Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Development ; 138(12): 2511-21, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21561988

ABSTRACT

RanBPM is a recently identified scaffold protein that links and modulates interactions between cell surface receptors and their intracellular signaling pathways. RanBPM has been shown to interact with a variety of functionally unrelated proteins; however, its function remains unclear. Here, we show that RanBPM is essential for normal gonad development as both male and female RanBPM(-/-) mice are sterile. In the mutant testis there was a marked decrease in spermatogonia proliferation during postnatal development. Strikingly, the first wave of spermatogenesis was totally compromised, as seminiferous tubules of homozygous mutant animals were devoid of post-meiotic germ cells. We determined that spermatogenesis was arrested around the late pachytene-diplotene stages of prophase I; surprisingly, without any obvious defect in chromosome synapsis. Interestingly, RanBPM deletion led to a remarkably quick disappearance of all germ cell types at around one month of age, suggesting that spermatogonia stem cells are also affected by the mutation. Moreover, in chimeric mice generated with RanBPM(-/-) embryonic stem cells all mutant germ cells disappeared by 3 weeks of age suggesting that RanBPM is acting in a cell-autonomous way in germ cells. RanBPM homozygous mutant females displayed a premature ovarian failure due to a depletion of the germ cell pool at the end of prophase I, as in males. Taken together, our results highlight a crucial role for RanBPM in mammalian gametogenesis in both genders.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Nuclear Proteins/physiology , Oogenesis , Spermatogenesis , Animals , Female , Gonads/growth & development , Male , Meiotic Prophase I , Mice , Primary Ovarian Insufficiency/etiology
2.
J Neurosci ; 32(7): 2252-62, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22396401

ABSTRACT

The molecular mechanisms underlying the effects of electroconvulsive shock (ECS) therapy, a fast-acting and very effective antidepressant therapy, are poorly understood. Changes related to neuroplasticity, including enhanced adult hippocampal neurogenesis and neuronal arborization, are believed to play an important role in mediating the effects of ECS. Here we show a dynamic upregulation of the scaffold protein tamalin, selectively in the hippocampus of animals subjected to ECS. Interestingly, this gene upregulation is functionally significant because tamalin deletion in mice abrogated ECS-induced neurogenesis in the adult mouse hippocampus. Furthermore, loss of tamalin blunts mossy fiber sprouting and dendritic arborization caused by ECS. These data suggest an essential role for tamalin in ECS-induced adult neuroplasticity and provide new insight into the pathways that are involved in mediating ECS effects.


Subject(s)
Carrier Proteins/physiology , Electroshock , Hippocampus/growth & development , Membrane Proteins/physiology , Neuronal Plasticity/physiology , Animals , Electroshock/methods , Embryonic Stem Cells/physiology , Female , Hippocampus/cytology , Hippocampus/physiology , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/deficiency , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Neurogenesis/physiology , Random Allocation
3.
Am J Physiol Cell Physiol ; 302(1): C141-53, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21865582

ABSTRACT

Neurotrophin-dependent activation of the tyrosine kinase receptor trkB.FL modulates neuromuscular synapse maintenance and function; however, it is unclear what role the alternative splice variant, truncated trkB (trkB.T1), may have in the peripheral neuromuscular axis. We examined this question in trkB.T1 null mice and demonstrate that in vivo neuromuscular performance and nerve-evoked muscle tension are significantly increased. In vitro assays indicated that the gain-in-function in trkB.T1(-/-) animals resulted specifically from an increased muscle contractility, and increased electrically evoked calcium release. In the trkB.T1 null muscle, we identified an increase in Akt activation in resting muscle as well as a significant increase in trkB.FL and Akt activation in response to contractile activity. On the basis of these findings, we conclude that the trkB signaling pathway might represent a novel target for intervention across diseases characterized by deficits in neuromuscular function.


Subject(s)
Muscle Contraction/genetics , Neuromuscular Junction/genetics , Receptor, trkB/deficiency , Receptor, trkB/genetics , Animals , Calcium/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Muscle Contraction/physiology , Neuromuscular Junction/physiology , Receptor, trkB/physiology
4.
Exp Neurol ; 337: 113576, 2021 03.
Article in English | MEDLINE | ID: mdl-33359475

ABSTRACT

The pathophysiology of Amyotrophic Lateral Sclerosis (ALS), a disease caused by the gradual degeneration of motoneurons, is still largely unknown. Insufficient neurotrophic support has been cited as one of the causes of motoneuron cell death. Neurotrophic factors such as BDNF have been evaluated in ALS human clinical trials, but yielded disappointing results attributed to the poor pharmacokinetics and pharmacodynamics of BDNF. In the inherited ALS G93A SOD1 animal model, deletion of the BDNF receptor TrkB.T1 delays spinal cord motoneuron cell death and muscle weakness through an unknown cellular mechanism. Here we show that TrkB.T1 is expressed ubiquitously in the spinal cord and its deletion does not change the SOD1 mutant spinal cord inflammatory state suggesting that TrkB.T1 does not influence microglia or astrocyte activation. Although TrkB.T1 knockout in astrocytes preserves muscle strength and co-ordination at early stages of disease, its specific conditional deletion in motoneurons or astrocytes does not delay motoneuron cell death during the early stage of the disease. These data suggest that TrkB.T1 may limit the neuroprotective BDNF signaling to motoneurons via a non-cell autonomous mechanism providing new understanding into the reasons for past clinical failures and insights into the design of future clinical trials employing TrkB agonists in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Membrane Glycoproteins/genetics , Protein-Tyrosine Kinases/genetics , Receptor, trkB/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/psychology , Animals , Calcium Signaling , Gene Deletion , Interleukin-1beta/metabolism , Macrophage Activation , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Motor Neurons/pathology , Psychomotor Performance , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Neuron ; 51(1): 21-8, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16815329

ABSTRACT

Imbalances in neurotrophins or their high-affinity Trk receptors have long been reported in neurodegenerative diseases. However, a molecular link between these gene products and neuronal cell death has not been established. In the trisomy 16 (Ts16) mouse there is increased apoptosis in the cortex, and hippocampal neurons undergo accelerated cell death that cannot be rescued by administration of brain-derived neurotrophic factor (BDNF). Ts16 neurons have normal levels of the TrkB tyrosine kinase receptor but an upregulation of the TrkB.T1 truncated receptor isoform. Here we show that restoration of the physiological level of the TrkB.T1 receptor by gene targeting rescues Ts16 cortical cell and hippocampal neuronal death. Moreover, it corrects resting Ca2+ levels and restores BDNF-induced intracellular signaling mediated by full-length TrkB in Ts16 hippocampal neurons. These data provide a direct link between neuronal cell death and abnormalities in Trk neurotrophin receptor levels.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Nerve Degeneration/genetics , Neurons/metabolism , Receptor, trkB/genetics , Trisomy/genetics , Animals , Apoptosis/genetics , Brain/cytology , Brain-Derived Neurotrophic Factor/pharmacology , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Disease Models, Animal , Down-Regulation/genetics , Gene Targeting/methods , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout , Mice, Neurologic Mutants , Molecular Weight , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recovery of Function/genetics , Signal Transduction/genetics , Trisomy/physiopathology
6.
J Neurosci ; 29(3): 678-85, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-19158294

ABSTRACT

Pathological or in vitro overexpression of the truncated TrkB (TrkB.T1) receptor inhibits signaling through the full-length TrkB (TrkB.FL) tyrosine kinase receptor. However, to date, the role of endogenous TrkB.T1 is still unknown. By studying mice lacking the truncated TrkB.T1 isoform but retaining normal spatiotemporal expression of TrkB.FL, we have analyzed TrkB.T1-specific physiological functions and its effect on endogenous TrkB kinase signaling in vivo. We found that TrkB.T1-deficient mice develop normally but show increased anxiety in association with morphological abnormalities in the length and complexity of neurites of neurons in the basolateral amygdala. However, no behavioral abnormalities were detected in hippocampal-dependent memory tasks, which correlated with lack of any obvious hippocampal morphological deficits or alterations in basal synaptic transmission and long-term potentiation. In vivo reduction of TrkB signaling by removal of one BDNF allele could be partially rescued by TrkB.T1 deletion, which was revealed by an amelioration of the enhanced aggression and weight gain associated with BDNF haploinsufficiency. Our results suggest that, at the physiological level, TrkB.T1 receptors are important regulators of TrkB.FL signaling in vivo. Moreover, TrkB.T1 selectively affects dendrite complexity of certain neuronal populations.


Subject(s)
Brain/abnormalities , Brain/anatomy & histology , Mutation , Neurons/physiology , Receptor, trkB/genetics , Animals , Body Weight/genetics , Brain/ultrastructure , Brain-Derived Neurotrophic Factor/genetics , Conditioning, Psychological/physiology , Exploratory Behavior/physiology , Fear , Hippocampus/cytology , In Vitro Techniques , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, trkB/deficiency , Silver Staining/methods
7.
Nat Commun ; 11(1): 1950, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327658

ABSTRACT

BDNF signaling in hypothalamic circuitries regulates mammalian food intake. However, whether BDNF exerts metabolic effects on peripheral organs is currently unknown. Here, we show that the BDNF receptor TrkB.T1 is expressed by pancreatic ß-cells where it regulates insulin release. Mice lacking TrkB.T1 show impaired glucose tolerance and insulin secretion. ß-cell BDNF-TrkB.T1 signaling triggers calcium release from intracellular stores, increasing glucose-induced insulin secretion. Additionally, BDNF is secreted by skeletal muscle and muscle-specific BDNF knockout phenocopies the ß-cell TrkB.T1 deletion metabolic impairments. The finding that BDNF is also secreted by differentiated human muscle cells and induces insulin secretion in human islets via TrkB.T1 identifies a new regulatory function of BDNF on metabolism that is independent of CNS activity. Our data suggest that muscle-derived BDNF may be a key factor mediating increased glucose metabolism in response to exercise, with implications for the treatment of diabetes and related metabolic diseases.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Calcium/metabolism , Cells, Cultured , Glucose/metabolism , Glucose Intolerance , Humans , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, trkB/chemistry , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction
8.
Mol Cell Biol ; 26(14): 5249-58, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809763

ABSTRACT

Trafficking and cell adhesion are key properties of cells of the immune system. However, the molecular pathways that control these cellular behaviors are still poorly understood. Cybr is a scaffold protein highly expressed in the hematopoietic/immune system whose physiological role is still unknown. In vitro studies have shown it regulates LFA-1, a crucial molecule in lymphocyte attachment and migration. Cybr also binds cytohesin-1, a guanine nucleotide exchange factor for the ARF GTPases, which affects actin cytoskeleton remodeling during cell migration. Here we show that expression of Cybr in vivo is differentially modulated by type 1 cytokines during lymphocyte maturation. In mice, Cybr deficiency negatively affects leukocytes circulating in blood and lymphocytes present in the lymph nodes. Moreover, in a Th1-polarized mouse model, lymphocyte trafficking is impaired by loss of Cybr, and Cybr-deficient mice with aseptic peritonitis have fewer cells than controls present in the peritoneal cavity, as well as fewer leukocytes leaving the bloodstream. Mutant mice injected with Moloney murine sarcoma/leukemia virus develop significantly larger tumors than wild-type mice and have reduced lymph node enlargement, suggesting reduced cytotoxic T-lymphocyte migration. Taken together, these data support a role for Cybr in leukocyte trafficking, especially in response to proinflammatory cytokines in stress conditions.


Subject(s)
Cytokines/physiology , Cytoskeletal Proteins/physiology , Leukocytes/physiology , Animals , Cell Differentiation , Cell Movement , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Gene Expression , Leukocytes/cytology , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Moloney murine sarcoma virus , Peritonitis/immunology , Peritonitis/pathology , Retroviridae Infections/immunology , Retroviridae Infections/pathology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology
9.
Elife ; 82019 08 20.
Article in English | MEDLINE | ID: mdl-31429825

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a potent modulator of brain synaptic plasticity. Signaling defects caused by dysregulation of its Ntrk2 (TrkB) kinase (TrkB.FL) and truncated receptors (TrkB.T1) have been linked to the pathophysiology of several neurological and neurodegenerative disorders. We found that upregulation of Rbfox1, an RNA binding protein associated with intellectual disability, epilepsy and autism, increases selectively hippocampal TrkB.T1 isoform expression. Physiologically, increased Rbfox1 impairs BDNF-dependent LTP which can be rescued by genetically restoring TrkB.T1 levels. RNA-seq analysis of hippocampi with upregulation of Rbfox1 in conjunction with the specific increase of TrkB.T1 isoform expression also shows that the genes affected by Rbfox1 gain of function are surprisingly different from those influenced by Rbfox1 deletion. These findings not only identify TrkB as a major target of Rbfox1 pathophysiology but also suggest that gain or loss of function of Rbfox1 regulate different genetic landscapes.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/physiology , Long-Term Potentiation , Membrane Glycoproteins/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , RNA Splicing Factors/biosynthesis , Up-Regulation , Animals , Gene Expression Profiling , Mice , Protein Isoforms/biosynthesis , Sequence Analysis, RNA
10.
J Cell Biol ; 210(6): 1003-12, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26347138

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is critical for mammalian development and plasticity of neuronal circuitries affecting memory, mood, anxiety, pain sensitivity, and energy homeostasis. Here we report a novel unexpected role of BDNF in regulating the cardiac contraction force independent of the nervous system innervation. This function is mediated by the truncated TrkB.T1 receptor expressed in cardiomyocytes. Loss of TrkB.T1 in these cells impairs calcium signaling and causes cardiomyopathy. TrkB.T1 is activated by BDNF produced by cardiomyocytes, suggesting an autocrine/paracrine loop. These findings unveil a novel signaling mechanism in the heart that is activated by BDNF and provide evidence for a global role of this neurotrophin in the homeostasis of the organism by signaling through different TrkB receptor isoforms.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cardiomyopathies/enzymology , Membrane Glycoproteins/metabolism , Muscle Strength , Myocardial Contraction , Myocytes, Cardiac/enzymology , Protein-Tyrosine Kinases/metabolism , Animals , Autocrine Communication , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Calcium Signaling , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Enzyme Activation , Genotype , Isolated Heart Preparation , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice, Knockout , Mice, Transgenic , Muscle Strength/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Paracrine Communication , Phenotype , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Signal Transduction , Time Factors
11.
Nat Med ; 19(6): 695-703, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644514

ABSTRACT

Anticancer chemotherapy drugs challenge hematopoietic tissues to regenerate but commonly produce long-term sequelae. Chemotherapy-induced deficits in hematopoietic stem or stromal cell function have been described, but the mechanisms mediating hematopoietic dysfunction remain unclear. Administration of multiple cycles of cisplatin chemotherapy causes substantial sensory neuropathy. Here we demonstrate that chemotherapy-induced nerve injury in the bone marrow of mice is a crucial lesion impairing hematopoietic regeneration. Using pharmacological and genetic models, we show that the selective loss of adrenergic innervation in the bone marrow alters its regeneration after genotoxic insult. Sympathetic nerves in the marrow promote the survival of constituents of the stem cell niche that initiate recovery. Neuroprotection by deletion of Trp53 in sympathetic neurons or neuroregeneration by administration of 4-methylcatechol or glial-derived neurotrophic factor (GDNF) promotes hematopoietic recovery. These results demonstrate the potential benefit of adrenergic nerve protection for shielding hematopoietic niches from injury.


Subject(s)
Antineoplastic Agents/toxicity , Bone Marrow/drug effects , Hematopoietic Stem Cells/drug effects , Regeneration/drug effects , Sympathetic Nervous System/drug effects , Animals , Bone Marrow/innervation , Cell Survival , Female , Hematopoietic Stem Cell Mobilization , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Receptors, Adrenergic, beta/physiology , Sympathetic Nervous System/physiology
12.
PLoS One ; 7(6): e39946, 2012.
Article in English | MEDLINE | ID: mdl-22761934

ABSTRACT

Brain Derived Neurotrophic Factor (BDNF) exerts strong pro-survival effects on developing and injured motoneurons. However, in clinical trials, BDNF has failed to benefit patients with amyotrophic lateral sclerosis (ALS). To date, the cause of this failure remains unclear. Motoneurons express the TrkB kinase receptor but also high levels of the truncated TrkB.T1 receptor isoform. Thus, we investigated whether the presence of this receptor may affect the response of diseased motoneurons to endogenous BDNF. We deleted TrkB.T1 in the hSOD1(G93A) ALS mouse model and evaluated the impact of this mutation on motoneuron death, muscle weakness and disease progression. We found that TrkB.T1 deletion significantly slowed the onset of motor neuron degeneration. Moreover, it delayed the development of muscle weakness by 33 days. Although the life span of the animals was not affected we observed an overall improvement in the neurological score at the late stage of the disease. To investigate the effectiveness of strategies aimed at bypassing the TrkB.T1 limit to BDNF signaling we treated SOD1 mutant mice with the adenosine A2A receptor agonist CGS21680, which can activate motoneuron TrkB receptor signaling independent of neurotrophins. We found that CGS21680 treatment slowed the onset of motor neuron degeneration and muscle weakness similarly to TrkB.T1 removal. Together, our data provide evidence that endogenous TrkB.T1 limits motoneuron responsiveness to BDNF in vivo and suggest that new strategies such as Trk receptor transactivation may be used for therapeutic intervention in ALS or other neurodegenerative disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Animals , Disease Models, Animal , Disease Progression , Mice , Phenethylamines/pharmacology , Receptor, trkB/genetics , Superoxide Dismutase/genetics
13.
Science ; 338(6112): 1357-60, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23224557

ABSTRACT

How neural circuits associated with sexually dimorphic organs are differentially assembled during development is unclear. Here, we report a sexually dimorphic pattern of mouse mammary gland sensory innervation and the mechanism of its formation. Brain-derived neurotrophic factor (BDNF), emanating from mammary mesenchyme and signaling through its receptor TrkB on sensory axons, is required for establishing mammary gland sensory innervation of both sexes at early developmental stages. Subsequently, in males, androgens promote mammary mesenchymal expression of a truncated form of TrkB, which prevents BDNF-TrkB signaling in sensory axons and leads to a rapid loss of mammary gland innervation independent of neuronal apoptosis. Thus, sex hormone regulation of a neurotrophic factor signal directs sexually dimorphic axonal growth and maintenance, resulting in generation of a sex-specific neural circuit.


Subject(s)
Axons/physiology , Brain-Derived Neurotrophic Factor/metabolism , Mammary Glands, Animal/embryology , Mammary Glands, Animal/innervation , Sex Characteristics , Androgens/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Male , Mice , Mice, Inbred C57BL , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction
14.
Development ; 131(20): 5185-95, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15459109

ABSTRACT

The nerve growth factor (NGF) receptor TrkA is widely expressed in non-neural tissues suggesting pleiotropic functions outside the nervous system. Based on pharmacological and immuno-depletion experiments, it has been hypothesized that NGF plays an important role in the normal development and function of the immune system. However, attempts to unravel these functions by conventional gene targeting in mice have been hampered by the early postnatal lethality caused by null mutations. We have developed a novel 'reverse conditional' gene targeting strategy by which TrkA function is restored specifically in the nervous system. Mice lacking TrkA in non-neuronal tissues are viable and appear grossly normal. All major immune system cell populations are present in normal numbers and distributions. However, mutant mice have elevated serum levels of certain immunoglobulin classes and accumulate B1 cells with aging. These data, confirmed in a classical reconstitution model using embryonic fetal liver from TrkA-null mice, demonstrate that endogenous NGF modulates B cell development through TrkA in vivo. Furthermore, they demonstrate that many of the dramatic effects previously reported by pharmacological or immuno-depletion approaches do not reflect physiological developmental roles of TrkA in the immune system.


Subject(s)
B-Lymphocytes/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , B-Lymphocytes/immunology , Immune System/embryology , Immunoglobulins/blood , Immunologic Memory/genetics , Immunologic Memory/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Nerve Growth Factor/deficiency , Nerve Growth Factor/genetics , Receptor, trkA/deficiency , Receptor, trkA/genetics , Receptor, trkA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL