Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Am Soc Nephrol ; 27(3): 766-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26376860

ABSTRACT

Hyponatremia is the most common clinical electrolyte disorder. Once thought to be asymptomatic in response to adaptation by the brain, recent evidence suggests that chronic hyponatremia may be linked to attention deficits, gait disturbances, risk of falls, and cognitive impairments. Such neurologic defects are associated with a reduction in quality of life and may be a significant cause of mortality. However, because underlying diseases such as adrenal insufficiency, heart failure, liver cirrhosis, and cancer may also affect brain function, the contribution of hyponatremia alone to neurologic manifestations and the underlying mechanisms remain unclear. Using a syndrome of inappropriate secretion of antidiuretic hormone rat model, we show here that sustained reduction of serum sodium ion concentration induced gait disturbances; facilitated the extinction of a contextual fear memory; caused cognitive impairment in a novel object recognition test; and impaired long-term potentiation at hippocampal CA3-CA1 synapses. In vivo microdialysis revealed an elevated extracellular glutamate concentration in the hippocampus of chronically hyponatremic rats. A sustained low extracellular sodium ion concentration also decreased glutamate uptake by primary astrocyte cultures, suggesting an underlying mechanism of impaired long-term potentiation. Furthermore, gait and memory performances of corrected hyponatremic rats were equivalent to those of control rats. Thus, these results suggest chronic hyponatremia in humans may cause gait disturbance and cognitive impairment, but these abnormalities are reversible and careful correction of this condition may improve quality of life and reduce mortality.


Subject(s)
Gait Disorders, Neurologic/etiology , Hyponatremia/complications , Inappropriate ADH Syndrome/physiopathology , Memory Disorders/etiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/physiopathology , Cells, Cultured , Chronic Disease , Cognition Disorders/blood , Cognition Disorders/etiology , Disease Models, Animal , Fear/physiology , Gait Disorders, Neurologic/blood , Glutamic Acid/metabolism , Hyponatremia/blood , Hyponatremia/psychology , Inappropriate ADH Syndrome/complications , Inappropriate ADH Syndrome/psychology , Male , Memory Disorders/blood , Microdialysis , Neuronal Plasticity , Rats , Rats, Sprague-Dawley , Sodium/blood , Sodium/pharmacology , Synapses/physiology
2.
J Neurosci ; 31(16): 6067-78, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21508232

ABSTRACT

Mutations of the myosin Va gene cause the neurological diseases Griscelli syndrome type 1 and Elejalde syndrome in humans and dilute phenotypes in rodents. To understand the pathophysiological mechanisms underlying the neurological disorders in myosin Va diseases, we conducted an integrated analysis at the molecular, cellular, electrophysiological, and behavioral levels using the dilute-neurological (d-n) mouse mutant. These mice manifest an ataxic gait and clonic seizures during postnatal development, but the neurological disorders are ameliorated in adulthood. We found that smooth endoplasmic reticulum (SER) rarely extended into the dendritic spines of Purkinje cells (PCs) of young d-n mice, and there were few, if any, IP(3) receptors. Moreover, long-term depression (LTD) at parallel fiber-PC synapses was abolished, consistent with our previous observations in juvenile lethal dilute mutants. Young d-n mice exhibited severe impairment of cerebellum-dependent motor learning. In contrast, adult d-n mice showed restoration of motor learning and LTD, and these neurological changes were associated with accumulation of SER and IP(3) receptors in some PC spines and the expression of myosin Va proteins in the PCs. RNA interference-mediated repression of myosin Va caused a reduction in the number of IP(3) receptor-positive spines in cultured PCs. These findings indicate that myosin Va function is critical for subsequent processes in localization of SER and IP(3) receptors in PC spines, LTD, and motor learning. Interestingly, d-n mice had defects of motor coordination from young to adult ages, suggesting that the role of myosin Va in PC spines is not sufficient for motor coordination.


Subject(s)
Cerebellum/physiology , Learning/physiology , Motor Activity/physiology , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Analysis of Variance , Animals , Blotting, Western , Conditioning, Classical/physiology , Conditioning, Eyelid/physiology , Dendritic Spines/metabolism , Electrophysiology , Endoplasmic Reticulum, Smooth/metabolism , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Neurologic Mutants , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Rotarod Performance Test , Synapses/physiology
3.
Gene ; 411(1-2): 94-102, 2008 Mar 31.
Article in English | MEDLINE | ID: mdl-18280673

ABSTRACT

Using forward genetic analysis, we identified the insertion of an intracisternal A particle (IAP) retrotransposon element in the plasma membrane calcium ATPase 2 gene (Pmca2/Atp2b2) in the joggle mouse, a novel mutant that displays ataxic gait by postnatal day 12. Expression of Pmca2 mRNA in the joggle mouse is only 5% of that in the wild type mouse. The insertion is located 15 bp downstream of the donor splice site of the exon containing the initiation codon. Chimeric mRNA composed of the 5'-region of Pmca2 and the IAP element were detected, indicating that some of the primary transcripts are terminated by polyadenylation signals in long terminal repeats of the IAP element. We also identified cryptic splice sites in the IAP element that are likely involved in aberrant splicing of the Pmca2 primary transcripts that leads to rapid degradation of mRNA through nonsense mediated mRNA decay. Ataxia was observed in compound heterozygous mice carrying the joggle mutation and the wriggle mutation, a previously reported missense Pmca2 mutant. Thus, we attributed ataxia in joggle mice to reduced expression of Pmca2, resulting from insertion of the IAP element.


Subject(s)
Ataxia/genetics , Brain/metabolism , Genes, Intracisternal A-Particle/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Retroelements/genetics , Animals , Chromosome Mapping , Gene Expression , Mice , Mice, Mutant Strains , Mutagenesis, Insertional , Phenotype , Plasma Membrane Calcium-Transporting ATPases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
Exp Anim ; 56(5): 389-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18075201

ABSTRACT

The joggle mouse is a recessive ataxic mutant carrying an unknown mutation in a C3H/He (C3H)-derived chromosomal segment. Taking advantage of the mouse genome database, we selected 127 DNA microsatellite markers showing heterozygosity between C3H and C57BL/6J (B6) and a first round of screening for the joggle mutation was performed on B6-jog/+ partial congenic mice (N4). We identified 4 chromosomal regions in which 13 microsatellite markers show heterozygosity between C3H and B6. Then, we analyzed the genotype of these 4 chromosomal regions in mice that showed the joggle phenotype and mapped the jog locus between markers D6Mit104 (111.4 Mb) and D6Mit336 (125.1 Mb) (an interval of 13.7 Mb) on chromosome 6. By using a partial congenic strain together with the mouse genome database, we successfully mapped the chromosomal localization of the jog locus much more efficiently than by conventional linkage analysis.


Subject(s)
Chromosome Mapping/methods , Mice, Mutant Strains/genetics , Animals , Heterozygote , Mice , Mice, Inbred C3H/genetics , Microsatellite Repeats
5.
J Invest Dermatol ; 126(3): 638-47, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16410790

ABSTRACT

Desmosterolosis is an autosomal recessive disorder due to mutations in the 3beta-hydroxysterol-Delta24 reductase (DHCR24) gene that encodes an enzyme catalyzing the conversion of desmosterol to cholesterol. To date, only two patients have been reported with severe developmental defects including craniofacial abnormalities and limb malformations. We employed mice with targeted disruption of DHCR24 to understand the pathophysiology of desmosterolosis. All DHCR24-/- mice died within a few hours after birth. Their skin was wrinkleless and less pliant, leading to restricted movement and inability to suck (empty stomach). DHCR24 gene was expressed abundantly in the epidermis of control but not of DHCR24-/- mice. Accordingly, cholesterol was not detected whereas desmosterol was abundant in the epidermis of DHCR24-/- mice. Skin histology revealed thickened epidermis with few and smaller keratohyaline granules. Aberrant expression of keratins such as keratins 6 and 14 suggested hyperproliferative hyperkeratosis with undifferentiated keratinocytes throughout the epidermis. Altered expression of filaggrin, loricrin, and involcrin were also observed in the epidermis of DHCR24-/-. These findings suggested impaired skin barrier function. Indeed, increased trans-epidermal water loss and permeability of Lucifer yellow were observed in DHCR24-/- mice. DHCR24 thus plays crucial role for skin development and its proper function.


Subject(s)
Desmosterol/metabolism , Nerve Tissue Proteins/physiology , Oxidoreductases Acting on CH-CH Group Donors/physiology , Skin Diseases/etiology , Skin/pathology , Animals , Apoptosis , Caveolin 1/analysis , Cell Differentiation , Cell Proliferation , Ceramides/analysis , Cholesterol/biosynthesis , Fatty Acids, Nonesterified/analysis , Immunohistochemistry , Keratinocytes/cytology , Keratins/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Permeability , Skin/metabolism , Skin Diseases/pathology
6.
Ann N Y Acad Sci ; 1086: 66-80, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17185506

ABSTRACT

A spontaneous neurological mutation, dilute-opisthotonus (dop), was discovered in our breeding colony of Wistar rats. We found that the mutation affected the gene encoding Myosin Va (MyoVA), an actin-based molecular motor. Analysis of the myosin Va (Myo5a) gene of the dop genome showed the presence of a complex rearrangement consisting of a 306-bp inversion associated with 217-bp and 17-bp deletions. A 141-bp exon is skipped in the dop transcript, producing a dop cDNA with a 141 in-frame deletion in the sequences encoding the head region. Expression of the MyoVA protein is severely impaired in the brains of dop homozygous rats, suggesting they have a null mutation for Myo5a. In a morphological analysis of the cerebella of dop rats, we found an absence of smooth endoplasmic reticulum (SER) and of inositol 1,4,5-triphosphate (IP3) receptors in the dendritic spines of Purkinje cells (PC). The SER acts as an intracellular Ca(2+) store and IP3-mediated Ca(2+) signaling in dendritic spines plays a critical role in synaptic regulation. We therefore measured synaptic transmission and long-term depression (LTD), a form of synaptic plasticity underlying cerebellar motor learning, at PC synapses in the cerebella of dop rats. We found that synaptic transmission at the PC synapses is largely normal, whereas the LTD is deficient due to a decrease in IP3-mediated Ca(2+) release from the SER in the PC spines of the dop cerebella. These findings may account for the ataxic movements and clonic convulsions displayed by dop rats. They also contribute to our understanding of the neurological disease mechanisms of the human hereditary disease Griscelli syndrome type 1, which is caused by mutation of the Myo5a gene.


Subject(s)
Disease Models, Animal , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myosin Heavy Chains/physiology , Myosin Type V/physiology , Nervous System Diseases/pathology , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Brain/pathology , Dendrites/metabolism , Dendrites/pathology , Endoplasmic Reticulum, Smooth/metabolism , Endoplasmic Reticulum, Smooth/pathology , Molecular Sequence Data , Mutation , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Purkinje Cells/metabolism , Purkinje Cells/pathology , Rats , Rats, Wistar , Synaptic Transmission , Syndrome
7.
Exp Anim ; 55(4): 411-4, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16880690

ABSTRACT

While establishing a new mouse strain, we discovered a novel mutant mouse that exhibited ataxia. Mating experiments showed that the mutant phenotype was due to a single autosomal recessive gene, which we have termed joggle (gene symbol: jog). The ataxia becomes apparent around postnatal day 12, when the mice first attempt to walk, and worsens thereafter. The life span of the mutant mouse is comparable to that of the wild-type mouse. After 21 days of age, the cerebellum weights of the jog/jog mice are significantly lower than those of the wild-type mice. These observations indicate that jog/jog mutant mice could be useful models for biomedical research.


Subject(s)
Ataxia/genetics , Mice, Mutant Strains , Animals , Ataxia/pathology , Cerebellum/pathology , Disease Models, Animal , Female , Genes, Recessive , Male , Mice , Phenotype
8.
Neurobiol Aging ; 43: 34-46, 2016 07.
Article in English | MEDLINE | ID: mdl-27255813

ABSTRACT

Emerging evidence suggests that axonal degeneration is a disease mechanism in various neurodegenerative diseases and that the paranodes at the nodes of Ranvier may be the initial site of pathogenesis. We investigated the pathophysiology of the disease process in the central and peripheral nervous systems of a Caspr mutant mouse, shambling (shm), which is affected by disrupted paranodal structures and impaired nerve conduction of myelinated nerves. The shm mice manifest a progressive neurological phenotype as mice age. We found extensive axonal degeneration and a loss of neurons in the central nervous system and peripheral nervous system in aged shm mice. Axonal alteration of myelinated nerves was defined by abnormal distribution and expression of neurofilaments and derangements in the status of phosphorylated and non/de-phosphorylated neurofilaments. Autophagy-related structures were also accumulated in degenerated axons and neurons. In conclusion, our results suggest that disrupted axon-glia interactions at the paranode cause the cytoskeletal alteration in myelinated axons leading to neuronal cell death, and the process involves detrimental autophagy and aging as factors that promote the pathogenesis.


Subject(s)
Aging/pathology , Axons/pathology , Axons/physiology , Nerve Degeneration , Nerve Fibers, Myelinated/pathology , Neuroglia/pathology , Neuroglia/physiology , Neurons/pathology , Action Potentials , Animals , Cell Death , In Vitro Techniques , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/physiology , Sciatic Nerve/pathology , Sciatic Nerve/physiology , Sciatic Nerve/physiopathology
9.
J Neurosci ; 24(50): 11368-80, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15601943

ABSTRACT

The functions of synapsins were examined by characterizing the phenotype of mice in which all three synapsin genes were knocked out. Although these triple knock-out mice were viable and had normal brain anatomy, they exhibited a number of behavioral defects. Synaptic transmission was altered in cultured neurons from the hippocampus of knock-out mice. At excitatory synapses, loss of synapsins did not affect basal transmission evoked by single stimuli but caused a threefold increase in the rate of synaptic depression during trains of stimuli. This suggests that synapsins regulate the reserve pool of synaptic vesicles. This possibility was examined further by measuring synaptic vesicle density in living neurons transfected with green fluorescent protein-tagged synaptobrevin 2, a marker of synaptic vesicles. The relative amount of fluorescent synaptobrevin was substantially lower at synapses of knock-out neurons than of wild-type neurons. Electron microscopy also revealed a parallel reduction in the number of vesicles in the reserve pool of vesicles >150 nm away from the active zone at excitatory synapses. Thus, synapsins are required for maintaining vesicles in the reserve pool at excitatory synapses. In contrast, basal transmission at inhibitory synapses was reduced by loss of synapsins, but the kinetics of synaptic depression were unaffected. In these terminals, there was a mild reduction in the total number of synaptic vesicles, but this was not restricted to the reserve pool of vesicles. Thus, synapsins maintain the reserve pool of glutamatergic vesicles but regulate the size of the readily releasable pool of GABAergic vesicles.


Subject(s)
Brain/physiology , Synapses/physiology , Synapsins/physiology , Action Potentials/physiology , Animals , Brain/anatomy & histology , Brain/ultrastructure , Evoked Potentials/physiology , Hippocampus/physiology , Learning/physiology , Mice , Mice, Knockout , Multigene Family , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Neurotransmitter Agents/metabolism , Phenotype , Synapses/classification , Synapsins/genetics , Synaptic Transmission/physiology , Synaptic Vesicles/physiology
10.
Cardiovasc Res ; 53(1): 89-96, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11744016

ABSTRACT

OBJECTIVE: Intercellular coupling through gap junctions allows the morphologically and functionally heterogeneous sinoatrial node to synchronize and drive the atrial muscle. The purpose of this study was to identify the connexin isotypes expressed by sinoatrial node cells and to analyse the density of connexins in relation to cell size. METHODS: Labeling for the different connexins using isotype-specific antibodies was assessed in cells isolated from the rabbit sinoatrial node by immunoconfocal microscopy. RESULTS: Sinoatrial node cells with a cell projection area smaller than 800 microm(2) were devoid of immunolabeling for connexin43. Such small cells showed high levels of connexin45 labeling (compared to that in large cells) and low levels of connexin40 labeling. Sinoatrial node cells with a projection area between 800 and 1200 microm(2) had a lower amount of connexin45 label and again a small amount of connexin40 but an increased amount of connexin43 label. In the larger sinoatrial node cells, some colocalization of connexin45 and connexin43 immunolabeled spots was observed. CONCLUSIONS: Rabbit sinoatrial node cells are heterogeneous in terms of connexin expression, and there is a clear cell size-dependence in pattern of connexin expression. Small (putative central) cells express connexin45 but not connexin43, whereas larger (putative peripheral) cells express both connexin45 and connexin43. The co-localization of connexin43 and connexin45 in larger cells raises the possibility that heterotypic or heteromeric connexin43/connexin45 channels could be present in gap junctions at the periphery of the sinoatrial node.


Subject(s)
Connexins/analysis , Sinoatrial Node/metabolism , Animals , Cell Size , Cells, Cultured , Connexin 43/analysis , Immunohistochemistry/methods , Microscopy, Confocal , Protein Isoforms/metabolism , Rabbits , Gap Junction alpha-5 Protein
11.
PLoS One ; 10(7): e0133812, 2015.
Article in English | MEDLINE | ID: mdl-26192435

ABSTRACT

Animal models with defective glucagon action show hyperplasia of islet α-cells, however, the regulatory mechanisms underlying the proliferation of islet endocrine cells remain largely to be elucidated. The Gcggfp/gfp mice, which are homozygous for glucagon/green fluorescent protein knock-in allele (GCGKO), lack all proglucagon-derived peptides including glucagon and GLP-1. The present study was aimed to characterize pancreatic neuroendocrine tumors (panNETs), which develop in the GCGKO mice. At 15 months of age, macroscopic GFP-positive tumors were identified in the pancreas of all the GCGKO mice, but not in that of the control heterozygous mice. The tumor manifested several features that were consistent with pancreatic neuroendocrine tumors (panNETs), such as organoid structures with trabecular and cribriform patterns, and the expression of chromogranin A and synaptophysin. Dissemination of GFP-positive cells was observed in the liver and lungs in 100% and 95%, respectively, of 15-month-old GCGKO mice. To elucidate the regulatory mechanism for tumor growth, PanNET grafts were transplanted into subrenal capsules in GCGKO and control mice. Ki-67 positive cells were identified in panNET grafts transplanted to GCGKO mice 1 month after transplantation, but not in those to control mice. These results suggest that humoral factors or conditions specific to GCGKO mice, are involved in the proliferation of panNETs. Taken together, GCGKO mice are novel animal model for studying the development, pathogenesis, and metastasis panNETs.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucagon/metabolism , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Glucagon/genetics , Glucagon-Like Peptide 1/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proglucagon/genetics , Proglucagon/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Histochem Cytochem ; 50(3): 311-24, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11850434

ABSTRACT

We investigated the densities of the L-type Ca(2+) current, i(Ca,L), and various Ca(2+) handling proteins in rabbit sinoatrial (SA) node. The density of i(Ca,L), recorded with the whole-cell patch-clamp technique, varied widely in sinoatrial node cells. The density of i(Ca,L) was significantly (p<0.001) correlated with cell capacitance (measure of cell size) and the density was greater in larger cells (likely to be from the periphery of the SA node) than in smaller cells (likely to be from the center of the SA node). Immunocytochemical labeling of the L-type Ca(2+) channel, Na(+)-Ca(2+) exchanger, sarcoplasmic reticulum Ca(2+) release channel (RYR2), and sarcoplasmic reticulum Ca(2+) pump (SERCA2) also varied widely in SA node cells. In all cases there was significantly (p<0.05) denser labeling of cells from the periphery of the SA node than of cells from the center. In contrast, immunocytochemical labeling of the Na(+)-K(+) pump was similar in peripheral and central cells. We conclude that Ca(2+) handling proteins are sparse and poorly organized in the center of the SA node (normally the leading pacemaker site), whereas they are more abundant in the periphery (at the border of the SA node with the surrounding atrial muscle).


Subject(s)
Calcium Channels, L-Type/analysis , Calcium/metabolism , Animals , Calcium-Transporting ATPases/analysis , Immunohistochemistry , Rabbits , Ryanodine Receptor Calcium Release Channel/analysis , Sarcolemma/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/analysis , Sodium-Potassium-Exchanging ATPase/analysis , Tissue Distribution
13.
PLoS One ; 8(5): e64415, 2013.
Article in English | MEDLINE | ID: mdl-23671715

ABSTRACT

Defects in glucagon action can cause hyperplasia of islet α-cells, however, the underlying mechanisms remain largely to be elucidated. Mice homozygous for a glucagon-GFP knock-in allele (Gcg(gfp/gfp) ) completely lack proglucagon-derived peptides and exhibit hyperplasia of GFP-positive α-like cells. Expression of the transcription factor, aristaless-related homeobox (ARX), is also increased in the Gcg(gfp/gfp) pancreas. Here, we sought to elucidate the role of ARX in the hyperplasia of α-like cells through analyses of two Arx mutant alleles (Arx(P355L/Y) and Arx ([330insGCG]7/Y) ) that have different levels of impairment of their function. Expression of Gfp and Arx genes was higher and the size and number of islets increased in the Gcg(gfp/gfp) pancreas compared to and Gcg(gfp/+) pancreas at 2 weeks of age. In male Gcg(gfp/gfp) mice that are hemizygous for the Arx(P355L/Y) mutation that results in a protein with a P355L amino acid substitution, expression of Gfp mRNA in the pancreas was comparable to that in control Gcg(gfp/+)Arx(+/Y) mice. The increases in islet size and number were also reduced in these mice. Immunohistochemical analysis showed that the number of GFP-positive cells was comparable in Gcg(gfp/gfp) Arx(P355L/Y) and Gcg(gfp/+)Arx(+/Y) mice. These results indicate that the hyperplasia is reduced by introduction of an Arx mutation. Arx(P355L/Y) mice appeared to be phenotypically normal; however, Arx ([330insGCG]7/Y) mice that have a mutant ARX protein with expansion of the polyalanine tract had a reduced body size and shortened life span. The number of GFP positive cells was further reduced in the Gcg(gfp/gfp) Arx ([330insGCG]7/Y) mice. Taken together, our findings show that the function of ARX is one of the key modifiers for hyperplasia of islet α-like cells in the absence of proglucagon-derived peptides.


Subject(s)
Homeodomain Proteins/metabolism , Islets of Langerhans/metabolism , Peptides/metabolism , Proglucagon/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Female , Gene Expression , Glucagon/genetics , Glucagon/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Hyperplasia , Immunohistochemistry , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Peptides/genetics , Proglucagon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
14.
PLoS One ; 8(7): e65202, 2013.
Article in English | MEDLINE | ID: mdl-23843937

ABSTRACT

Molecular signaling of cardiac autonomic innervation is an unresolved issue. Here, we show that glial cell line-derived neurotrophic factor (GDNF) promotes cardiac sympathetic innervation in vitro and in vivo. In vitro, ventricular myocytes (VMs) and sympathetic neurons (SNs) isolated from neonatal rat ventricles and superior cervical ganglia were cultured at a close distance. Then, morphological and functional coupling between SNs and VMs was assessed in response to GDNF (10 ng/ml) or nerve growth factor (50 ng/ml). As a result, fractions of neurofilament-M-positive axons and synapsin-I-positive area over the surface of VMs were markedly increased with GDNF by 9-fold and 25-fold, respectively, compared to control without neurotrophic factors. Pre- and post-synaptic stimulation of ß1-adrenergic receptors (BAR) with nicotine and noradrenaline, respectively, resulted in an increase of the spontaneous beating rate of VMs co-cultured with SNs in the presence of GDNF. GDNF overexpressing VMs by adenovirus vector (AdGDNF-VMs) attracted more axons from SNs compared with mock-transfected VMs. In vivo, axon outgrowth toward the denervated myocardium in adult rat hearts after cryoinjury was also enhanced significantly by adenovirus-mediated GDNF overexpression. GDNF acts as a potent chemoattractant for sympathetic innervation of ventricular myocytes, and is a promising molecular target for regulation of cardiac function in diseased hearts.


Subject(s)
Axons/drug effects , Ganglia, Sympathetic/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Myocytes, Cardiac/metabolism , Neurons/drug effects , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Fibers, Postganglionic/growth & development , Animals , Autonomic Denervation , Axons/metabolism , Coculture Techniques , Excitation Contraction Coupling/physiology , Ganglia, Sympathetic/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Nerve Growth Factor/pharmacology , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Neurons/metabolism , Rats , Receptors, Adrenergic, beta-1/metabolism
15.
Biomed Res Int ; 2013: 532720, 2013.
Article in English | MEDLINE | ID: mdl-24066291

ABSTRACT

The aim of myocardial tissue engineering is to repair or regenerate damaged myocardium with engineered cardiac tissue. However, this strategy has been hampered by lack of functional integration of grafts with native myocardium. Autonomic innervation may be crucial for grafts to function properly with host myocardium. In this study, we explored the feasibility of in vivo induction of autonomic innervation to engineered myocardial tissue using genetic modulation by adenovirus encoding glial cell line derived neurotrophic factor (GDNF). GFP-transgene (control group) or GDNF overexpressing (GDNF group) engineered cardiomyocyte sheets were transplanted on cryoinjured hearts in rats. Nerve fibers in the grafts were examined by immunohistochemistry at 1, 2, and 4 weeks postoperatively. Growth associated protein-43 positive growing nerves and tyrosine hydroxylase positive sympathetic nerves were first detected in the grafts at 2 weeks postoperatively in control group and 1 week in GDNF group. The densities of growing nerve and sympathetic nerve in grafts were significantly increased in GDNF group. No choline acetyltransferase immunopositive parasympathetic nerves were observed in grafts. In conclusion, sympathetic innervation could be effectively induced into engrafted engineered cardiomyocyte sheets using GDNF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/genetics , Heart Transplantation , Heart/innervation , Myocytes, Cardiac/metabolism , Tissue Engineering , Adenoviridae , Animals , Humans , Myocytes, Cardiac/transplantation , Neuroglia/metabolism , Neuroglia/pathology , Rats , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/surgery , Transplants/growth & development , Transplants/innervation
16.
Diabetes ; 62(2): 510-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23099862

ABSTRACT

Glucagon and glucagon-like peptide-1 (GLP-1) are produced in pancreatic α-cells and enteroendocrine L-cells, respectively, in a tissue-specific manner from the same precursor, proglucagon, that is encoded by glucagon gene (Gcg), and play critical roles in glucose homeostasis. Here, we studied glucose homeostasis and ß-cell function of Gcg-deficient mice that are homozygous for a Gcg-GFP knock-in allele (Gcg(gfp/gfp)). The Gcg(gfp/gfp) mice displayed improved glucose tolerance and enhanced insulin secretion, as assessed by both oral glucose tolerance test (OGTT) and intraperitoneal glucose tolerance test (IPGTT). Responses of glucose-dependent insulinotropic polypeptide (GIP) to both oral and intraperitoneal glucose loads were unexpectedly enhanced in Gcg(gfp/gfp) mice, and immunohistochemistry localized GIP to pancreatic ß-cells of Gcg(gfp/gfp) mice. Furthermore, secretion of GIP in response to glucose was detected in isolated islets of Gcg(gfp/gfp) mice. Blockade of GIP action in vitro and in vivo by cAMP antagonism and genetic deletion of the GIP receptor, respectively, almost completely abrogated enhanced insulin secretion in Gcg(gfp/gfp) mice. These results indicate that ectopic GIP expression in ß-cells maintains insulin secretion in the absence of proglucagon-derived peptides (PGDPs), revealing a novel compensatory mechanism for sustaining incretin hormone action in islets.


Subject(s)
Gastric Inhibitory Polypeptide/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Proglucagon/metabolism , Animals , Cyclic AMP/antagonists & inhibitors , Gastric Inhibitory Polypeptide/genetics , Gene Deletion , Gene Knock-In Techniques , Glucagon-Like Peptide-1 Receptor , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Incretins/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Mice , Proglucagon/analysis , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/metabolism
17.
Neurosci Res ; 73(1): 56-60, 2012 May.
Article in English | MEDLINE | ID: mdl-22381959

ABSTRACT

Delayed onset muscle soreness (DOMS) appears with some delay after unaccustomed, strenuous exercise, especially after lengthening contraction (LC). It is characterized by tenderness and movement related pain, namely muscular mechanical hyperalgesia. To clarify the involvement of C-fibers in this mechanical hyperalgesia, we examined whether DOMS could be induced in rats treated neonatally with capsaicin. We confirmed that a large portion of unmyelinated afferent fibers were lost in capsaicin treated rats. In these animals, LC failed to induce muscular mechanical hyperalgesia. mRNA of nerve growth factor (NGF) in the muscle, which plays a pivotal role in maintaining mechanical hyperalgesia, was upregulated in the capsaicin treated animals similar to the vehicle treated animals. These results demonstrate that C-fiber afferents are essential in transmitting the nociceptive information from exercised muscle in DOMS.


Subject(s)
Capsaicin/toxicity , Hyperalgesia/prevention & control , Hyperalgesia/physiopathology , Pain/physiopathology , Physical Conditioning, Animal/methods , Animals , Animals, Newborn , Hyperalgesia/chemically induced , Male , Muscle Contraction/physiology , Nerve Fibers, Unmyelinated/physiology , Pain/chemically induced , Pain/pathology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Time Factors
18.
Diabetes ; 61(1): 74-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22187375

ABSTRACT

Glucagon is believed to be one of the most important peptides for upregulating blood glucose levels. However, homozygous glucagon-green fluorescent protein (gfp) knock-in mice (Gcg(gfp/gfp): GCGKO) are normoglycemic despite the absence of proglucagon-derived peptides, including glucagon. To characterize metabolism in the GCGKO mice, we analyzed gene expression and metabolome in the liver. The expression of genes encoding rate-limiting enzymes for gluconeogenesis was only marginally altered. On the other hand, genes encoding enzymes involved in conversion of amino acids to metabolites available for the tricarboxylic acid cycle and/or gluconeogenesis showed lower expression in the GCGKO liver. The expression of genes involved in the metabolism of fatty acids and nicotinamide was also altered. Concentrations of the metabolites in the GCGKO liver were altered in manners concordant with alteration in the gene expression patterns, and the plasma concentrations of amino acids were elevated in the GCGKO mice. The insulin concentration in serum and phosphorylation of Akt protein kinase in liver were reduced in GCGKO mice. These results indicated that proglucagon-derived peptides should play important roles in regulating various metabolic pathways, especially that of amino acids. Serum insulin concentration is lowered to compensate the impacts of absent proglucagon-derived peptide on glucose metabolism. On the other hand, impacts on other metabolic pathways are only partially compensated by reduced insulin action.


Subject(s)
Amino Acids/blood , Liver/metabolism , Metabolic Diseases/metabolism , Proglucagon/deficiency , Proglucagon/genetics , Amino Acids/metabolism , Animals , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver/enzymology , Male , Metabolic Diseases/blood , Metabolic Diseases/genetics , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Peptide Fragments/chemistry , Peptide Fragments/deficiency , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proglucagon/chemistry , Proglucagon/metabolism , Up-Regulation
19.
PLoS One ; 6(1): e14605, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21298050

ABSTRACT

Obesity is a serious international health problem that increases the risk of several diet-related chronic diseases. The genetic factors predisposing to obesity are little understood. Rcan2 was originally identified as a thyroid hormone-responsive gene. In the mouse, two splicing variants that harbor distinct tissue-specific expression patterns have been identified: Rcan2-3 is expressed predominately in the brain, whereas Rcan2-1 is expressed in the brain and other tissues such as the heart and skeletal muscle. Here, we show that Rcan2 plays an important role in the development of age- and diet-induced obesity. We found that although the loss of Rcan2 function in mice slowed growth in the first few weeks after birth, it also significantly ameliorated age- and diet-induced obesity in the mice by causing a reduction in food intake rather than increased energy expenditure. Rcan2 expression was most prominent in the ventromedial, dorsomedial and paraventricular hypothalamic nuclei governing energy balance. Fasting and refeeding experiment showed that only Rcan2-3 mRNA expression is up-regulated in the hypothalamus by fasting, and loss of Rcan2 significantly attenuates the hyperphagic response to starvation. Using double-mutant (Lep(ob/ob) Rcan2(-/-)) mice, we were also able to demonstrate that Rcan2 and leptin regulate body weight through different pathways. Our findings indicate that there may be an Rcan2-dependent mechanism which regulates food intake and promotes weight gain through a leptin-independent pathway. This study provides novel information on the control of body weight in mice and should improve our understanding of the mechanisms of obesity in humans.


Subject(s)
Aging , Eating/genetics , Obesity/etiology , Proteins/genetics , Animal Feed , Animals , Body Weight , Intracellular Signaling Peptides and Proteins , Leptin/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Obesity/prevention & control
20.
Biomed Res ; 31(6): 353-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21187646

ABSTRACT

Molecular signaling of sympathetic innervation of myocardium is an unresolved issue. The purpose of this study was to investigate the effect of neurotrophic factors on sympathetic neurite growth towards cardiomyocytes. Cardiomyocytes (CMs) and sympathetic neurons (SNs) were isolated from neonatal rat hearts and superior cervical ganglia, and were co-cultured, either in a random or localized way. Neurite growth from SNs toward CMs was assessed by immunohistochemistry for neurofilament M and α-actinin in response to neurotrophic factors-nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), ciliary neurotrophic factor (CNTF) and a chemical repellent, semaphorin 3A. As a result, GDNF as well as NGF and BDNF stimulated neurite growth. GDNF enhanced neurite outgrowth even under the NGF-depleted culture condition, excluding an indirect effect of GDNF via NGF. Quantification of mRNA and protein by real-time PCR and immunohistochemistry at different developmental stages revealed that GDNF is abundantly expressed in the hearts of embryos and neonates, but not in adult hearts. GDNF plays an important role in inducing cardiac sympathetic innervation at the early developmental stages. A possible role in (re)innervation of injured or transplanted or cultured and transplanted myocardium may deserve investigation.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Heart/embryology , Myocytes, Cardiac/cytology , Neurites/physiology , Sympathetic Nervous System/cytology , Actinin , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Ganglia/cytology , Gene Expression Regulation, Developmental , Nerve Growth Factor/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Sympathetic Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL