Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 394
Filter
1.
Sci Rep ; 11(1): 22745, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815492

ABSTRACT

Although Krüppel-associated box domain-containing zinc-finger proteins (K-ZNFs) may be associated with sophisticated gene regulation in higher organisms, the physiological functions of most K-ZNFs remain unknown. The Zfp212 protein was highly conserved in mammals and abundant in the brain; it was mainly expressed in the cerebellum (Cb). Zfp212 (mouse homolog of human ZNF212) knockout (Zfp212-KO) mice showed a reduction in survival rate compared to wild-type mice after 20 months of age. GABAergic Purkinje cell degeneration in the Cb and aberrant locomotion were observed in adult Zfp212-KO mice. To identify genes related to the ataxia-like phenotype of Zfp212-KO mice, 39 ataxia-associated genes in the Cb were monitored. Substantial alterations in the expression of ataxin 10, protein phosphatase 2 regulatory subunit beta, protein kinase C gamma, and phospholipase D3 (Pld3) were observed. Among them, Pld3 alone was tightly regulated by Flag-tagged ZNF212 overexpression or Zfp212 knockdown in the HT22 cell line. The Cyclic Amplification and Selection of Targets assay identified the TATTTC sequence as a recognition motif of ZNF212, and these motifs occurred in both human and mouse PLD3 gene promoters. Adeno-associated virus-mediated introduction of human ZNF212 into the Cb of 3-week-old Zfp212-KO mice prevented Purkinje cell death and motor behavioral deficits. We confirmed the reduction of Zfp212 and Pld3 in the Cb of an alcohol-induced cerebellar degeneration mouse model, suggesting that the ZNF212-PLD3 relationship is important for Purkinje cell survival.


Subject(s)
Ataxia/pathology , DNA-Binding Proteins/metabolism , Gait Disorders, Neurologic/pathology , Nerve Tissue Proteins/physiology , Phospholipase D/antagonists & inhibitors , Purkinje Cells/pathology , Animals , Ataxia/etiology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , Gait Disorders, Neurologic/etiology , Humans , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism
2.
J Neurochem ; 158(5): 1172-1185, 2021 09.
Article in English | MEDLINE | ID: mdl-34287909

ABSTRACT

Neuropeptide cocaine- and amphetamine-regulated transcript (CART) is known to influence the activity of the canonical mesolimbic dopaminergic pathway and modulate reward seeking behaviour. CART neurons of the lateral hypothalamus (LH) send afferents to the ventral tegmental area (VTA) and paraventricular thalamic nucleus (PVT) and these nuclei, in turn, send secondary projections to nucleus accumbens. We try to dissect the precise sites of CART's action in these circuits in promoting reward. Rats were implanted with bipolar electrode targeted at the lateral hypothalamus-medial forebrain bundle (LH-MFB) and trained to press the lever through intracranial self-stimulation (ICSS) protocol. CART (55-102) administered directly into posterior VTA (pVTA) or PVT of the conditioned rats significantly increased the number of lever presses, indicating reward-promoting activity of the peptide. Concomitant increase in dopamine (DA) and 3, 4-dihydroxyphenylacetic acid (DOPAC) efflux was noted in the microdialysate collected from the nucleus accumbens shell (AcbSh). On the other hand, immunoneutralization of endogenous CART with CART antibodies injected directly in the pVTA or PVT reduced the lever press activity as well as DA and DOPAC efflux in the AcbSh. Injection of CART (1-39) in pVTA or PVT was ineffective. We suggest that CART cells in the LH-MFB area send afferents to (a) pVTA and influence dopaminergic neurons projecting to AcbSh and (b) PVT, from where the secondary neurons may feed into the AcbSh. Excitation of the CARTergic pathway to the pVTA as well as the PVT seems to promote DA release in the AcbSh and contribute to the generation of reward.


Subject(s)
Dopamine/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/metabolism , Reward , Animals , Electrodes, Implanted , Male , Microdialysis/methods , Nerve Net/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Wistar
3.
J Neuroinflammation ; 18(1): 53, 2021 Feb 21.
Article in English | MEDLINE | ID: mdl-33612107

ABSTRACT

BACKGROUND: Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is a recently discovered autoimmune syndrome associated with psychosis, dyskinesia, and seizures. However, the underlying mechanisms of this disease remain unclear, in part because of a lack of suitable animal models. METHODS: This study describes a novel female C57BL/6 mouse model of anti-NMDAR encephalitis that was induced by active immunization against NMDARs using an amino terminal domain (ATD) peptide from the GluN1 subunit (GluN1356-385). RESULTS: Twelve weeks after immunization, the immunized mice showed significant memory loss. Furthermore, antibodies from the cerebrospinal fluid of immunized mice decreased the surface NMDAR cluster density in hippocampal neurons which was similar to the effect induced by the anti-NMDAR encephalitis patients' antibodies. Immunization also impaired long-term potentiation at Schaffer collateral-CA1 synapses and reduced NMDAR-induced calcium influx. CONCLUSION: We established a novel anti-NMDAR encephalitis model using active immunization with peptide GluN1356-385 targeting the ATD of GluN1. This novel model may allow further research into the pathogenesis of anti-NMDAR encephalitis and aid in the development of new therapies for this disease.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/chemically induced , Nerve Tissue Proteins/administration & dosage , Peptide Fragments/administration & dosage , Receptors, N-Methyl-D-Aspartate/administration & dosage , Vaccination/adverse effects , Amino Acid Sequence , Animals , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/genetics , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Autoantibodies/genetics , Autoantibodies/immunology , Cells, Cultured , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/immunology , Vaccination/methods
4.
CNS Neurosci Ther ; 27(1): 60-70, 2021 01.
Article in English | MEDLINE | ID: mdl-33314664

ABSTRACT

Recent clinical studies suggest that pentraxin 3 (PTX3), which is known as an acute-phase protein that is produced rapidly at local sites of inflammation, may be a new biomarker of disease risk for central nervous system disorders, including stroke. However, the effects of PTX3 on cerebrovascular function in the neurovascular unit (NVU) after stroke are mostly unknown, and the basic research regarding the roles of PTX3 in NVU function is still limited. In this reverse translational study, we prepared mouse models of white matter stroke by vasoconstrictor (ET-1 or L-Nio) injection into the corpus callosum region to examine the roles of PTX3 in the pathology of cerebral white matter stroke. PTX3 expression was upregulated in GFAP-positive astrocytes around the affected region in white matter for at least 21 days after vasoconstrictor injection. When PTX3 expression was reduced by PTX3 siRNA, blood-brain barrier (BBB) damage at day 3 after white matter stroke was exacerbated. In contrast, when PTX3 siRNA was administered at day 7 after white matter stroke, compensatory angiogenesis at day 21 was promoted. In vitro cell culture experiments confirmed the inhibitory effect of PTX3 in angiogenesis, that is, recombinant PTX3 suppressed the tube formation of cultured endothelial cells in a Matrigel-based in vitro angiogenesis assay. Taken together, our findings may support a novel concept that astrocyte-derived PTX3 plays biphasic roles in cerebrovascular function after white matter stroke; additionally, it may also provide a proof-of-concept that PTX3 could be a therapeutic target for white matter-related diseases, including stroke.


Subject(s)
Blood-Brain Barrier/metabolism , C-Reactive Protein/biosynthesis , Nerve Tissue Proteins/biosynthesis , Recovery of Function/physiology , Stroke/drug therapy , Stroke/metabolism , White Matter/metabolism , Aged , Aged, 80 and over , Animals , Blood-Brain Barrier/drug effects , C-Reactive Protein/administration & dosage , C-Reactive Protein/antagonists & inhibitors , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Rats , Recovery of Function/drug effects , Stroke/pathology , White Matter/drug effects , White Matter/pathology
5.
Transl Stroke Res ; 12(1): 125-135, 2021 02.
Article in English | MEDLINE | ID: mdl-32378028

ABSTRACT

Adipose-derived mesenchymal stem cells markedly attenuated brain infarct size and improved neurological function in rats. The mechanisms for neuronal cell death have previously been defined in stress states to suggest that an influx of calcium ions into the neurons activates calpain cleavage of p35 into p25 forming a hyperactive complex that induces cell death. Now we report that p5, a 24-residue peptide derived from p35, offers protection to neurons and endothelial cells in vitro. In vivo administration of human adipose-derived mesenchymal stem cells (hADMSCs) loaded with this therapeutic peptide to post-stroke rats had no effect on the infarct volume. Nevertheless, the treatment led to improvement in functional recovery in spatial learning and memory (water maze), bilateral coordination and sensorimotor function (rotating pole), and asymmetry of forelimb usage (cylinder test). However, the treatment may not impact on cutaneous sensitivity (adhesive tape removal test). In addition, the double immunofluorescence with human cell-specific antibodies revealed that the number of surviving transplanted cells was higher in the peri-infarcted area of animals treated with hADMSCs + P5 than that in hADMSC-treated or control animals, concomitant with reduced number of phagocytic, annexin3-positive cells in the peri-infarcted region. However, the combination therapy did not increase the vascular density in the peri-infarcted area after stroke. In conclusion, administration of hADMSC-loaded p5 peptide to post-stroke rats created conditions that supported survival of drug-loaded hADMSCs after cerebral ischemia, suggesting its therapeutic potential in patients with stroke.


Subject(s)
Adipose Tissue/transplantation , Brain Ischemia/therapy , Disease Models, Animal , Mesenchymal Stem Cell Transplantation/methods , Nerve Tissue Proteins/administration & dosage , Peptide Fragments/administration & dosage , Recovery of Function/drug effects , Adipose Tissue/physiology , Amino Acid Sequence , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Line, Tumor , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mesenchymal Stem Cells/physiology , Nerve Tissue Proteins/genetics , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy , Peptide Fragments/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology
6.
J Endocrinol ; 244(1): 1-12, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31536964

ABSTRACT

We recently discovered a novel gene encoding a small secretory protein, neurosecretory protein GL (NPGL), which stimulates feeding behavior in mice following acute administration. These findings suggest that dysregulation of NPGL contributes to obesity and metabolic disease. To explore this possibility, we investigated the impact of prolonged exposure to NPGL through 13 days of chronic intracerebroventricular (i.c.v.) infusion and examined feeding behavior, body composition, expressions of lipid metabolic factors, respiratory metabolism, locomotor activity, and food preference. Under standard chow diet, NPGL increased white adipose tissue (WAT) mass without affecting feeding behavior and body mass. In contrast, when fed a high-calorie diet, NPGL stimulated feeding behavior and increased body mass concomitant with marked fat accumulation. Quantitative reverse transcription polymerase chain reaction (RT-PCR) analysis revealed that mRNA expressions for key enzymes and related factors involved in lipid metabolism were increased in WAT and liver. Likewise, analyses of respiratory metabolism and locomotor activity revealed that energy expenditure and locomotor activity were significantly decreased by NPGL. In contrast, selective feeding of macronutrients did not alter food preference in response to NPGL, although total calorie intake was increased. Immunohistochemical analysis revealed that NPGL-containing cells produce galanin, a neuropeptide that stimulates food intake. Taken together, these results provide further support for NPGL as a novel regulator of fat deposition through changes in energy intake and locomotor activity.


Subject(s)
Adipose Tissue, White/drug effects , Body Composition/drug effects , Feeding Behavior/drug effects , Nerve Tissue Proteins/administration & dosage , Animal Feed , Animals , Energy Metabolism/drug effects , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects
7.
Brain Res ; 1728: 146595, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31830460

ABSTRACT

Reward deficit, expressed as anhedonia, is one of the major symptoms associated with neuropsychiatric disorders, but the underlying maladaptations have not been understood. Herein, we test the hypothesis that the neuropeptide cocaine- and amphetamine-regulated transcript (CART) may participate in the process. The study is justified since the peptide is a major player in inducing satiety and also processing of reward. The rats were socially isolated to induce reward deficit and conditioned to self-stimulate via an electrode in lateral hypothalamus (LH)-medial forebrain bundle (MFB) region. Compared to group-housed control rats, the socially isolated animals showed decreased lever press activity and elevated ICSS threshold indicating anhedonia-like condition. However, the effects of social isolation were alleviated by CART administered via intracerebroventricular route. The changes in the expression of CART protein and mRNA were screened using immunofluorescence and qRT-PCR methods, respectively. Socially isolated rats showed reduction in the expression of CART in the LH, nucleus accumbens shell (AcbSh) and posterior ventral tegmental area (pVTA) and CART mRNA in the Acb and LH. Double immunostaining with antibodies against CART and synaptophysin revealed significant loss of colabeled elements in LH, AcbSh and pVTA. We suggest that down-regulation of endogenous CARTergic system in the LH-pVTA-AcbSh reward circuitry may be causal to motivational anhedonia like phenotype seen in neuropsychiatric conditions.


Subject(s)
Nerve Tissue Proteins/physiology , Reward , Social Isolation , Anhedonia , Animals , Hypothalamic Area, Lateral/metabolism , Locomotion , Male , Medial Forebrain Bundle/metabolism , Motivation , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Rats , Rats, Wistar , Self Stimulation/physiology
8.
Cells ; 8(8)2019 07 29.
Article in English | MEDLINE | ID: mdl-31362466

ABSTRACT

NK cells (natural killer cells) being a part of the innate immune system have been shown to be involved in immunoregulation of autoimmune diseases. Previously we have shown that HINT1/Hsp70 treatment induced regulatory NK cells ameliorating experimental autoimmune encephalomyelitis (EAE) course and CD4+ T cells proliferation. NK cells were isolated from mice treated with HINT1/Hsp70 and co-cultured with proteolipid protein (PLP)-stimulated CD4+ T cells isolated from EAE mice. Cell proliferation was assessed by thymidine uptake, cytotoxicity by lactate dehydrogenase (LDH) release assay and fluorescence activated cell sorting (FACS) analysis, protein expression by Western blot, mRNA by quantitative RT-PCR. Gene related to anergy in lymphocytes (GRAIL) expression was downregulated by specific siRNA and GRAIL overexpression was induced by pcDNA-GRAIL transfection. HINT1/Hsp70 pretreatment of EAE SJL/J mice ameliorated EAE course, suppressed PLP-induced T cell proliferation by enhancing T cell expression of GRAIL as GRAIL downregulation restored T cell proliferation. HINT1/Hsp70 treatment induced immunoregulatory NK cells which inhibited PLP-stimulated T cell proliferation not depending on T cell necrosis and apoptosis. This immunoregulatory NK cell function depended on NK cell expression of GRAIL as GRAIL downregulation diminished inhibition of NK cell suppression of T cell proliferation. Similarly GRAIL overexpression in NK cells induced their regulatory function. HINT1/Hsp70 treatment generated regulatory NK cells characterized by expression of GRAIL.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , HSP70 Heat-Shock Proteins/administration & dosage , Killer Cells, Natural/cytology , Nerve Tissue Proteins/administration & dosage , Ubiquitin-Protein Ligases/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Clonal Anergy , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , HSP70 Heat-Shock Proteins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mice , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/metabolism , Nerve Tissue Proteins/pharmacology , Proteolipids/pharmacology , Ubiquitin-Protein Ligases/metabolism
9.
Pain ; 160(8): 1847-1855, 2019 08.
Article in English | MEDLINE | ID: mdl-31335652

ABSTRACT

Data from preclinical research have been suggested to suffer from a lack of inherent reproducibility across laboratories. The goal of our study was to replicate findings from a previous report that demonstrated positive effects of Meteorin, a novel neurotrophic factor, in a rat model of neuropathic pain induced by chronic constriction injury (CCI). Notably, 5 to 6 intermittent subcutaneous (s.c.) injections of Meteorin had been reported to produce reversal of mechanical allodynia/thermal hyperalgesia after injury, wherein maximum efficacy of Meteorin was reached slowly and outlasted the elimination of the compound from the blood by several weeks. Here, we evaluated the efficacy of Meteorin in reversing hindpaw mechanical hyperalgesia and cold allodynia in male, Sprague-Dawley rats with CCI. Nociceptive behavior was monitored before and after CCI, and after drug treatment until day 42 after injury. Systemic administration of recombinant mouse Meteorin (0.5 and 1.8 mg/kg, s.c.) at days 10, 12, 14, 17, and 19 after CCI produced a prolonged reversal of neuropathic hypersensitivity with efficacy comparable with that obtained with gabapentin (100 mg/kg, orally). Despite some protocol deviations (eg, nociceptive endpoint, animal vendor, testing laboratory, investigator, etc.) being incurred, these did not affect study outcome. By paying careful attention to key facets of study design, using bioactive material, and confirming drug exposure, the current data have replicated the salient findings of the previous study, promoting confidence in further advancement of this novel molecule as a potential therapy for neuropathic pain.


Subject(s)
Analgesics/therapeutic use , Hyperalgesia/drug therapy , Nerve Tissue Proteins/therapeutic use , Neuralgia/drug therapy , Analgesics/administration & dosage , Animals , Disease Models, Animal , Hyperalgesia/etiology , Male , Nerve Tissue Proteins/administration & dosage , Neuralgia/etiology , Pain Measurement , Pain Threshold/drug effects , Peripheral Nerve Injuries/complications , Rats , Rats, Sprague-Dawley , Reproducibility of Results
10.
Pharmacol Biochem Behav ; 181: 101-109, 2019 06.
Article in English | MEDLINE | ID: mdl-31054945

ABSTRACT

While insulin secreted from pancreas plays a pivotal role in the control of glucose homeostasis, it also interacts with hypothalamic sites and negatively influences the energy balance. The present study was undertaken to reveal the functional interaction between cocaine- and amphetamine-regulated transcript (CART), a well-known anorexic peptide, and insulin within the framework of hypothalamus in the regulation of feeding behavior and body weight. Insulin was administered daily by intracerebroventricular (icv) route, alone or in combination with CART (icv) for a period of seven days. Immediately thereafter, preweighed food was offered to the animals at the commencement of the dark phase. The food intake and body weight were measured daily just prior to next injection. Furthermore, brains of insulin-treated rats were processed for the immunohistochemical analysis of CART-containing elements in the hypothalamus. Treatment with insulin (6 mU, icv) for a period of 7 days caused a significant decrease in food intake and body weight as compared to control. Concomitant administration of CART (0.5 µg, icv) potentiated insulin-induced anorexia and weight loss. Insulin administration resulted in a significant increase in CART immunoreactivity in the hypothalamic arcuate, paraventricular, dorsomedial and ventromedial nuclei. We suggest that increased CART contents in the hypothalamus may be causally linked with anorexia and weight loss induced by insulin.


Subject(s)
Body Weight/drug effects , Feeding Behavior/drug effects , Insulin/pharmacology , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/pharmacology , Neuropeptides/immunology , Neuropeptides/pharmacology , Animals , Anorexia/chemically induced , Antibodies, Monoclonal/pharmacology , Eating/drug effects , Hypothalamus/drug effects , Hypothalamus/immunology , Immunohistochemistry , Insulin/administration & dosage , Male , Nerve Tissue Proteins/administration & dosage , Neuropeptides/administration & dosage , Photoperiod , Rats , Rats, Sprague-Dawley , Weight Loss/drug effects
11.
J Mol Histol ; 50(3): 253-261, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30937700

ABSTRACT

Nel-like molecule-1 (NELL-1) is a novel highly specific growth factor that can induce osteoblast differentiation and bone formation as well as odontoblast differentiation. Recent studies have suggested that NELL-1 can synergistically increase bone formation and regeneration with bone morphogenetic protein 2 (BMP2) and inhibit adverse effects induced by BMP2. This study aimed to evaluate the combined effects of NELL-1 and BMP2 on rat pulp repair. The experiment used healthy non-carious maxillary first molars from 60 Wistar rats. Exposed pulps were capped with NELL-1 plus BMP2, NELL-1 alone, and BMP2 alone, and each was absorbed onto a sterile collagen sponge. In the control samples, the collagen sponge alone and Dycal were used as capping agents. After l, 2 and 4 weeks, the rats were sacrificed. The formation of reparative dentin, as well the situation of pulp repair, was detected by hematoxylin-eosin (HE) staining; moreover, the expression of dentin specific protein-dentin sialophosphoprotein (DSPP) and the pro-inflammatory cytokines interleukin-6 (IL6) and interleukin-8 (IL8) was detected by immunohistochemical staining. Quantitative real-time PCR experiment was used to investigate the mRNA levels of IL6 and IL8. The results showed that pulp capping with NELL-1 plus BMP2 in rats had superior ability in inducing reparative dentin formation with dentin tubules and in reducing the inflammatory cell response compared with the other groups. These findings suggested that combined use of NELL-1 and BMP2 could positively regulate pulp repair.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Dental Pulp Capping , Dental Pulp/growth & development , Nerve Tissue Proteins/genetics , Animals , Bone Morphogenetic Protein 2/administration & dosage , Cell Differentiation/drug effects , Dental Pulp/metabolism , Dentin/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Interleukin-6/genetics , Interleukin-8/genetics , Nerve Tissue Proteins/administration & dosage , Osteogenesis/drug effects , Osteogenesis/genetics , Phosphoproteins/genetics , Rats , Rats, Wistar , Sialoglycoproteins/genetics
12.
Mol Med Rep ; 19(4): 3255-3262, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816469

ABSTRACT

Repulsive guidance molecule a (RGMa) is a membrane­associated glycoprotein that regulates axonal guidance and inhibits axon outgrowth. In our previous study, we hypothesized that RGMa may be involved in temporal lobe epilepsy (TLE) via the repulsive guidance molecule a (RGMa)­focal adhesion kinase (FAK)­Ras signaling pathway. To investigate the role of RGMa in epilepsy, recombinant RGMa protein and FAK inhibitor 14 was intracerebroventricularly injected into a pentylenetetrazol (PTZ) kindling model and Timm staining, co­immunoprecipitation and western blotting analyses were subsequently performed. The results of the present study revealed that intracerebroventricular injection of recombinant RGMa protein reduced the phosphorylation of FAK (Tyr397) and intracerebroventricular injection of FAK inhibitor 14 reduced the interaction between FAK and p120GAP, as wells as Ras expression. Recombinant RGMa protein and FAK inhibitor 14 exerted seizure­suppressant effects; however, recombinant RGMa protein but not FAK inhibitor 14 suppressed mossy fiber sprouting in the PTZ kindling model. Collectively, these results demonstrated that RGMa may be considered as a potential therapeutic agent for epilepsy, and that RGMa may exert the aforementioned biological effects partly via the FAK­p120GAP­Ras signaling pathway.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Membrane Glycoproteins/metabolism , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/metabolism , Seizures/metabolism , Seizures/physiopathology , Signal Transduction , p120 GTPase Activating Protein/metabolism , ras Proteins/metabolism , Animals , Disease Models, Animal , GPI-Linked Proteins , Gene Expression , Male , Membrane Glycoproteins/administration & dosage , Mossy Fibers, Hippocampal/physiopathology , Nerve Tissue Proteins/administration & dosage , Pentylenetetrazole/adverse effects , Phosphorylation , Protein Binding , Rats , Recombinant Proteins , Seizures/drug therapy , Seizures/etiology , Signal Transduction/drug effects , ras Proteins/genetics
13.
Brain Res ; 1710: 117-124, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30610873

ABSTRACT

Cocaine- and amphetamine-regulated transcript peptide (CARTp) is present in neurons and varicose fibers in the rostral ventrolateral medulla (RVLM) that is crucial in the control of cardiovascular function. Prior research indicated that intracisternal administration of CARTp evokes hypertension and accumulation of Fos in the RVLM. Despite the interaction among CARTp, cardiovascular effect, and the RVLM, no studies have directly examined whether CARTp participates in cardiovascular regulation in the RVLM. The current study directly examined the modulation of blood pressure and baroreflex sensitivity by CARTp in the RVLM in the different strain of rats. Immunohistochemical study showed that CARTp immunoreactive (CART-IR) cell bodies and varicose CART-IR fibers were observed throughout the RVLM in the SD, WKY, and SHRs. Varicose CART-IR nerve fibers were particularly abundant in the WKY and SHRs. Bilateral microinjection of CARTp (30 pmol) into the RVLM caused a significant increase in mean arterial pressure (MAP) in WKY and SHRs. Bilateral microinjection of CARTp antibody (1:5000) into the RVLM displayed a fall in the basal level of the MAP in SHRs but had no effects in WKY rats. In SD rats, bilateral microinjection of CARTp (6, 30 or 60 pmol) into the RVLM did not change the MAP but attenuated phenylephrine-induced bradycardia in a dose-dependent manner. We propose that CARTp acting in the RVLM may involvement in the cardiovascular regulation either by increases in the blood pressure or by decreases in the baroreflex sensitivity in rats. Moreover, endogenous CARTp in the RVLM is associated with the maintenance of basal blood pressure of SHRs.


Subject(s)
Arterial Pressure , Baroreflex , Medulla Oblongata/physiology , Nerve Tissue Proteins/physiology , Animals , Arterial Pressure/drug effects , Baroreflex/drug effects , Cardiovascular Physiological Phenomena/drug effects , Male , Medulla Oblongata/drug effects , Nerve Tissue Proteins/administration & dosage , Neurons/physiology , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley
14.
Nutrients ; 10(12)2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30513901

ABSTRACT

Nesfatin-1 is a well-established anorexigenic peptide. Recent studies indicated an association between nesfatin-1 and anxiety/depression-like behavior. However, it is unclear whether this effect is retained in obesity. The aim was to investigate the effect of nesfatin-130-59-the active core of nesfatin-1-on anxiety and depression-like behavior in normal weight (NW) and diet-induced (DIO) obese rats. Male rats were intracerebroventricularly (ICV) cannulated and received nesfatin-130-59 (0.1, 0.3, or 0.9 nmol/rat) or vehicle 30 min before testing. Nesfatin-130-59 at a dose of 0.3 nmol reduced sucrose consumption in the sucrose preference test in NW rats compared to vehicle (⁻33%, p < 0.05), indicating depression-like/anhedonic behavior. This dose was used for all following experiments. Nesfatin-130-59 also reduced cookie intake during the novelty-induced hypophagia test (-62%, p < 0.05). Moreover, nesfatin-130-59 reduced the number of entries into the center zone in the open field test (-45%, p < 0.01) and the visits of open arms in the elevated zero maze test (-39%, p < 0.01) in NW rats indicating anxiety. Interestingly, DIO rats showed no behavioral alterations after the injection of nesfatin-130-59 (p > 0.05). These results indicate an implication of nesfatin-130-59 in the mediation of anxiety and depression-like behavior/anhedonia under normal weight conditions, while in DIO rats, a desensitization might occur.


Subject(s)
Anhedonia/drug effects , Anxiety/chemically induced , Calcium-Binding Proteins/adverse effects , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/adverse effects , DNA-Binding Proteins/chemistry , Depression/chemically induced , Nerve Tissue Proteins/adverse effects , Nerve Tissue Proteins/chemistry , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Animals , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Dose-Response Relationship, Drug , Feeding Behavior , Injections, Intraventricular , Male , Nerve Tissue Proteins/administration & dosage , Nucleobindins , Obesity , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley
15.
Peptides ; 109: 14-22, 2018 11.
Article in English | MEDLINE | ID: mdl-30261207

ABSTRACT

Nesfatin-1 is an 82-amino acid protein derived from nucleobindin 2 (NUCB2), which could inhibit food intake in fish and mammals. However, the neuroendocrine mechanism of nesfatin-1 in animal appetite regulation is unclear. To explore the feeding mechanism of nesfatin-1 in Siberian sturgeon (Acipenser baerii), intraperitoneal injections of nesfatin-1 and sulfated cholecystokinin octapeptide (CCK8), Lorglumide (CCK1R selective antagonist), or LY 225,910 (CCK2R selective antagonist) were performed. Co-injection of nesfatin-1 and CCK8 synergistically significantly decreased the food intake in 1 h. Lorglumide reversed the anorectic effect of nesfatin-1, but LY 225,910 had no effect. Moreover, Lorglumide could also reverse the expressions of appetite factors including nucb2, cck, unc3, cart, apelin, pyy, and npy induced by nesfatin-1 in the brain, stomach, and liver, while LY 225,910 partially reversed these changes. These results indicate that nesfatin-1 inhibits the appetite of Siberian sturgeon mainly through the CCK-CCK1R signaling pathway.


Subject(s)
Appetite/drug effects , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Eating/drug effects , Fishes/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Animals , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/pharmacology , Fishes/physiology , Injections, Intraperitoneal , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Nucleobindins
16.
J Mol Neurosci ; 65(3): 377-384, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29956089

ABSTRACT

Nesfatin-1, a recently discovered peptide, is involved in important functions such as food intake regulation and energy homeostasis. Previous studies have demonstrated that it has protective effects following myocardial injury and also protects dopaminergic cells against neurotoxicity with the anti-inflammatory and anti-apoptotic mechanisms. In this study, we aimed to assay the neuroprotective effects of Nesfatin-1 after brain ischemia/reperfusion. Twenty-eight male Wistar rats were randomly selected and allocated in the form of four groups (sham, Nesfatin-1, ischemia, ischemia+Nesfatin-1). Ischemia was created by obstruction couple common carotid arteries in 20-min period. Saline as a vehicle and Nesfatin-1 (20 µg/kg, intraperitoneally) were injected at the time of reperfusion. Spatial memory performances were evaluated by the Morris water maze. The level of protein expression was determined by immunohistochemical and immunofluorescence staining. Nesfatin-1 significantly reduced caspase-3 (P < 0.01) and microglial activation (P < 0.01) and improved spatial memory impairments (P < 0.05) induced by brain ischemia. Nesfatin-1 has significant neuroprotective effects and can be introduced as a therapeutic agent against cerebral ischemia-induced injuries.


Subject(s)
Brain Ischemia/drug therapy , Calcium-Binding Proteins/therapeutic use , DNA-Binding Proteins/therapeutic use , Memory , Microglia/drug effects , Nerve Tissue Proteins/therapeutic use , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/pharmacology , Caspase 3/metabolism , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/pharmacology , Male , Microglia/metabolism , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Nucleobindins , Rats , Rats, Wistar
17.
Neuropeptides ; 70: 9-15, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29751997

ABSTRACT

Nesfatin-1, a peptide whose receptor is yet to be identified, has been shown to be involved in the modulation of feeding, stress, and metabolic responses. Recently, increasing evidence has supported a modulatory role of nesfatin-1 in cardiovascular activity. We have previously reported that nesfatin-1 causes an increase in blood pressure in normotensive and hypotensive rats by increasing plasma catecholamine, vasopressin, and renin levels. Recent reports suggest that nesfatin-1 may activate the central cholinergic system. However, there is no evidence showing an interaction between central nesfatin-1 and the cholinergic system. Therefore, this study aimed to determine whether the central cholinergic system may have a functional role in the nesfatin-1-induced cardiovascular effect observed in normotensive rats. Intracerebroventricular injection of nesfatin-1 caused short-term increases in mean arterial pressure and heart rate responses including bradycardic/tachycardic phases in normotensive animals. Central injection of nesfatin-1 increased the acetylcholine and choline levels in the posterior hypothalamus, as shown in microdialysis studies. Central pretreatment with the cholinergic muscarinic receptor antagonist atropine and/or nicotinic receptor antagonist mecamylamine blocked nesfatin-1-induced cardiovascular effects. In conclusion, the results show that centrally administered nesfatin-1 produces a pressor effect on blood pressure and heart rate responses including bradycardic/tachycardic phases in normotensive rats. Moreover, according to our findings, the central cholinergic system can modulate nesfatin-1-evoked cardiovascular activity.


Subject(s)
Blood Pressure/drug effects , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Hypotension/etiology , Nerve Tissue Proteins/pharmacology , Vasoconstrictor Agents/pharmacology , Acetylcholine/metabolism , Animals , Brain/drug effects , Calcium-Binding Proteins/administration & dosage , Catecholamines/metabolism , Cholinergic Agents/pharmacology , DNA-Binding Proteins/administration & dosage , Heart Rate/drug effects , Male , Mecamylamine/blood , Nerve Tissue Proteins/administration & dosage , Nucleobindins , Rats, Sprague-Dawley , Vasopressins/blood
18.
Neuropeptides ; 69: 53-59, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29705514

ABSTRACT

By screening for neuropeptides in the mouse spinal cord using mass spectrometry (MS), we have previously demonstrated that one of the 78 peptides that is expressed predominantly (> 6-fold) in the dorsal horn compared to the ventral spinal cord is the atypical peptide desCER [des-Ser1]-cerebellin, which originates from the precursor protein cerebellin 1 (CBLN1). Furthermore, we found that intrathecal injection of desCER induces mechanical hypersensitivity in a dose dependent manner. The current study was designed to further investigate the relative expression of other CBLN derived peptides in the spinal cord and to examine whether they share similar nociceptive properties. In addition to the peptides cerebellin (CER) and desCER we identified and relatively quantified nine novel peptides originating from cerebellin precursor proteins CBLN1 (two peptides), CBLN2 (three peptides) and CBLN4 (four peptides). Ten out of eleven peptides displayed statistically significantly (p < 0.05) higher expression levels (200-350%) in the dorsal horn compared to the ventral horn. Intrathecal injection of three of the four CBLN1 and two of the three CBLN2 derived peptides induced mechanical hypersensitivity in response to von Frey filament testing in mice during the first 6 h post-injection compared to saline injected mice, while none of the four CBLN4 derived peptides altered withdrawal thresholds. This study demonstrates that high performance MS is an effective tool for detecting novel neuropeptides in CNS tissues. We show the presence of nine novel atypical peptides originating from CBLN1, CBLN2 and CBLN4 precursor proteins in the mouse dorsal horn, whereof five peptides induce pain-like behavior upon intrathecal injection. Further studies are required to investigate the mechanisms by which CBLN1 and CBLN2 derived peptides facilitate nociceptive signal transmission.


Subject(s)
Nerve Tissue Proteins/physiology , Nociception/physiology , Pain Threshold , Spinal Cord/physiopathology , Animals , Injections, Spinal , Male , Mass Spectrometry , Mice, Inbred C57BL , Nerve Tissue Proteins/administration & dosage , Neuropeptides/administration & dosage , Neuropeptides/isolation & purification , Nociception/drug effects , Physical Stimulation , Spinal Cord/drug effects
19.
Sci Rep ; 8(1): 6184, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29670152

ABSTRACT

Patients and rodents with cerebellar damage display ataxic gaits characterized by impaired coordination of limb movements. Here, gait ataxia in mice with a null mutation of the gene for the cerebellin 1 precursor protein (cbln1-null mice) was investigated by kinematic analysis of hindlimb movements during locomotion. The Cbln1 protein is predominately produced and secreted from cerebellar granule cells. The cerebellum of cbln1-null mice is characterized by an 80% reduction in the number of parallel fiber-Purkinje cell synapses compared with wild-type mice. Our analyses identified prominent differences in the temporal parameters of locomotion between cbln1-null and wild-type mice. The cbln1-null mice displayed abnormal hindlimb movements that were characterized by excessive toe elevation during the swing phase, and by severe hyperflexion of the ankles and knees. When recombinant Cbln1 protein was injected into the cerebellum of cbln1-null mice, the step cycle and stance phase durations increased toward those of wild-type mice, and the angular excursions of the knee during a cycle period showed a much closer agreement with those of wild-type mice. These findings suggest that dysfunction of the parallel fiber-Purkinje cell synapses might underlie the impairment of hindlimb movements during locomotion in cbln1-null mice.


Subject(s)
Cerebellar Ataxia/physiopathology , Cerebellum/drug effects , Cerebellum/physiopathology , Gait/drug effects , Nerve Tissue Proteins/administration & dosage , Protein Precursors/administration & dosage , Animals , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/etiology , Cerebellum/metabolism , Disease Models, Animal , Injections , Locomotion/drug effects , Mice , Mice, Knockout , Phenotype , Treatment Outcome
20.
Neuroscience ; 376: 204-223, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29555037

ABSTRACT

Collapsin Response Mediator Protein 2 (CRMP2) is an intracellular protein involved in axon and dendrite growth and specification. In this study, CRMP2 was identified in a conditioned media derived from degenerated sciatic nerves (CM). On cultured rat hippocampal neurons, acute extracellular application of CM or partially purified recombinant CRMP2 produced an increase in cytoplasmic calcium. The increase in cytoplasmic calcium was mostly mediated through NMDA receptors, with a minor contribution of N-type VDCC, and it was maintained as long as CM was present. By using live-labeling of CRMP2, Ca2+ channel binding domain 3 (CBD3) peptide derived from CRMP2, and recombinant CRMP2, we demonstrated that that this effect was mediated by an action on the extracellular side of the NMDA receptor. This is the first report of an extracellular action of CRMP2. Prolonged exposure to extracellular CRMP2, may contribute to neuronal calcium dysregulation and neuronal damage.


Subject(s)
Calcium/metabolism , Central Nervous System Agents/administration & dosage , Cytoplasm/drug effects , Intercellular Signaling Peptides and Proteins/administration & dosage , Nerve Tissue Proteins/administration & dosage , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cations, Divalent/metabolism , Cells, Cultured , Central Nervous System Agents/isolation & purification , Culture Media, Conditioned , Cytoplasm/metabolism , Extracellular Space , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Molecular Docking Simulation , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Optic Nerve/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Sciatic Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...