Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 130(3): 373-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26025657

ABSTRACT

Diabetic neuropathy (DNP), afflicting sensory and motor nerve fibers, is a major complication in diabetes. The underlying cellular mechanisms of axon degeneration are poorly understood. IGFBP5, an inhibitory binding protein for insulin-like growth factor 1 (IGF1) is highly up-regulated in nerve biopsies of patients with DNP. We investigated the pathogenic relevance of this finding in transgenic mice overexpressing IGFBP5 in motor axons and sensory nerve fibers. These mice develop motor axonopathy and sensory deficits similar to those seen in DNP. Motor axon degeneration was also observed in mice in which the IGF1 receptor (IGF1R) was conditionally depleted in motoneurons, indicating that reduced activity of IGF1 on IGF1R in motoneurons is responsible for the observed effect. These data provide evidence that elevated expression of IGFBP5 in diabetic nerves reduces the availability of IGF1 for IGF1R on motor axons, thus leading to progressive neurodegeneration. Inhibition of IGFBP5 could thus offer novel treatment strategies for DNP.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/physiopathology , Motor Neurons/physiology , Nerve Degeneration/physiopathology , Animals , Axons/pathology , Cell Enlargement , Cell Survival/physiology , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/pathology , Humans , Mice, Transgenic , Motor Activity/physiology , Motor Neurons/pathology , Nerve Degeneration/pathology , Phrenic Nerve/pathology , Phrenic Nerve/physiopathology , Receptor, IGF Type 1/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Sensation/physiology
2.
Amyotroph Lateral Scler ; 13(5): 418-29, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22871074

ABSTRACT

Insulin-like growth factor I (IGF-I) has been successfully tested in the SOD1-G93A mouse model of familial amyotrophic lateral sclerosis (ALS) and proposed for clinical treatment. However, beneficial effects required gene therapy or intrathecal application. Circumventing the dosing issues we recently found that polyethylene glycol (PEG) modified IGF-I (PEG-IGF-I) modulated neuromuscular function after systemic application, and protected against disease progression in a motor neuron disease model. Here we investigated its effects in two SOD1-G93A mouse lines, the G1L with a milder and the G1H with a more severe phenotype. Results showed that in G1L mice, PEG-IGF-I treatment significantly improved muscle force, motor coordination and animal survival. In contrast, treatment of G1H mice with PEG-IGF-I or IGF-I even at high doses did not beneficially affect survival or functional outcomes despite increased signalling in brain and spinal cord by both agents. In conclusion, the data point towards further investigation of the therapeutic potential of PEG-IGF-I in ALS patients with less severe clinical phenotypes.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Insulin-Like Growth Factor I/therapeutic use , Motor Neurons/drug effects , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Disease Progression , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Severity of Illness Index
3.
Int J Oncol ; 40(4): 1079-88, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22200760

ABSTRACT

The microtubule associated tumor suppressor gene 1 (MTUS1) is a recently published tumor suppressor gene, which has also been shown to act as an early component in the growth inhibitory signaling cascade of the angiotensin II type 2 receptor (AT2R). In this study we report the generation of MTUS1 knock-out (KO) mice, which develop normally but reveal higher body weights and slightly decreased blood pressure levels. Twenty-eight percent of the studied MTUS1 KO mice also developed heart hypertrophy and 12% developed nephritis, independent of blood pressure levels. Forty-three percent of the MTUS1 KO mice revealed lymphoid hyperplasia affecting spleen (20%), kidney (37%), lung (23%), lymph nodes (17%), and liver (17%) accompanied with leukocytosis, lymphocytosis, and mild anemia. One animal (3%) developed a marginal zone B-cell lymphoma affecting submandibular salivary gland and regional lymph nodes. The symptoms of all mentioned animals are consistent with a B-cell lymphoproliferative disease with features of systemic lupus erythematosus. In addition, body weight of the MTUS1 KO mice was significantly increased and isolated skin fibroblasts showed increased cell proliferation and decreased cell size, compared to wild-type (WT) fibroblasts in response to depleted FCS concentration and lack of growth factors. In conclusion we herein report the first generation of a MTUS1 KO mouse, developing spontaneous heart hypertrophy and increased cell proliferation, confirming once more the anti-proliferative effect of MTUS1, and a SLE-like lymphoproliferative disease suggesting crucial role in regulation of inflammation. These MTUS1 KO mice can therefore serve as a model for further investigations in cardiovascular disease, autoimmune disease and carcinogenesis.


Subject(s)
Cardiomegaly/pathology , Lymphoma, B-Cell, Marginal Zone/pathology , Lymphoproliferative Disorders/pathology , Tumor Suppressor Proteins/deficiency , Animals , Cardiomegaly/blood , Cardiomegaly/genetics , Cardiomegaly/metabolism , Carrier Proteins/genetics , Cell Growth Processes/physiology , Fibroblasts/pathology , Immunohistochemistry , Lymphoma, B-Cell, Marginal Zone/blood , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoproliferative Disorders/blood , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Mice , Mice, Knockout , Skin/pathology , Tumor Suppressor Proteins/genetics
4.
Exp Neurol ; 232(2): 261-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21963648

ABSTRACT

Based on its potent neurotrophic and myotrophic activities, insulin-like growth factor I (IGF-I) has been proposed for treatment of neuromuscular disorders such as muscular dystrophies and amyotrophic lateral sclerosis (ALS). However, the short half life in the circulation limits its use in vivo. At least in mouse models, beneficial effects are generally only observed by dosing regimens such as minipumps or gene therapy that are difficult to translate to patients. We have developed a polyethylene glycol coupled IGF-I (PEG-IGF-I) that could circumvent these problems by longer half-life, showing all features of a therapeutic agent supporting muscular and neuronal function. Here we investigated its effects in the pmn mutant mouse, a model with typical dying-back motoneuron degeneration. In vitro, PEG-IGF-I and rhIGF-I profoundly promoted survival axonal growth of wild-type as well as pmn mutant embryonic motoneurons, suggesting that PEG-IGF-I had a fully conserved neurotrophic activity via its receptor. In vivo, treatment of pmn mutant mice with PEG-IGF-I prolonged survival, protected against late stage weight loss and significantly maintained muscle force and motor coordination. Consistently, PEG-IGF-I treatment rescued facial and lumbar motoneurons from cell death and partially preserved phrenic nerve myelinated axons. The data support that PEG-IGF-I could be used for treatment of neuromuscular diseases in a clinically feasible manner.


Subject(s)
Drug Design , Insulin-Like Growth Factor I/analogs & derivatives , Insulin-Like Growth Factor I/pharmacokinetics , Motor Neuron Disease/drug therapy , Motor Neurons/drug effects , Polyethylene Glycols/pharmacokinetics , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Female , Fetus/cytology , In Vitro Techniques , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Motor Neurons/cytology , Nerve Fibers, Myelinated/drug effects , Phrenic Nerve/cytology , Pregnancy , Recombinant Proteins/pharmacology , Spinal Cord/cytology
5.
Mol Cell Neurosci ; 42(2): 134-41, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19555761

ABSTRACT

Mutations in Ribosomal s6 kinase 2 (Rsk2) are associated with severe neuronal dysfunction in Coffin-Lowry syndrome (CLS) patients, flies and mice. So far, the mechanisms of how Rsk2 regulates development, maintenance and activity of neurons are not understood. We have investigated the consequences of Rsk2 deficiency in mouse spinal motoneurons. Survival of isolated Rsk2 deficient motoneurons is not reduced, but these cells grow significantly longer neurites. Conversely, overexpression of a constitutively active form of Rsk2 leads to reduced axon growth. Increased axon growth in Rsk2 deficient neurons was accompanied by higher Erk 1/2 phosphorylation, and the knockout phenotype could be rescued by pharmacological inhibition of MAPK/Erk kinase (Mek). These data indicate that Rsk2 negatively regulates axon elongation via the MAPK pathway. Thus, the functional defects observed in the nervous system of CLS patients and animal models with Rsk2 deficiency might be caused by dysregulated neurite growth rather than primary neurodegeneration.


Subject(s)
Axons/physiology , Motor Neurons , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cell Survival , Female , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/physiology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Spinal Cord/cytology
6.
J Neural Transm (Vienna) ; 115(8): 1127-32, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18665319

ABSTRACT

The relative contribution of the two tryptophan hydroxylase (TPH) isoforms, TPH1 and TPH2, to brain serotonergic system function is controversial. To investigate the respective role of TPH2 in neuron serotonin (5-HT) synthesis and the role of 5-HT in brain development, mice with a targeted disruption of Tph2 were generated. The preliminary results indicate that in Tph2 knockout mice raphe neurons are completely devoid of 5-HT, whereas no obvious alteration in morphology and fiber distribution are observed. The findings confirm the exclusive specificity of Tph2 in brain 5-HT synthesis and suggest that Tph2-synthesized 5-HT is not required for serotonergic neuron formation.


Subject(s)
Brain Chemistry/genetics , Brain/cytology , Neurons/physiology , Serotonin/deficiency , Tryptophan Hydroxylase/physiology , Animals , Gene Deletion , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Raphe Nuclei/cytology , Raphe Nuclei/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/biosynthesis , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Tryptophan Hydroxylase/genetics
7.
J Immunol ; 180(4): 2204-13, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250427

ABSTRACT

The neurotrophic cytokines ciliary neurotrophic factor and leukemia inhibitory factor (LIF) play a key role in neuronal and oligodendrocyte survival and as protective factors in neuroinflammation. To further elucidate the potential of endogenous LIF in modulating neuroinflammation, we studied myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis in LIF knockout mice (LIF(-/-) mice). In the late phase of active myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, LIF(-/-) mice exhibited a markedly milder disease course. The inflammatory infiltrate in LIF(-/-) mice was characterized by an increase in neutrophilic granulocytes early and fewer infiltrating macrophages associated with less demyelination later in the disease. In good correlation with an effect of endogenous LIF on the immune response, we found an Ag-specific T cell-priming defect with impaired IFN-gamma production in LIF(-/-) mice. On the molecular level, the altered recruitment of inflammatory cells is associated with distinct patterns of chemokine production in LIF(-/-) mice with an increase of CXCL1 early and a decrease of CCL2, CCL3, and CXCL10 later in the disease. These data reveal that endogenous LIF is an immunologically active molecule in neuroinflammation. This establishes a link between LIF and the immune system which was not observed in the ciliary neurotrophic factor knockout mouse.


Subject(s)
Chemokines/physiology , Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Leukemia Inhibitory Factor/deficiency , Leukemia Inhibitory Factor/physiology , Animals , Cell Line , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Genetic Predisposition to Disease , Glycoproteins/immunology , Glycoproteins/toxicity , Leukemia Inhibitory Factor/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Peptide Fragments/toxicity , Receptors, OSM-LIF/biosynthesis , Receptors, OSM-LIF/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
8.
Mol Cell Biol ; 27(24): 8797-806, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923692

ABSTRACT

Recent evidence has shown that the activation of receptor tyrosine kinases is not only dependent on binding of their ligands but in addition requires adhesion molecules as coreceptors. We have identified CD44v6 as a coreceptor for c-Met in several tumor and primary cells. The CD44v6 ectodomain is required for c-Met activation, whereas the cytoplasmic tail recruits ERM proteins and the cytoskeleton into a signalosome complex. Here we demonstrate that c-Met (and hepatocyte growth factor and Gab1) is haploinsufficient in a cd44-/- background, as the cd44-/-; met+/- (and cd44-/-; hgf+/- and cd44-/-; gab1+/-) mice die at birth. They have impaired synaptic transmission in the respiratory rhythm-generating network and alterations in the phrenic nerve. These results are the first genetic data showing that CD44 and c-Met collaborate in vivo and that they are involved in synaptogenesis and axon myelination in the central and peripheral nervous systems.


Subject(s)
Haploidy , Hyaluronan Receptors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Animals, Newborn , Brain/pathology , Hepatocyte Growth Factor/metabolism , Lung/abnormalities , Lung/pathology , Mice , Mice, Inbred C57BL , Motor Neurons/pathology , Nerve Fibers/pathology , Phrenic Nerve/pathology , Phrenic Nerve/ultrastructure , Synaptic Transmission
9.
Proc Natl Acad Sci U S A ; 104(43): 17210-5, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17940030

ABSTRACT

Neurotrophins are potent survival factors for developing and injured neurons. However, they are not being used to treat neurodegenerative diseases because of difficulties in administration and numerous side effects that have been encountered in previous clinical trials. Their biological activities use Trk (tropomyosin-related kinase) transmembrane tyrosine kinases. Therefore, one alternative approach is to use transactivation pathways such as adenosine 2A receptor agonists, which can activate Trk receptor signaling independent of neurotrophin binding. However, the relevance in vivo and applicability of these transactivation events during neurodegenerative and injury conditions have never been extensively studied. Here we demonstrate that motoneuron survival after facial nerve lesioning is significantly enhanced by transactivation of Trk receptor tyrosine kinases by adenosine agonists. Moreover, survival of motoneurons directly required the activation of the BDNF receptor TrkB and an increase in Akt (AKT8 virus oncogene cellular homolog) activity. The ability of small molecules to activate a trophic response by using Trk signaling provides a unique mechanism to promote survival signals in motoneurons and suggests new strategies for using transactivation in neurodegenerative diseases.


Subject(s)
Motor Neurons/cytology , Motor Neurons/enzymology , Receptor, trkB/genetics , Receptors, Adenosine A2/metabolism , Transcriptional Activation/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Animals , Axotomy , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/enzymology , Enzyme Activation/drug effects , Facial Nerve/drug effects , Facial Nerve/pathology , Mice , Motor Neurons/drug effects , Phenethylamines/pharmacology , Transcriptional Activation/drug effects
10.
Development ; 134(18): 3271-81, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17699610

ABSTRACT

The transcription factor Sox10 regulates early neural crest development, specification of neural crest-derived lineages and terminal differentiation of oligodendrocytes in the central nervous system. Here, we generated two novel hypomorphic Sox10 alleles in the mouse. Mutant mice either expressed a Sox10 protein with a triple alanine substitution in the dimerization domain, or a Sox10 protein with a deletion in the central portion that we define as a cell-specific transactivation domain. Phenotypic analysis revealed important roles for a functional dimerization domain and the newly defined novel transactivation domain in melanocyte and enteric nervous system development, whereas early neural crest development and oligodendrocyte differentiation were surprisingly little disturbed in both mutants. Unique requirements were additionally detected for the novel transactivation domain in satellite glia differentiation and during Schwann cell myelination, whereas DNA-dependent dimerization was needed for immature Schwann cells to enter the promyelinating stage. These two hypomorphic alleles thus uncover novel functions of Sox10 in satellite glia and Schwann cells during late developmental stages and reveal important developmental differences between these two types of peripheral glia and oligodendrocytes regarding their reliance on Sox10.


Subject(s)
Cell Lineage , High Mobility Group Proteins/physiology , Neuroglia/physiology , Peripheral Nervous System/embryology , Transcription Factors/physiology , Alleles , Animals , Cell Lineage/genetics , Dimerization , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Melanocytes/physiology , Mice , Mice, Mutant Strains , Mutation , Myelin Sheath , Neuroglia/cytology , Oligodendroglia/cytology , Oligodendroglia/physiology , SOXE Transcription Factors , Schwann Cells , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Hepatology ; 45(3): 639-48, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17326158

ABSTRACT

UNLABELLED: We previously reported that exogenous cardiotrophin-1 (CT-1), a member of the IL-6 family of cytokines, exerts hepatoprotective effects. Because CT-1 is expressed in the normal liver, we hypothesized that this cytokine may constitute an endogenous defense of the liver against proapoptotic stimuli. Here, we found that CT-1-/- mice died faster than wild-type animals after challenge with a lethal dose of the Fas agonist Jo-2. At sublethal doses of Jo-2, all wild-type mice survived whereas CT-1-/- animals developed extensive hepatocyte apoptosis with 50% mortality at 24 hours. Pretreatment with CT-1 improved survival and reduced injury in both CT-1-/- and wild-type animals. Upon Fas ligation the activation of STAT-3, a molecule that defends the liver against apoptosis, was lower in CT-1-/- mice than in wild-type animals despite similar IL-6 up-regulation in the 2 groups. Analysis of liver transcriptome in CT-1-/- and wild-type mice showed that 9 genes reported to be associated with cell survival/death functions were differentially expressed in the 2 groups. Four of these genes [IGFBP1, peroxiredoxin3, TNFR1, and calpastatin (endogenous inhibitor of calpain)] could be validated by real-time PCR. All of them were down-regulated in CT-1-/- mice and were modulated by CT-1 administration. Treatment of CT-1-/- animals with the calpain inhibitor MDL28170 afforded significant protection against Fas-induced liver injury. CONCLUSION: CT-1-/- mice are highly sensitive to Fas-mediated apoptosis due in part to deficient STAT-3 activation and inadequate control of calpain activity during the apoptotic process. Our data show that CT-1 is a natural defense of the liver against apoptosis. This cytokine may have therapeutic potential.


Subject(s)
Apoptosis/physiology , Cytokines/physiology , Hepatocytes/physiology , Liver/pathology , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Cytokines/genetics , Gene Expression Regulation , Hepatocytes/pathology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidases/genetics , Peroxidases/physiology , Peroxiredoxin III , Peroxiredoxins , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/physiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , fas Receptor/immunology
12.
J Neurochem ; 99(3): 900-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16925593

ABSTRACT

Transgenic mice expressing the superoxide dismutase G93A mutation (SOD1(G93A)) were used to investigate the role of glial inwardly rectifying K(+) (Kir)4.1 channels, which buffer extracellular K(+) increases in response to neuronal excitation. A progressive decrease in Kir4.1 immunoreactivity was observed predominantly in the ventral horn of SOD1(G93A) mutants. Immunoblotting of spinal cord extracts mirrored these changes by showing a loss of Kir4.1 channels from presymptomatic stages onwards. Kir4.1 channels were found to be expressed in the spinal cord grey matter, targetting astrocytes and clustering around capillaries, supporting their role in clearance of extracellular K(+). To understand the functional implications of extracellular K(+) increases, we challenged the NSC34 motor neurone cell line with increasing extracellular K(+) concentrations. Exposure to high extracellular K(+) induced progressive motor neurone cell death. We suggest that loss of Kir4.1 impairs perineural K(+) homeostasis and may contribute to motor neurone degeneration in SOD1(G93A) mutants by K(+) excitotoxic mechanisms.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Neuroglia/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Aquaporin 4/metabolism , Astrocytes/metabolism , Blotting, Western , Capillaries/cytology , Capillaries/metabolism , Cell Survival/physiology , Cells, Cultured , Extracellular Space/metabolism , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/physiology , Potassium/metabolism , Spinal Cord/cytology , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
13.
J Neurosci ; 26(6): 1823-32, 2006 Feb 08.
Article in English | MEDLINE | ID: mdl-16467531

ABSTRACT

Leukemia inhibitory factor (LIF) receptor beta (LIFRbeta) is a receptor for a variety of neurotrophic cytokines, including LIF, ciliary neurotrophic factor (CNTF), and cardiotrophin-1 (CT-1). These cytokines play an essential role for the survival and maintenance of developing and postnatal somatic motoneurons. CNTF may also serve the maintenance of autonomic, preganglionic sympathetic neurons (PSNs) in the spinal cord, as suggested by its capacity to prevent their death after destruction of one of their major targets, the adrenal medulla. Although somatic motoneurons and PSNs share a common embryonic origin, they are distinct in several respects, including responses to lesions. We have studied PSNs in mice with targeted deletions of the LIFRbeta or CT-1 genes, respectively. We show that LIF, CNTF, and CT-1 are synthesized in embryonic adrenal gland and spinal cord and that PSNs express LIFRbeta. In embryonic day 18.5 LIFRbeta (-/-) and CT-1 (-/-) mice, PSNs were reduced by approximately 20%. PSNs projecting to the adrenal medulla were more severely affected (-55%). Although LIFRbeta (-/-) mice revealed normal numbers of adrenal chromaffin cells and axons terminating on chromaffin cells, levels of adrenaline and numbers of adrenaline-synthesizing cells were significantly reduced. We conclude that activation of LIFRbeta is required for normal development of PSNs and one of their prominent targets, the adrenal medulla. Thus, both somatic motoneurons and PSNs in the spinal cord depend on LIFRbeta signaling for their development and maintenance, although PSNs seem to be overall less affected than somatic motoneurons by LIFRbeta deprivation.


Subject(s)
Adrenal Medulla/physiology , Cytokines/physiology , Interleukin-6/physiology , Nerve Fibers/physiology , Neurons/physiology , Actins/physiology , Adrenal Medulla/drug effects , Adrenal Medulla/innervation , Animals , Benzylamines/pharmacology , Cytokines/deficiency , Cytokines/genetics , Dizocilpine Maleate/pharmacology , Hippocampus/physiology , Interleukin-6/deficiency , Interleukin-6/genetics , Leukemia Inhibitory Factor , Mice , Nerve Fibers/drug effects , Neurons/drug effects , Oligopeptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Sympathetic Nervous System/embryology , Synapses/drug effects , Synapses/physiology , Synaptic Membranes/drug effects , Synaptic Membranes/physiology , Synaptosomes/drug effects , Synaptosomes/physiology
14.
Mol Cell Biol ; 25(20): 8960-70, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16199874

ABSTRACT

The Hey basic helix-loop-helix transcription factors are downstream effectors of Notch signaling in the cardiovascular system. Mice lacking Hey2 develop cardiac hypertrophy, often associated with congenital heart defects, whereas combined Hey1/Hey2 deficiency leads to severe vascular defects and embryonic lethality around embryonic day E9.5. The molecular basis of these disorders is poorly understood, however, since target genes of Hey transcription factors in the affected tissues remain elusive. To identify genes regulated by Hey factors we have generated a conditional Hey1 knockout mouse. This strain was used to generate paired Hey2- and Hey1/2-deficient embryonic stem cell lines. Comparison of these cell lines by microarray analysis identified GATA4 and GATA6 as differentially expressed genes. Loss of Hey1/2 leads to elevated GATA4/6 and ANF mRNA levels in embryoid bodies, while forced expression of Hey factors strongly represses expression of the GATA4 and GATA6 promoter in various cell lines. In addition, the promoter activity of the GATA4/6 target gene ANF was inhibited by Hey1, Hey2, and HeyL. Protein interaction and mutation analyses suggest that repression is due to direct binding of Hey proteins to GATA4 and GATA6, blocking their transcriptional activity. In Hey2-deficient fetal hearts we observed elevated mRNA levels of ANF and CARP. Expression of ANF and Hey2 is normally restricted to the trabecular and compact myocardial layer, respectively. Intriguingly, loss of Hey2 leads to ectopic ANF expression in the compact layer, suggesting a direct role for Hey2 in limiting ANF expression in this cardiac compartment.


Subject(s)
Atrial Natriuretic Factor/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Fetal Heart/metabolism , GATA4 Transcription Factor/genetics , Repressor Proteins/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , DNA, Complementary/genetics , Gene Expression Regulation , Gene Targeting , Helix-Loop-Helix Motifs/genetics , Histone Deacetylases/metabolism , Humans , Mice , Mice, Knockout , Muscle Proteins , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics
15.
Nat Neurosci ; 8(9): 1169-78, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116448

ABSTRACT

Bag1 is a cochaperone for the heat-shock protein Hsp70 that interacts with C-Raf, B-Raf, Akt, Bcl-2, steroid hormone receptors and other proteins. Here we use targeted gene disruption in mice to show that Bag1 has an essential role in the survival of differentiating neurons and hematopoietic cells. Cells of the fetal liver and developing nervous system in Bag1-/- mice underwent massive apoptosis. Lack of Bag1 did not disturb the primary function of Akt or Raf, as phosphorylation of the forkhead transcription factor FKHR and activation of extracellular signal-regulated kinase (Erk)-1/2 were not affected. However, the defect was associated with the disturbance of a tripartite complex formed by Akt, B-Raf and Bag1, in addition to the absence of Bad phosphorylation at Ser136. We also observed reduced expression of members of the inhibitor of apoptosis (IAP) family. Our data show that Bag1 is a physiological mediator of extracellular survival signals linked to the cellular mechanisms that prevent apoptosis in hematopoietic and neuronal progenitor cells.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/physiology , Membrane Proteins/physiology , Motor Neurons/physiology , Spinal Cord/cytology , Alkaline Phosphatase/metabolism , Analysis of Variance , Animals , Blotting, Southern/methods , Blotting, Western/methods , Carrier Proteins/metabolism , Cell Count/methods , Cell Survival/physiology , Cells, Cultured , DNA-Binding Proteins , Electron Transport Complex IV/metabolism , Embryo, Mammalian , Eye Proteins/metabolism , Fibroblasts/metabolism , HSP70 Heat-Shock Proteins/deficiency , Homeodomain Proteins/metabolism , Immunohistochemistry/methods , Immunoprecipitation/methods , In Situ Nick-End Labeling/methods , Intermediate Filament Proteins/metabolism , Isoenzymes/metabolism , Liver/cytology , Liver/growth & development , Liver/metabolism , Membrane Proteins/deficiency , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/metabolism , Nestin , Neurofilament Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , Propidium , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/biosynthesis , RNA, Small Interfering/metabolism , Rats , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord/growth & development , Spinal Cord/metabolism , Transcription Factors , Transfection/methods , bcl-Associated Death Protein
16.
J Neurosci ; 25(7): 1778-87, 2005 Feb 16.
Article in English | MEDLINE | ID: mdl-15716414

ABSTRACT

Members of the ciliary neurotrophic factor (CNTF)-leukemia inhibitory factor (LIF) gene family play an essential role for survival of developing and postnatal motoneurons. When subunits of the shared receptor complex are inactivated by homologous recombination, the mice die at approximately birth and exhibit reduced numbers of motoneurons in the spinal cord and brainstem nuclei. However, mice in which cntf, lif, or cardiotrophin-1 (ct-1) are inactivated can survive and show less motoneuron cell loss. This suggests cooperative and redundant roles of these ligands. However, their cooperative functions are not well understood. We generated cntf/lif/ct-1 triple-knock-out and combinations of double-knock-out mice to study the individual and combined roles of CNTF, LIF and CT-1 on postnatal motoneuron survival and function. Triple-knock-out mice exhibit increased motoneuron cell loss in the lumbar spinal cord that correlates with muscle weakness during early postnatal development. LIF deficiency leads to pronounced loss of distal axons and motor endplate alterations, whereas CNTF-and/or CT-1-deficient mice do not show significant changes in morphology of these structures. In cntf/lif/ct-1 triple-knock-out mice, various degrees of muscle fiber type grouping are found, indicating that denervation and reinnervation had occurred. We conclude from these findings that CNTF, LIF, and CT-1 have distinct functions for motoneuron survival and function and that LIF plays a more important role for postnatal maintenance of distal axons and motor endplates than CNTF or CT-1.


Subject(s)
Ciliary Neurotrophic Factor/physiology , Cytokines/physiology , Interleukin-6/physiology , Motor Neurons/physiology , Muscle Fibers, Skeletal/pathology , Muscle Weakness/physiopathology , Animals , Ciliary Neurotrophic Factor/deficiency , Ciliary Neurotrophic Factor/genetics , Cytokines/deficiency , Cytokines/genetics , Hand Strength , Interleukin-6/genetics , Leukemia Inhibitory Factor , Mice , Mice, Knockout , Motor Endplate/ultrastructure , Muscle Denervation , Muscle Weakness/genetics , Muscle Weakness/pathology , Nerve Degeneration , Peripheral Nerves/pathology , Spinal Cord/pathology
17.
J Cell Sci ; 117(Pt 24): 5803-14, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15507485

ABSTRACT

The Trk family of receptor tyrosine kinases and the p75 receptor (p75NTR) mediate the effects of neurotrophins on neuronal survival, differentiation and synaptic plasticity. The neurotrophin BDNF and its cognate receptor tyrosine kinase, TrkB.FL, are highly expressed in neurons of the central nervous system. At later stages in postnatal development the truncated TrkB splice variants (TrkB.T1, TrkB.T2) become abundant. However, the signalling and function of these truncated receptors remained largely elusive. We show that overexpression of TrkB.T1 in hippocampal neurons induces the formation of dendritic filopodia, which are known precursors of synaptic spines. The induction of filopodia by TrkB.T1 occurs independently of neurotrophin binding and of kinase activity of endogenous TrkB.FL. Coexpression of a p75NTR lacking an intracellular domain inhibits the TrkB.T1-induced effect in a dominant negative manner. Steric hindrance of extracellular p75NTR interactions with a specific antibody, or absence of p75NTR with an intact extracellular domain also inhibit this TrkB.T1-induced effect. We thus propose a novel signalling pathway initiated by neurotrophin-independent extracellular or intramembrane interaction of TrkB.T1 with the p75NTR receptor, which modulates dendritic growth via p75NTR signalling cascades.


Subject(s)
Dendrites/physiology , Hippocampus/metabolism , Pseudopodia/metabolism , Receptor, trkB/physiology , Receptors, Nerve Growth Factor/physiology , Animals , COS Cells , Cell Differentiation , Cloning, Molecular , Dendrites/metabolism , Dose-Response Relationship, Drug , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Microscopy, Fluorescence , Models, Biological , Nerve Growth Factors/metabolism , Neurons/metabolism , PC12 Cells , Protein Binding , Protein Structure, Tertiary , Rats , Receptor, Nerve Growth Factor , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Time Factors , Transfection
18.
Hum Mol Genet ; 13(18): 2031-42, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15269181

ABSTRACT

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is caused by recessive mutations of the IGHMBP2 gene. The role of IGHMBP2 (immunoglobulin mu-binding protein 2) in the pathomechanism of motor neuron disease is unknown. We have generated antibodies against Ighmbp2 and showed that low levels of Ighmbp2 immunoreactivity are present in the nucleus of spinal motor neurons and high levels in cell bodies, axons and growth cones. Ighmbp2 protein levels are strongly reduced in neuromuscular degeneration (nmd) mice, the mouse model of SMARD1. Mutant mice show severe motor neuron degeneration before first clinical symptoms become apparent. The loss of motor neuron cell bodies in lumbar spinal cord is followed by axonal degeneration in corresponding nerves such as the femoral quadriceps and sciatic nerve and loss of axon terminals at motor endplates. Motor neuron degeneration and clinical symptoms then slowly progress until the mice die at the age of 3-4 months. In addition, myopathic changes seem to contribute to muscle weakness and especially to respiratory failure, which is characteristic of the disorder in humans. Cultured motor neurons from embryonic nmd mice did not show any abnormality with respect to survival, axonal growth or growth cone size, thus differing from motor neurons derived from, e.g. Smn (survival motor neuron) deficient mice, the model of spinal muscular atrophy (SMA). Our data suggest that the pathomechanism in SMARD1 is clearly distinct from other motor neuron diseases such as classic SMA.


Subject(s)
DNA-Binding Proteins/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/pathology , Transcription Factors/metabolism , Action Potentials/physiology , Animals , Antibodies/immunology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Disease Models, Animal , Electromyography , Humans , Mice , Mice, Neurologic Mutants , Motor Neurons/chemistry , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Phenotype , Rotarod Performance Test , Spinal Cord/pathology , Transcription Factors/analysis , Transcription Factors/genetics
19.
Glia ; 42(4): 340-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12730954

ABSTRACT

The nodes of Ranvier are sites of specific interaction between Schwann cells and axons. Besides their crucial role in transmission of action potentials, the nodes of Ranvier and in particular the paranodal axon-Schwann cell networks (ASNs) are thought to function as local centers in large motor axons for removal, degradation, and disposal of organelles. In order to test whether ciliary neurotrophic factor (CNTF), which is present at high levels in the Schwann cell cytoplasm, is involved in the maintenance of these structures, we have examined lumbar ventral root nerve fibers of alpha-motor neurons by electron microscopy in 3- and 9-month-old Cntf null ((-/-)) mutant mice. Nerve fibers and nodes of Ranvier in 3-month-old Cntf(-/-) mutants appeared morphologically normal, except that ASNs were more voluminous in the mutants than in wild-type control animals at this age. In 9-month-old Cntf(-/-) animals, morphological changes, such as reduction in nerve fiber and axon diameter, myelin sheath disruption, and loss of ASNs at nodes of Ranvier, were observed. These findings suggest that endogenous CNTF, in addition to its role in promoting motor neuron survival and regeneration, is needed for long-term maintenance of alpha-motor nerve fibers. The premature loss of paranodal ASNs in animals lacking CNTF, which seems to be a defect related to a disturbed interaction in the nodal region between the axon and its myelinating Schwann cells, could impede the maintenance of a normal milieu in the motor axon, preceding more general neuronal damage.


Subject(s)
Axons/pathology , Ciliary Neurotrophic Factor/deficiency , Motor Neurons/pathology , Nerve Degeneration/pathology , Ranvier's Nodes/pathology , Animals , Axons/ultrastructure , Ciliary Neurotrophic Factor/genetics , Male , Mice , Mice, Knockout , Microscopy, Electron , Motor Neurons/ultrastructure , Ranvier's Nodes/ultrastructure , Schwann Cells/pathology , Schwann Cells/ultrastructure , Spinal Nerve Roots/pathology
20.
Biochem J ; 370(Pt 2): 397-402, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12521380

ABSTRACT

Selenoprotein P (SePP), the major selenoprotein in plasma, has been implicated in selenium transport, selenium detoxification or antioxidant defence. We generated SePP-knockout mice that were viable, but exhibited reduced growth and developed ataxia. Selenium content was elevated in liver, but low in plasma and other tissues, and selenoenzyme activities changed accordingly. Our data reveal that SePP plays a pivotal role in delivering hepatic selenium to target tissues.


Subject(s)
Proteins/genetics , Proteins/metabolism , Selenium/metabolism , Animals , Biological Transport/genetics , Gene Deletion , Mice , Mice, Transgenic , Selenoprotein P , Selenoproteins
SELECTION OF CITATIONS
SEARCH DETAIL
...