is a transmembrane protein that shares homology with the cholesterol binding domain (START domain) of the steroidogenic acute regulatory protein. The steroidogenic acute regulatory protein is located in the inner membrane of mitochondria, where it facilitates cholesterol import into the mitochondria. Crystallographic analysis showed that the START domain of MLN64 is a cholesterol-binding domain. The present work was undertaken to determine which step of the intracellular cholesterol pathway MLN64 participates in. Using immunocytofluorescence, MLN64 colocalizes with LBPA, a lipid found specifically in late endosomes. Electron microscopy indicates that MLN64 is restricted to the limiting membrane of late endosomes. Microinjection or endocytosis of specific antibodies shows that the START domain of MLN64 is cytoplasmic. Deletion and mutagenesis experiments demonstrate that the amino-terminal part of MLN64 is responsible for its addressing. Although this domain does not contain conventional dileucine- or tyrosine-based targeting signals, we show that a dileucine motif (Leu(66)-Leu(67)) and a tyrosine residue (Tyr(89)) are critical for the targeting or the proper folding of the molecule. Finally, MLN64 colocalizes with cholesterol and Niemann Pick C1 protein in late endosomes. However, complementation assays show that MLN64 is not involved in the Niemann Pick C2 disease which, results in cholesterol lysosomal accumulation. Together, our results show that MLN64 plays a role at the surface of the late endosomes, where it might shuttle cholesterol from the limiting membrane to cytoplasmic acceptor(s).


Subject(s)
Cholesterol/metabolism , Endosomes/metabolism , Phosphoproteins/metabolism , Animals , Base Sequence , Biological Transport , Carrier Proteins/metabolism , Cell Line , Cricetinae , DNA Primers , Fluorescent Antibody Technique , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Mutagenesis, Site-Directed , Niemann-Pick C1 Protein , Phosphoproteins/genetics , Protein Binding , Tyrosine/metabolism
9.
Nature ; 408(6809): 199-203, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11089973

ABSTRACT

Signalling through dopamine D2 receptors governs physiological functions related to locomotion, hormone production and drug abuse. D2 receptors are also known targets of antipsychotic drugs that are used to treat neuropsychiatric disorders such as schizophrenia. By a mechanism of alternative splicing, the D2 receptor gene encodes two molecularly distinct isoforms, D2S and D2L, previously thought to have the same function. Here we show that these receptors have distinct functions in vivo; D2L acts mainly at postsynaptic sites and D2S serves presynaptic autoreceptor functions. The cataleptic effects of the widely used antipsychotic haloperidol are absent in D2L-deficient mice. This suggests that D2L is targeted by haloperidol, with implications for treatment of neuropsychiatric disorders. The absence of D2L reveals that D2S inhibits D1 receptor-mediated functions, uncovering a circuit of signalling interference between dopamine receptors.


Subject(s)
Protein Isoforms/physiology , Receptors, Dopamine D2/physiology , Animals , Apomorphine/pharmacology , Benzazepines/pharmacology , Catalepsy/metabolism , Chimera , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Protein Isoforms/chemistry , Quinpirole/pharmacology , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Signal Transduction , Spiperone/pharmacology , Synapses/physiology
10.
Development ; 127(19): 4277-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10976058

ABSTRACT

The functions of estrogen receptors (ERs) in mouse ovary and genital tracts were investigated by generating null mutants for ERalpha (ERalphaKO), ERbeta (ERbetaKO) and both ERs (ERalphabetaKO). All ERalphaKO females are sterile, whereas ERbetaKO females are either infertile or exhibit variable degrees of subfertility. Mast cells present in adult ERalphaKO and ERalphabetaKO ovaries could participate in the generation of hemorrhagic cysts. Folliculogenesis proceeds normally up to the large antral stage in both ERalphaKO and ERbetaKO adults, whereas large antral follicles of ERalpha+/-ERbetaKO and ERalphabetaKO adults are markedly deficient in granulosa cells. Similarly, prematurely developed follicles found in prepubertal ERalphaKO ovaries appear normal, but their ERalphabetaKO counterparts display only few granulosa cell layers. Upon superovulation treatment, all prepubertal ERalphaKO females form numerous preovulatory follicles of which the vast majority do not ovulate. The same treatment fails to elicit the formation of preovulatory follicles in half of the ERbetaKO mice and in all ERalpha+/-/ERbetaKO mice. These and other results reveal a functional redundancy between ERalpha and ERbeta for ovarian folliculogenesis, and strongly suggest that (1) ERbeta plays an important role in mediating the stimulatory effects of estrogens on granulosa cell proliferation, (2) ERalpha is not required for follicle growth under wild type conditions, while it is indispensable for ovulation, and (3) ERalpha is also necessary for interstitial glandular cell development. Our data also indicate that ERbeta exerts some function in ERalphaKO uterus and vagina. ERalphabetaKO granulosa cells localized within degenerating follicles transform into cells displaying junctions that are unique to testicular Sertoli cells. From the distribution pattern of anti-Müllerian hormone (AMH) in ERalphabetaKO ovaries, it is unlikely that an elevated AMH level is the cause of Sertoli cell differentiation. Our results also show that cell proliferation in the prostate and urinary bladder of old ERbetaKO and ERalphabetaKO males is apparently normal.


Subject(s)
Fertility/genetics , Genitalia, Female/anatomy & histology , Genitalia, Male/anatomy & histology , Glycoproteins , Receptors, Estrogen/genetics , Animals , Anti-Mullerian Hormone , Cysts , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Gonadotropins/pharmacology , Growth Inhibitors/isolation & purification , Infertility, Female/genetics , Intercellular Junctions/ultrastructure , Male , Mast Cells , Mice , Mice, Knockout , Ovarian Follicle/growth & development , Ovarian Follicle/ultrastructure , Ovary/drug effects , Ovary/ultrastructure , Ovulation , Phenotype , Sertoli Cells/ultrastructure , Sexual Maturation , Testicular Hormones/isolation & purification
11.
Proc Natl Acad Sci U S A ; 97(17): 9508-13, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944220

ABSTRACT

Several lines of evidence suggest that the serotonin (5-hydroxytryptamine, 5-HT) regulates cardiovascular functions during embryogenesis and adulthood. 5-HT binds to numerous cognate receptors to initiate its biological effects. However, none of the 5-HT receptor disruptions in mice have yet resulted in embryonic defects. Here we show that 5-HT(2B) receptor is an important regulator of cardiac development. We found that inactivation of 5-HT(2B) gene leads to embryonic and neonatal death caused by heart defects. 5-HT(2B) mutant embryos exhibit a lack of trabeculae in the heart and a specific reduction in the expression levels of a tyrosine kinase receptor, ErbB-2, leading to midgestation lethality. These in vivo data suggest that the Gq-coupled receptor 5-HT(2B) uses the signaling pathway of tyrosine kinase receptor ErbB-2 for cardiac differentiation. All surviving newborn mice display a severe ventricular hypoplasia caused by impaired proliferative capacity of myocytes. In adult mutant mice, cardiac histopathological changes including myocyte disarray and ventricular dilation were consistently observed. Our results constitute genetic evidence that 5-HT via 5-HT(2B) receptor regulates differentiation and proliferation of developing and adult heart. This mutation provides a genetic model for cardiopathy and should facilitate studies of both the pathogenesis and therapy of cardiac disorders in humans.


Subject(s)
Heart/embryology , Myocardium/metabolism , Receptors, Serotonin/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Division , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryo, Mammalian/physiopathology , Female , Fetal Death , Gene Deletion , Genes, erbB-2/genetics , Heart/physiopathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Kinetics , Male , Mice , Mice, Knockout , Myocardium/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, Serotonin, 5-HT2B , Receptors, Serotonin/genetics , Signal Transduction
12.
Nat Genet ; 25(2): 195-200, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835636

ABSTRACT

The role of the opioid system in controlling pain, reward and addiction is well established, but its role in regulating other emotional responses is poorly documented in pharmacology. The mu-, delta- and kappa- opioid receptors (encoded by Oprm, Oprd1 and Oprk1, respectively) mediate the biological activity of opioids. We have generated Oprd1-deficient mice and compared the behavioural responses of mice lacking Oprd1, Oprm (ref. 6) and Oprk1 (ref. 7) in several models of anxiety and depression. Our data show no detectable phenotype in Oprk1-/- mutants, suggesting that kappa-receptors do not have a role in this aspect of opioid function; opposing phenotypes in Oprm-/- and Oprd1-/- mutants which contrasts with the classical notion of similar activities of mu- and delta-receptors; and consistent anxiogenic- and depressive-like responses in Oprd1-/- mice, indicating that delta-receptor activity contributes to improvement of mood states. We conclude that the Oprd1-encoded receptor, which has been proposed to be a promising target for the clinical management of pain, should also be considered in the treatment of drug addiction and other mood-related disorders.


Subject(s)
Anxiety/metabolism , Depression/metabolism , Gene Deletion , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Anxiety/genetics , Binding Sites , Darkness , Depression/genetics , Electroshock , Female , Light , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Pain Threshold/drug effects , Phenotype , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Sex Characteristics , Swimming
13.
Development ; 127(13): 2955-63, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10851139

ABSTRACT

TIF1beta, a member of the transcriptional intermediary factor 1 family, has been reported to function as a corepressor for the large class of KRAB domain-containing zinc finger proteins of the Krüppel type. To address the biological function of TIF1beta, we have generated TIF1beta-deficient mice by gene disruption. TIF1beta protein was detected in wild-type but not TIF1beta(-/-) blastocysts. Homozygous mutant embryos, which developed normally until the blastocyst stage and underwent uterine implantation, were arrested in their development at the early egg-cylinder stage at about embryonic day (E) 5.5 and were completely resorbed by E8.5. Taken together, these results provide genetic evidence that TIF1beta is a developmental regulatory protein that exerts function(s) essential for early postimplantation development.


Subject(s)
DNA-Binding Proteins/physiology , Embryo, Mammalian/physiology , Fetal Proteins , Nuclear Proteins , Repressor Proteins/physiology , Transcription Factors , Alleles , Animals , Blastocyst/physiology , Blotting, Western , Crosses, Genetic , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Fluorescent Antibody Technique , Gastrula/physiology , Genotype , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis , Phenotype , Protein Structure, Tertiary , Repressor Proteins/genetics , Stem Cells , T-Box Domain Proteins/metabolism , Time Factors , Tripartite Motif-Containing Protein 28 , Zinc Fingers
14.
Hum Mol Genet ; 9(8): 1219-26, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10767347

ABSTRACT

Friedreich ataxia (FRDA), the most common autosomal recessive ataxia, is caused in almost all cases by homozygous intronic expansions resulting in the loss of frataxin, a mitochondrial protein conserved through evolution, and involved in mitochondrial iron homeostasis. Yeast knockout models, and histological and biochemical data from patient heart biopsies or autopsies indicate that the frataxin defect causes a specific iron-sulfur protein deficiency and mitochondrial iron accumulation leading to the pathological changes. Affected human tissues are rarely available to further examine this hypothesis. To study the mechanism of the disease, we generated a mouse model by deletion of exon 4 leading to inactivation of the Frda gene product. We show that homozygous deletions cause embryonic lethality a few days after implantation, demonstrating an important role for frataxin during early development. These results suggest that the milder phenotype in humans is due to residual frataxin expression associated with the expansion mutations. Surprisingly, in the frataxin knockout mouse, no iron accumulation was observed during embryonic resorption, suggesting that cell death could be due to a mechanism independent of iron accumulation.


Subject(s)
Fetal Death/genetics , Friedreich Ataxia/genetics , Iron-Binding Proteins , Iron/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Apoptosis , Decidua/cytology , Decidua/pathology , Embryo, Mammalian/pathology , Exons , Female , Genotype , Homozygote , Humans , Introns , Iron-Sulfur Proteins/deficiency , Iron-Sulfur Proteins/genetics , Mice , Mice, Knockout , Necrosis , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Pregnancy , Frataxin
15.
Hum Mol Genet ; 9(5): 849-58, 2000 Mar 22.
Article in English | MEDLINE | ID: mdl-10749994

ABSTRACT

Deletion of the murine survival of motor neuron gene (SMN) exon 7, the most frequent mutation found in spinal muscular atrophy (SMA) patients, directed to neurons but not to skeletal muscle, enabled generation of a mouse model of SMA providing evidence that motor neurons are the primary target of the gene defect. Moreover, the mutated SMN protein (SMNDeltaC15) is dramatically reduced in the motor neuron nuclei and causes a lack of gems associated with large aggregates of coilin, a coiled-body-specific protein. These results identify the lack of the nuclear targeting of SMN as the biochemical defect in SMA.


Subject(s)
Cell Nucleus/metabolism , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Animals , Base Sequence , Cyclic AMP Response Element-Binding Protein , DNA Primers , Disease Models, Animal , Exons , Gene Deletion , Genes, Lethal , Homozygote , Mice , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Nerve Tissue Proteins/chemistry , Phenotype , RNA-Binding Proteins , SMN Complex Proteins
16.
Proc Natl Acad Sci U S A ; 97(4): 1607-11, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677506

ABSTRACT

In the mammalian pancreas, the endocrine cell types of the islets of Langerhans, including the alpha-, beta-, delta-, and pancreatic polypeptide cells as well as the exocrine cells, derive from foregut endodermal progenitors. Recent genetic studies have identified a network of transcription factors, including Pdx1, Isl1, Pax4, Pax6, NeuroD, Nkx2.2, and Hlxb9, regulating the development of islet cells at different stages, but the molecular mechanisms controlling the specification of pancreatic endocrine precursors remain unknown. neurogenin3 (ngn3) is a member of a family of basic helix-loop-helix transcription factors that is involved in the determination of neural precursor cells in the neuroectoderm. ngn3 is expressed in discrete regions of the nervous system and in scattered cells in the embryonic pancreas. We show herein that ngn3-positive cells coexpress neither insulin nor glucagon, suggesting that ngn3 marks early precursors of pancreatic endocrine cells. Mice lacking ngn3 function fail to generate any pancreatic endocrine cells and die postnatally from diabetes. Expression of Isl1, Pax4, Pax6, and NeuroD is lost, and endocrine precursors are lacking in the mutant pancreatic epithelium. Thus, ngn3 is required for the specification of a common precursor for the four pancreatic endocrine cell types.


Subject(s)
Islets of Langerhans/embryology , Nerve Tissue Proteins/genetics , Pancreas/embryology , Transcription Factors/metabolism , Xenopus Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development , Eye Proteins , Glucagon/metabolism , Helix-Loop-Helix Motifs , Histocytochemistry , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , In Situ Hybridization , In Situ Nick-End Labeling , Insulin/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , RNA, Messenger/metabolism , Repressor Proteins , Somatostatin/metabolism , Trans-Activators/metabolism
17.
J Clin Invest ; 104(12): 1731-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606627

ABSTRACT

ADP is a key agonist in hemostasis and thrombosis. ADP-induced platelet activation involves the purinergic P2Y(1) receptor, which is responsible for shape change through intracellular calcium mobilization. This process also depends on an unidentified P2 receptor (P2cyc) that leads to adenylyl cyclase inhibition and promotes the completion and amplification of the platelet response. P2Y(1)-null mice were generated to define the role of the P2Y(1) receptor and to determine whether the unidentified P2cyc receptor is distinct from P2Y(1). These mice are viable with no apparent abnormalities affecting their development, survival, reproduction, or the morphology of their platelets, and the platelet count in these animals is identical to that of wild-type mice. However, platelets from P2Y(1)-deficient mice are unable to aggregate in response to usual concentrations of ADP and display impaired aggregation to other agonists, while high concentrations of ADP induce platelet aggregation without shape change. In addition, ADP-induced inhibition of adenylyl cyclase still occurs, demonstrating the existence of an ADP receptor distinct from P2Y(1). P2Y(1)-null mice have no spontaneous bleeding tendency but are resistant to thromboembolism induced by intravenous injection of ADP or collagen and adrenaline. Hence, the P2Y(1) receptor plays an essential role in thrombotic states and represents a potential target for antithrombotic drugs.


Subject(s)
Platelet Aggregation , Receptors, Purinergic P2/physiology , Thrombosis/prevention & control , Adenosine Diphosphate/pharmacology , Animals , Bleeding Time , Female , Male , Mice , Mice, Inbred C57BL , Platelet Activation , Receptors, Purinergic P2Y1 , Recombination, Genetic
18.
Mol Cell Biol ; 19(10): 7076-87, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490644

ABSTRACT

The ternary complex factors (TCFs) are targets for Ras/mitogen-activated protein kinase signalling pathways. They integrate the transcriptional response at the level of serum response elements in early-response genes, such as the c-fos proto-oncogene. An important aim is to understand the individual roles played by the three TCFs, Net, Elk1, and Sap1a. Net, in contrast to Elk1 and Sap1a, is a strong repressor of transcription. We now show that Net is regulated by nuclear-cytoplasmic shuttling in response to specific signalling pathways. Net is mainly nuclear under both normal and basal serum conditions. Net contains two nuclear localization signals (NLSs); one is located in the Ets domain, and the other corresponds to the D box. Net also has a nuclear export signal (NES) in the conserved Ets DNA binding domain. Net is apparently unique among Ets proteins in that a particular leucine in helix 1, a structural element, generates a NES. Anisomycin, UV, and heat shock induce active nuclear exclusion of Net through a pathway that involves c-Jun N-terminal kinase kinase and is inhibited by leptomycin B. Nuclear exclusion relieves transcriptional repression by Net. The specific induction of nuclear exclusion of Net by particular signalling pathways shows that nuclear-cytoplasmic transport of transcription factors can add to the specificity of the response to signalling cascades.


Subject(s)
Amino Acid Motifs , Cell Nucleus/metabolism , JNK Mitogen-Activated Protein Kinases , Oncogene Proteins , Repressor Proteins/metabolism , Transcription Factors/metabolism , Anisomycin/pharmacology , Biological Transport , Cell Compartmentation , Gene Expression Regulation , Hot Temperature , Leucine , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Signal Transduction , Transcription, Genetic , Ultraviolet Rays
19.
EMBO J ; 18(18): 4903-14, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10487743

ABSTRACT

The gene encoding cellular retinol (ROL, vitA)-binding protein type I (CRBPI) has been inactivated. Mutant mice fed a vitA-enriched diet are healthy and fertile. They do not present any of the congenital abnormalities related to retinoic acid (RA) deficiency, indicating that CRBPI is not indispensable for RA synthesis. However, CRBPI deficiency results in an approximately 50% reduction of retinyl ester (RE) accumulation in hepatic stellate cells. This reduction is due to a decreased synthesis and a 6-fold faster turnover, which are not related to changes in the levels of RE metabolizing enzymes, but probably reflect an impaired delivery of ROL to lecithin:retinol acyltransferase. CRBPI-null mice fed a vitA-deficient diet for 5 months fully exhaust their RE stores. Thus, CRBPI is indispensable for efficient RE synthesis and storage, and its absence results in a waste of ROL that is asymptomatic in vitA-sufficient animals, but leads to a severe syndrome of vitA deficiency in animals fed a vitA-deficient diet.


Subject(s)
Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/metabolism , Vitamin A/metabolism , Animals , Female , Homeostasis , In Situ Hybridization , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Retinol-Binding Proteins/deficiency , Retinol-Binding Proteins, Cellular , Vitamin A/administration & dosage , Vitamin A Deficiency/metabolism , Vitamin A Deficiency/pathology
20.
Proc Natl Acad Sci U S A ; 96(18): 10338-43, 1999 Aug 31.
Article in English | MEDLINE | ID: mdl-10468609

ABSTRACT

MHC class II (MHC-II) molecules play a central role in the selection of the T cell repertoire, in the establishment and regulation of the adaptive immune response, and in autoimmune deviation. We have generated knockout mice lacking all four of the classical murine MHC-II genes (MHCII(Delta/Delta) mice), via a large (80-kilobase) deletion of the entire class II region that was engineered by homologous recombination and Cre recombinase-mediated excision. These mice feature immune system perturbations like those of Aalpha and Abeta knockout animals, notably a dearth of CD4(+) lymphocytes in the thymus and spleen. No new anatomical or physiological abnormalities were observed in MHCII(Delta/Delta) mice. Because these animals are devoid of all classical MHC-II chains, even unpaired chains, they make excellent recipients for MHC-II transgenes from other species, avoiding the problem of interspecies cross-pairing of MHC-II chains. Therefore, they should be invaluable for engineering "humanized" mouse models of human MHC-II-associated autoimmune disorders.


Subject(s)
Genes, MHC Class II , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , T-Lymphocytes/immunology , Viral Proteins , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Bone Marrow Cells/immunology , Chromosome Deletion , Humans , Immunophenotyping , Integrases , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Stem Cells/immunology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...