Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 395
Filter
Add more filters

Publication year range
1.
Gen Physiol Biophys ; 43(5): 457-467, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140686

ABSTRACT

In this study, we investigated the effects of peripheral nesfatin-1 on basal brain activity and 4-aminopyridine (4-AP)-induced epileptiform activity, and its relationship with the electrocorticogram (ECoG) power spectrum and EEG bands. Forty-nine male Wistar rats were divided into seven groups: control sham, 4-AP (2.5 mg/kg i.p.), Nesfatin-1 (1, 2, and 4 µg/kg i.p.), Nesfatin-1 (2 µg/kg) post-treatment, and Nesfatin-1 (2 µg/kg) pre-treatment. Recordings were conducted for 70 min under ketamine/xylazine (90/10 mg/kg) anesthesia. In the post-treatment group, nesfatin-1 was injected 20 min after 4-AP induction. In the pre-treatment groups, nesfatin-1 was administered following basal recordings and before 4-AP injection. 4-AP induced epileptiform activity in all animals, peaking at 30 min. Nesfatin-1 (2 µg/kg) reduced basal brain activity (p < 0.05) and decreased alpha, delta, and theta bands in ECoG. Post-treatment of nesfatin-1 did not affect 4-AP-induced activity (p > 0.05) but increased gamma band activity (p > 0.05). Pre-treatment of nesfatin-1 reduced epileptiform activity between 50 and 60 min (p < 0.05), decreased delta bands, and increased gamma bands (p > 0.05). We conclude that peripheral nesfatin-1 modulates normal brain activity but has limited effects on abnormal discharges.


Subject(s)
Brain , Epilepsy , Nucleobindins , Rats, Wistar , Animals , Male , Rats , Epilepsy/physiopathology , Epilepsy/chemically induced , Epilepsy/blood , Brain/drug effects , Brain/metabolism , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/administration & dosage , Electroencephalography , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Treatment Outcome , Anticonvulsants/pharmacology , Anticonvulsants/administration & dosage
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.
Gen Comp Endocrinol ; 265: 71-76, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29155267

ABSTRACT

Recently we discovered a small hypothalamic protein in the chicken, named neurosecretory protein GL (NPGL), which is associated with body growth and energy metabolism in birds and rodents. Genome database analysis suggested that the NPGL gene has a paralogous gene in vertebrates, named neurosecretory protein GM (NPGM). However, the biological action of NPGM remains unclear. In this study, we investigated whether NPGM affects body growth in chicks. We found that subcutaneous infusion of NPGM for six days increased body mass gain in a dose-dependent manner. Despite the observed increase in body mass, infusion of NPGM did not alter food and water intake. Of note, we observed tendency of mass increase of several peripheral tissues, specifically. When we compared several tissue types, NPGM seemed to induce the largest growth increase in white adipose tissue mass. These results suggest that NPGM may accelerate fat accumulation and body growth. In addition, we analyzed whether NPGM increases body growth through the action of pituitary hormones. However, we observed no significant changes in mRNA expression of pituitary hormones or plasma levels of growth hormone in NPGM-treated chicks. This is the first report describing the biological action of NPGM in vertebrates.


Subject(s)
Chickens/growth & development , Nerve Tissue Proteins/administration & dosage , Weight Gain , Amino Acid Sequence , Animals , Body Composition/drug effects , Chickens/metabolism , Drinking/drug effects , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Hormones/genetics , Hormones/metabolism , Infusions, Subcutaneous , Male , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Pituitary Gland/metabolism
5.
Gen Comp Endocrinol ; 256: 37-42, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28554734

ABSTRACT

Recently, we discovered a novel cDNA encoding the precursor of a small secretory protein, neurosecretory protein GL (NPGL), in the chicken mediobasal hypothalamus. In this study, immunohistochemical analysis revealed that NPGL was produced in the infundibular and medial mammillary nuclei of the mediobasal hypothalamus, with immunoreactive fibers also detected in the hypothalamus and the median eminence. As it is known that these regions are involved in feeding behavior in chicks, we surveyed the effects of chronic intracerebroventricular infusion of NPGL on feeding behavior and body mass for a period of two weeks. NPGL stimulated food and water intake, with a concomitant increase in body mass. However, NPGL did not influence mRNA expression of several hypothalamic ingestion-related neuropeptides. Our data suggest that NPGL may be a novel neuronal regulator involved in growth processes in chicks.


Subject(s)
Body Weight , Chickens/metabolism , Drinking , Feeding Behavior/physiology , Infusions, Intraventricular , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Animals , Body Weight/drug effects , DNA, Complementary/metabolism , Drinking/drug effects , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Biochem Biophys Res Commun ; 485(2): 409-413, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28213131

ABSTRACT

Neuroendocrine regulatory peptide (NERP)-2 is derived from a distinct region of VGF, a neurosecretory protein originally identified as a product of a nerve growth factor-responsive gene in rat PC12 cells. Colocalization of NERP-2 with orexin-A in the lateral hypothalamus increases orexin-A-induced feeding and energy expenditure in both rats and mice. Orexigenic and anorectic peptides in the hypothalamus modulate gastric function. In this study, we investigated the effect of NERP-2 on gastric function in rats. Intracerebroventricular administration of NERP-2 to rats increased gastric acid secretion and gastric emptying, whereas peripheral administration did not affect gastric function. NERP-2-induced gastric acid secretion and gastric emptying were blocked by an orexin 1 receptor antagonist, SB334867. NERP-2 also induced Fos expression in the lateral hypothalamus and the dorsomotor nucleus of the vagus X, which are key sites in the central nervous system for regulation of gastric function. Atropine, a blocker of vagal efferent signal transduction, completely blocked NERP-2-induced gastric acid secretion. These results demonstrate that central administration of NERP-2 activates the orexin pathway, resulting in elevated gastric acid secretion and gastric emptying.


Subject(s)
Gastric Acid/metabolism , Gastric Emptying/drug effects , Nerve Tissue Proteins/pharmacology , Orexin Receptors/metabolism , Signal Transduction/drug effects , Animals , Atropine/pharmacology , Benzoxazoles/pharmacology , Injections, Intraventricular , Male , Naphthyridines , Nerve Tissue Proteins/administration & dosage , Parasympatholytics/pharmacology , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Urea/analogs & derivatives , Urea/pharmacology , Vagus Nerve/drug effects , Vagus Nerve/physiology
7.
Clin Exp Rheumatol ; 35(5): 735-738, 2017.
Article in English | MEDLINE | ID: mdl-28850023

ABSTRACT

OBJECTIVES: Pentraxin 3 (PTX3) is a multifunctional soluble factor. PTX3 can be involved in the regulation of vasculitis and is expressed in the cytoplasm of neutrophils. As anti-neutrophil cytoplasmic antibody (ANCA) is recognised as a cause of vasculitis, we aimed to discover the role of PTX3 in ANCA production in vivo. METHODS: To this end, we used aluminum salt (alum), which induces neutrophil extracellular traps, as an adjuvant for producing anti-myeloperoxidase-ANCA (MPO-ANCA). Specifically, we intraperitoneally injected alum and recombinant MPO (rMPO) into MPO-deficient mice and then measured the concentration of anti-MPO IgG in their blood. To show the involvement of extracellular PTX3 in this model, we assessed PTX3 protein content and host double-stranded DNA levels in the mice's peritoneal fluid after alum injection. In addition, we simultaneously administered recombinant PTX3, rMPO and alum to MPO-deficient mice to assess the function of PTX3 in producing anti-MPO IgG in vivo. RESULTS: Anti-MPO IgG was produced by the alum + rMPO immunisation model in MPO-deficient but not wildtype mice. Injection of alum induced extracellular PTX3 as well as double-stranded DNA and dead cells in MPO-deficient mice. Simultaneous injection of recombinant PTX3 with rMPO and alum attenuated the production of anti-MPO IgG in MPO-deficient mice. CONCLUSIONS: Our current findings provide evidence that PTX3 attenuates the production of murine MPO-ANCA.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Antibodies, Antineutrophil Cytoplasmic/blood , C-Reactive Protein/immunology , Immunoglobulin G/blood , Metabolism, Inborn Errors/immunology , Nerve Tissue Proteins/immunology , Peroxidase/immunology , Animals , Ascitic Fluid/immunology , Ascitic Fluid/metabolism , C-Reactive Protein/administration & dosage , C-Reactive Protein/metabolism , DNA/immunology , DNA/metabolism , Extracellular Traps/immunology , Extracellular Traps/metabolism , Female , Male , Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/enzymology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/metabolism , Peroxidase/administration & dosage , Peroxidase/deficiency , Peroxidase/genetics
8.
Brain ; 139(Pt 2): 365-79, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582558

ABSTRACT

Stiff-person syndrome is the prototype of a central nervous system disorder with autoantibodies targeting presynaptic antigens. Patients with paraneoplastic stiff-person syndrome may harbour autoantibodies to the BAR (Bin/Amphiphysin/Rvs) domain protein amphiphysin, which target its SH3 domain. These patients have neurophysiological signs of compromised central inhibition and respond to symptomatic treatment with medication enhancing GABAergic transmission. High frequency neurotransmission as observed in tonic GABAergic interneurons relies on fast exocytosis of neurotransmitters based on compensatory endocytosis. As amphiphysin is involved in clathrin-mediated endocytosis, patient autoantibodies are supposed to interfere with this function, leading to disinhibition by reduction of GABAergic neurotransmission. We here investigated the effects of human anti-amphiphysin autoantibodies on structural components of presynaptic boutons ex vivo and in vitro using electron microscopy and super-resolution direct stochastic optical reconstruction microscopy. Ultrastructural analysis of spinal cord presynaptic boutons was performed after in vivo intrathecal passive transfer of affinity-purified human anti-amphiphysin autoantibodies in rats and revealed signs of markedly disabled clathrin-mediated endocytosis. This was unmasked at high synaptic activity and characterized by a reduction of the presynaptic vesicle pool, clathrin coated intermediates, and endosome-like structures. Super-resolution microscopy of inhibitory GABAergic presynaptic boutons in primary neurons revealed that specific human anti-amphiphysin immunoglobulin G induced an increase of the essential vesicular protein synaptobrevin 2 and a reduction of synaptobrevin 7. This constellation suggests depletion of resting pool vesicles and trapping of releasable pool vesicular proteins at the plasma membrane. Similar effects were found in amphiphysin-deficient neurons from knockout mice. Application of specific patient antibodies did not show additional effects. Blocking alternative pathways of clathrin-independent endocytosis with brefeldin A reversed the autoantibody induced effects on molecular vesicle composition. Endophilin as an interaction partner of amphiphysin showed reduced clustering within presynaptic terminals. Collectively, these results point towards an autoantibody-induced structural disorganization in GABAergic synapses with profound changes in presynaptic vesicle pools, activation of alternative endocytic pathways, and potentially compensatory rearrangement of proteins involved in clathrin-mediated endocytosis. Our findings provide novel insights into synaptic pathomechanisms in a prototypic antibody-mediated central nervous system disease, which may serve as a proof-of-principle example in this evolving group of autoimmune disorders associated with autoantibodies to synaptic antigens.


Subject(s)
Autoantibodies/administration & dosage , Nerve Tissue Proteins/administration & dosage , Presynaptic Terminals/ultrastructure , Synaptic Vesicles/ultrastructure , Animals , Autoantibodies/blood , Cells, Cultured , Female , Humans , Injections, Spinal , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/blood , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Pregnancy , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Rats, Inbred Lew , Stiff-Person Syndrome/blood , Stiff-Person Syndrome/diagnosis , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism
9.
Neurobiol Dis ; 85: 164-173, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26550694

ABSTRACT

BACKGROUND AND PURPOSE: Peripheral immune cell infiltration to the brain tissue at the perisurgical site can promote neuroinflammation after surgical brain injury (SBI). Slit2, an extracellular matrix protein, has been reported to reduce leukocyte migration. This study evaluated the effect of recombinant Slit2 and the role of its receptor roundabout1 (Robo1) and its downstream mediator Slit-Robo GTPase activating protein 1 (srGAP1)-Cdc42 on peripheral immune cell infiltration after SBI in a rat model. METHODS: One hundred and fifty-three adult male Sprague-Dawley rats (280-350 g) were used. Partial resection of right frontal lobe was performed to induce SBI. Slit2 siRNA was administered by intracerebroventricular injection 24h before SBI. Recombinant Slit2 was injected intraperitoneally 1h before SBI. Recombinant Robo1 used as a decoy receptor was co-administered with recombinant Slit2. srGAP1 siRNA was administered by intracerebroventricular injection 24h before SBI. Post-assessments included brain water content measurement, neurological tests, ELISA, Western blot, immunohistochemistry, and Cdc42 activity assay. RESULTS: Endogenous Slit2 was increased after SBI. Robo1 was expressed by peripheral immune cells. Endogenous Slit2 knockdown worsened brain edema after SBI. Recombinant Slit2 administration reduced brain edema, neurological deficits, and pro-inflammatory cytokines after SBI. Recombinant Slit2 reduced peripheral immune cell markers cluster of differentiation 45 (CD45) and myeloperoxidase (MPO), as well as Cdc42 activity in the perisurgical brain tissue which was reversed by recombinant Robo1 co-administration and srGAP1 siRNA. CONCLUSIONS: Recombinant Slit2 improved outcomes by reducing neuroinflammation after SBI, possibly by decreasing peripheral immune cell infiltration to the perisurgical site through Robo1-srGAP1 mediated inhibition of Cdc42 activity. These results suggest that Slit2 may be beneficial to reduce SBI-induced neuroinflammation.


Subject(s)
Brain Injuries/immunology , Frontal Lobe/immunology , Frontal Lobe/injuries , Intercellular Signaling Peptides and Proteins/administration & dosage , Intraoperative Complications/immunology , Nerve Tissue Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Animals , Brain Edema/etiology , Brain Edema/immunology , Brain Edema/therapy , Brain Injuries/etiology , Brain Injuries/therapy , Disease Models, Animal , Frontal Lobe/surgery , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Knockdown Techniques , Genetic Therapy/methods , Infusions, Intraventricular , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intraoperative Complications/therapy , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats, Sprague-Dawley , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Proteins/genetics , Roundabout Proteins
10.
Hippocampus ; 26(10): 1313-27, 2016 10.
Article in English | MEDLINE | ID: mdl-27258934

ABSTRACT

Although cocaine- and amphetamine-regulated transcript peptide (CART) is detected in several cortical and subcortical areas, its role in higher functions has been largely ignored. We examined the significance of CART in memory formation and tested if the downstream actions of CART involve N-methyl-d-aspartate (NMDA) activated extra-cellular signal-regulated kinase (ERK). Newly formed memory was evaluated using novel object recognition test consisting of familiarization (T1) and choice trials (T2). The choice trials were performed at two time points: 30-min (T230-min ) and 24-h (T224-h ) postacquisition. In choice trial (T230-min ), vehicle control rats explored the novel object for significantly longer duration than the familiar object indicating intact memory formation. However, CART-antibody, U0126 [ERK antagonist, both via intracerebroventricular (icv) or intrahippocampal (ih) route] or MK-801 (NMDA antagonist; intraperitoneal) treated rats spent less time exploring novel objects; CART peptide (icv or ih) was ineffective. During choice trial at T224-h , a significant decrease in novel object exploration time was noticed in vehicle control rats suggesting amnesia. However, treatment with CART, prior to familiarization trial (T1), promoted exploration of the novel object even at T224-h . Pretreatment with U0126 or MK-801 blocked pro-cognitive-like effect of CART suggesting involvement of NMDA-ERK pathway in CART's action. Animals subjected to the object familiarization trial showed a drastic increase in the CART-immunoreactivity in the cells of cornu ammonis 3 and polymorph layer of dentate gyrus, and fibers within ento- (ENT) and peri-rhinal (PRH) cortices. Western blot analysis revealed that CART treatment significantly up-regulated the expression of phospo-ERK1/2 in hippocampus, ENT and PRH. This effect was attenuated following pretreatment with U0126 or MK-801, suggesting the activation of ERK signaling cascade through NMDA receptors. Thus, CART system seems to play an important role in recognition memory and that these effects may be mediated by NMDA receptors-ERK signaling in the ENT/PRH-hippocampal circuit. © 2016 Wiley Periodicals, Inc.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Recognition, Psychology/physiology , Animals , Butadienes/pharmacology , Choice Behavior/drug effects , Choice Behavior/physiology , Cognition/drug effects , Cognition/physiology , Dizocilpine Maleate/pharmacology , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hippocampus/drug effects , Learning/drug effects , Learning/physiology , Male , Nerve Tissue Proteins/administration & dosage , Neurons/drug effects , Neurons/physiology , Neuropsychological Tests , Nitriles/pharmacology , Perirhinal Cortex/drug effects , Perirhinal Cortex/metabolism , Psychotropic Drugs/pharmacology , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Recognition, Psychology/drug effects
11.
Biochem Biophys Res Commun ; 472(1): 201-8, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26920055

ABSTRACT

Nesfatin-1 is an 82 amino acid secreted peptide encoded in the precursor, nucleobindin-2 (NUCB2). It is an insulinotropic anorexigen abundantly expressed in the stomach and hypothalamus. Post-prandial insulin secretion is predominantly regulated by incretins glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). Nesfatin-1 was previously reported to modulate GLP-1 and GIP secretion in vitro in an enteroendocrine (STC-1) cell line. Intestine is a source of additional hormones including cholecystokinin (CCK) and peptide YY (PYY) that regulate metabolism. We hypothesized that nesfatin-1 modulates CCK and PYY secretion. Immunofluorescence histochemistry showed NUCB2/nesfatin-1 co-localizing CCK and PYY in the intestinal mucosa of mice. Static incubation of STC-1 cells with nesfatin-1 upregulated both CCK mRNA expression (1 and 10 nM) and secretion (0.1, 1 and 10 nM) at 1 h post-incubation. In contrast, nesfatin-1 treatment for 1 h downregulated PYY mRNA expression (all doses tested) and secretion (0.01 and 0.1 nM) in STC-1 cells. Continuous infusion of nesfatin-1 using osmotic mini-pumps for 12 h upregulated CCK mRNA expression in large intestine, and downregulated PYY mRNA expression in both large and small intestines of male C57BL/6J mice. In these tissues, Western blot analysis found a corresponding increase in CCK and a decrease in PYY content. Collectively, we provide new information on the cell specific localization of NUCB2/nesfatin-1 in the intestinal mucosa, and a novel function for nesfatin-1 in modulating intestinal CCK and PYY expression and secretion in mice.


Subject(s)
Calcium-Binding Proteins/physiology , Cholecystokinin/genetics , Cholecystokinin/metabolism , DNA-Binding Proteins/physiology , Intestinal Mucosa/metabolism , Nerve Tissue Proteins/physiology , Peptide YY/genetics , Peptide YY/metabolism , Animals , Calcium-Binding Proteins/administration & dosage , Cell Line , DNA-Binding Proteins/administration & dosage , Down-Regulation , Immunohistochemistry , Infusions, Subcutaneous , Intestine, Large/metabolism , Intestine, Small/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/administration & dosage , Nucleobindins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
12.
Crit Care Med ; 44(3): e146-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26491860

ABSTRACT

OBJECTIVE: Leucine-rich repeat and immunoglobulin domain-containing protein (LINGO)-1 is expressed in neural stem cells, and its neutralization results in sustained neuronal immaturity. Thus, targeted inhibition of LINGO-1 via RNA interference may enhance transplanted neural stem cell survival and neuronal differentiation in vivo. Furthermore, LINGO-1 RNA interference in neural stem cells represents a potential therapeutic strategy for spinal cord injury. DESIGN: Department of Spine Surgery, First Affiliated Hospital of Sun Yat-sen University. SETTING: Translational Medicine Center Research Laboratory, First Affiliated Hospital of Sun Yat-sen University. SUBJECTS: Female Sprague-Dawley rats. INTERVENTIONS: The animals were divided into three groups that underwent laminectomy and complete spinal cord transection accompanied by transplantation of control-RNA interference-treated or LINGO-1-RNA interference-treated neural stem cells at the injured site in vivo. In vitro, neural stem cells were divided into four groups for the following treatments: control, control RNA interference lentivirus, LINGO-1 RNA interference lentivirus and LINGO-1 complementary DNA lentivirusand the Key Projects of the Natural Science Foundation of Guangdong Province (No. S2013020012818). MEASUREMENTS AND MAIN RESULTS: Neural stem cells in each treatment group were examined for cell survival and neuronal differentiation in vitro and in vivo via immunofluorescence and Western blot analysis. Axonal regeneration and tissue repair were assessed via retrograde tracing using Fluorogold, electron microscopy, hematoxylin-eosin staining and MRI. Rats were also examined for functional recovery based on the measurement of spinal cord-evoked potentials and the Basso-Beattie-Bresnahan score. LINGO-1-RNA interference-treated neural stem cell transplantation increased tissue repair and functional recovery of the injured spinal cord in rats. Similarly, LINGO-1 RNA interference increased neural stem cell survival and neuronal differentiation in vitro. The mechanism underlying the effect of LINGO-1 RNA interference on the injured rat spinal cord may be that the significant inhibition of LINGO-1 expression in neural stem cells inactivated the RhoA and Notch signaling pathways, which act downstream of LINGO-1. CONCLUSIONS: Our findings indicate that transplantation of LINGO-1-RNA interference-treated neural stem cells facilitates functional recovery after spinal cord injury and represents a promising potential strategy for the repair of spinal cord injury.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/administration & dosage , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation/methods , Animals , Female , Genetic Vectors , Injections, Spinal , Laminectomy , Lentivirus/genetics , Nerve Regeneration , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , RNA Interference , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord/physiology
13.
NMR Biomed ; 29(3): 361-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26915794

ABSTRACT

Recent studies have shown that post-treatment with cocaine- and amphetamine-regulated transcript (CART) has neuroregenerative effects in animal models of stroke. The purpose of this study was to characterize CART-mediated neuronal and vascular repairments using non-invasive MRI techniques. Adult male rats were subjected to a 90 min middle cerebral artery occlusion (MCAo). Animals were separated into two groups with similar infarction sizes, measured by T2 -weighted MRI on Day 2 after MCAo, and were treated with CART or vehicle intranasally from Day 3 to Day 12. Diffusion tensor imaging was used to examine changes in plasticity of white matter elements. Susceptibility-weighted imaging (SWI) was used to measure angiogenesis. Post-treatment with CART significantly increased fractional anisotropy (FA) in lesioned cortex on Days 10 and 25 post stroke. A significant correlation between the behavioral recovery in body asymmetry and the change in FA was shown, suggesting that behavioral recovery was associated with reinnervation to the lesioned hemisphere. CART also increased the intensity of SWI and the immunoreactivity of the vascular marker alpha-smooth muscle actin in lesioned cortex. Together, our data support a non-invasive treatment strategy for stroke through angiogenesis and reinnervation by CART. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Infarction/complications , Magnetic Resonance Imaging/methods , Neovascularization, Physiologic , Nerve Tissue Proteins/therapeutic use , Stroke/drug therapy , Stroke/pathology , Animals , Anisotropy , Cerebral Cortex/pathology , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Male , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Rats, Sprague-Dawley , Stroke/complications
14.
Neurochem Res ; 41(6): 1375-80, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26846142

ABSTRACT

The present study was designed to construct a recombinant adeno-associated virus (rAAV) which can express NAP in the brain and examine whether this virus can produce antidepressant effects on C57 BL/6 mice that had been subjected to open field test and forced swimming test, via nose-to-brain pathway. When the recombinant plasmid pGEM-T Easy/NT4-NAP was digested by EcoRI, 297 bp fragments can be obtained and NT4-NAP sequence was consistent with the designed sequence confirmed by DNA sequencing. When the recombinant plasmid pSSCMV/NT4-NAP was digested by EcoRI, 297 bp fragments is visible. Immunohistochemical staining of fibroblasts revealed that expression of NAP was detected in NT4-NAP/AAV group. Intranasal delivery of NT4-NAP/AAV significantly reduced immobility time when the FST was performed after 1 day from the last administration. The effects observed in the FST could not be attributed to non-specific increases in activity since intranasal delivery of NT4-NAP/AAV did not alter the behavior of the mice during the open field test. The results indicated that a recombinant AAV vector which could express NAP in cells was successfully constructed and NAP may be a potential target for therapeutic action of antidepressant treatment.


Subject(s)
Antidepressive Agents/administration & dosage , Dependovirus , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Nerve Growth Factors/administration & dosage , Peptide Fragments/administration & dosage , Administration, Intranasal , Animals , Base Sequence , Dependovirus/genetics , Depression/drug therapy , Depression/genetics , Depression/psychology , Female , Genetic Vectors/genetics , HEK293 Cells , Homeodomain Proteins/administration & dosage , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Peptide Fragments/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Treatment Outcome
15.
Br J Clin Pharmacol ; 82(1): 108-17, 2016 07.
Article in English | MEDLINE | ID: mdl-27016000

ABSTRACT

AIMS: BG00010 is a protein in the glial cell line-derived neurotrophic factor (GDNF) family. It is a selective ligand for the GDNF family receptor alpha-3 (GFRα3) co-receptor that normalizes cellular changes resulting from damage or disease, and potentially alleviates neuropathic pain. The main objectives of this study were to evaluate the pharmacokinetic and safety profiles and to determine the effects on pain of ascending doses of intravenous injections of BG00010 in patients with sciatica. METHODS: This was a randomized, blinded, placebo-controlled multiple-dose study in subjects with sciatica. In Part I (16 patients), four IV dose levels were examined (50, 150, 400, 800 µg kg(-1) ) and in Part II (12 patients), three dose levels were examined (400, 600 and 1200 µg kg(-1) ). Safety and efficacy assessments were used as endpoints. RESULTS: The BG00010 concentration-time data indicated relatively low inter-patient variability and there was a dose-dependent (not dose-proportional) increase in serum exposure from 150 to 1200 µg kg(-1) . The effective half-life was between 40 and 60 h. The most frequently occurring adverse events (AEs) reported by patients receiving BG00010 were headache (67-83%), feeling hot (50-100%), and pruritus (42-67%). Most AEs were mild; no serious AEs or AEs leading to discontinuation occurred. Higher dose regimens of BG00010 resulted in greater pain reduction than placebo or lower dose regimens, although a clear dose-response relationship was not seen. CONCLUSIONS: The pharmacokinetic profile of BG00010 was characterized by low intra-patient variability. These data from a small sample suggest that BG00010 may have a benefit for patients with sciatica.


Subject(s)
Analgesics/administration & dosage , Hyperalgesia/drug therapy , Nerve Tissue Proteins/administration & dosage , Sciatica/drug therapy , Adult , Aged , Analgesics/pharmacokinetics , Analgesics/pharmacology , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Injections, Intravenous , Male , Middle Aged , Nerve Tissue Proteins/pharmacokinetics , Nerve Tissue Proteins/pharmacology , Treatment Outcome , Young Adult
16.
Pharm Res ; 33(6): 1383-98, 2016 06.
Article in English | MEDLINE | ID: mdl-26908049

ABSTRACT

PURPOSE: Oligosaccharides play diverse and unpredictable functional roles when attached to proteins and are a largely unexplored scaffold for deconstructing and attributing novel functions to proteins during drug development. Here, the glycoprotein Artemin (ART) was carefully assessed by multiple analytical methods that allow us to provide a comprehensive understanding of how N-linked glycosylation impact the structural and functional properties of ART. METHODS: Modification of the N-linked glycan of ART was performed by incubation with various enzymes. Biological assays and systems were used to examine the relative activity and pharmacokinetic properties of ART as a function of glycosylation. In order to reveal the conformational impact of glycosylation on ART, hydrogen/deuterium exchange mass spectrometry (HDX-MS) was employed in addition to differential scanning calorimetry. The colloidal stability of ART glycovariants was assessed by dynamic light scattering, viscometry, and solubility assays. RESULTS: No difference in pharmacokinetics or relative potency was revealed between glycosylated and nonglycosylated ART. Surprisingly, the HDX-MS data indicated that the glycan does not greatly influence the conformation and dynamics of the protein. In contrast, differences in thermal and colloidal stability clearly revealed a role of glycosylation in increasing the solubility and stability of ART. CONCLUSIONS: Our findings demonstrate how careful analysis using multiple advanced techniques can be used to identify and dissect the multiple potential functions of protein glycosylation and form a prerequisite for glycoengineering and drug development of glycoproteins.


Subject(s)
Nerve Tissue Proteins/chemistry , Protein Processing, Post-Translational , Animals , Colloids , Drug Stability , Dynamic Light Scattering , Glycosylation , Injections, Intravenous , Male , Models, Molecular , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacokinetics , Protein Conformation , Protein Stability , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Solubility , Structure-Activity Relationship , Temperature , Viscosity
17.
Stroke ; 46(2): 529-36, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25550365

ABSTRACT

BACKGROUND AND PURPOSE: Norrin and its receptor Frizzled-4 have important roles in the blood-brain barrier development. This study is to investigate a potential role and mechanism of Norrin/Frizzled-4 on protecting blood-brain barrier integrity after subarachnoid hemorrhage (SAH). METHODS: One hundred and seventy-eight male Sprague-Dawley rats were used. SAH model was induced by endovascular perforation. Frizzled-4 small interfering RNA was injected intracerebroventricularly 48 hours before SAH. Norrin was administrated intracerebroventricularly 3 hours after SAH. SAH grade, neurological scores, brain water content, Evans blue extravasation, western blots, and immunofluorescence were used to study the mechanisms of Norrin and its receptor regulation protein TSPAN12, as well as neurological outcome. RESULTS: Endogenous Norrin and TSPAN12 expression were increased after SAH, and Norrin was colocalized with astrocytes marker glial fibrillary acidic protein in cortex. Exogenous Norrin treatment significantly alleviated neurobehavioral dysfunction, reduced brain water content and Evans blue extravasation, promoted ß-catenin nuclear translocation, and increased Occludin, VE-Cadherin, and ZO-1 expressions. These effects were abolished by Frizzled-4 small interfering RNA pretreated before SAH. CONCLUSIONS: Norrin protected blood-brain barrier integrity and improved neurological outcome after SAH, and the action of Norrin appeared mediated by Frizzled-4 receptor activation, which promoted ß-catenin nuclear translocation, which then enhanced Occludin, VE-Cadherin, and ZO-1 expression. Norrin might have potential to protect blood-brain barrier after SAH.


Subject(s)
Blood-Brain Barrier/metabolism , Eye Proteins/biosynthesis , Frizzled Receptors/biosynthesis , Nerve Tissue Proteins/biosynthesis , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/prevention & control , beta Catenin/biosynthesis , Animals , Blood-Brain Barrier/drug effects , Eye Proteins/administration & dosage , Injections, Intraventricular , Male , Nerve Tissue Proteins/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley
18.
Eur J Neurosci ; 41(10): 1372-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25864922

ABSTRACT

The Reelin signaling pathway is implicated in processes controlling synaptic plasticity and hippocampus-dependent learning and memory. A single direct in vivo application of Reelin enhances long-term potentiation, increases dendritic spine density and improves associative and spatial learning and memory. Angelman syndrome (AS) is a neurological disorder that presents with an overall defect in synaptic function, including decreased long-term potentiation, reduced dendritic spine density, and deficits in learning and memory, making it an attractive model in which to examine the ability of Reelin to recover synaptic function and cognitive deficits. In this study, we investigated the effects of Reelin administration on synaptic plasticity and cognitive function in a mouse model of AS and demonstrated that bilateral, intraventricular injections of Reelin recover synaptic function and corresponding hippocampus-dependent associative and spatial learning and memory. Additionally, we describe alteration of the Reelin profile in tissue from both the AS mouse and post-mortem human brain.


Subject(s)
Angelman Syndrome/physiopathology , Angelman Syndrome/psychology , Cell Adhesion Molecules, Neuronal/administration & dosage , Extracellular Matrix Proteins/administration & dosage , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Nerve Tissue Proteins/administration & dosage , Serine Endopeptidases/administration & dosage , Angelman Syndrome/drug therapy , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Dendritic Spines/drug effects , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Female , HEK293 Cells , Hippocampus/physiopathology , Hippocampus/ultrastructure , Humans , Injections, Intraventricular , Male , Mice , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Spatial Learning/drug effects , Spatial Memory/drug effects
19.
Gen Physiol Biophys ; 34(1): 81-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25504061

ABSTRACT

Nesfatin is a peptide secreted by peripheral tissues, central and peripheral nervous system. It is involved in the regulation of homeostasis. Although the effects of nesfatin-1 on nutrition have been studied widely in the literature, the mechanisms of nesfatin-1 action and also relations with other physiological parameters are still not clarified well. We aimed to investigate the effect of peripheral chronic nesfatin-1 application on blood pressure regulation in normal and in rats exposed to restraint immobilization stress. In our study, three month-old male Wistar rats were used. Rats were divided into 4 groups as Control, Stress, Control+Nesfatin-1, Nesfatin-1+Stress. Angiotensinogen, angiotensin converting enzyme 2, angiotensin II, endothelin-1, endothelial nitric oxide synthase, aldosterone, cortisol, nesfatin-1 levels were determined in plasma samples by ELISA. Our results have shown that chronic peripheral nesfatin-1 administration increases blood pressure in normal and in rats exposed to chronic restraint stress. Effect of nesfatin-1 on circulating level of angiotensinogen, angiotensin converting enzyme 2, angiotensin II, endothelin-1, endothelial nitric oxide synthase, aldosterone and cortisol has been identified. We can conclude that elevated high blood pressure after chronic peripheral nesfatin-1 administration in rats exposed to chronic restraint stress may be related to decreased plasma level of endothelial nitric oxide synthase concentration.


Subject(s)
Blood Pressure/drug effects , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Gene Expression Regulation, Enzymologic , Nerve Tissue Proteins/administration & dosage , Animals , Body Weight , Endothelin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Male , Nitric Oxide Synthase Type III/metabolism , Nucleobindins , Rats , Rats, Wistar , Renin-Angiotensin System , Stress, Physiological
20.
Ren Fail ; 37(5): 882-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25707521

ABSTRACT

INTRODUCTION: The pathogenetic mechanisms underlying ischemia-reperfusion (I/R) injury involve oxidative stress, inflammation and apoptosis. Nesfatin-1, a novel peptide, has been reported to possess antioxidant, anti-inflammatory and anti-apoptic properties. The study was to examine the potential protective effects of nesfatin-1 on renal I/R injury. MATERIALS AND METHODS: I/R model was induced by placing a clamp across left renal artery for 45 min followed by 24 h reperfusion, along with a contralateral nephrectom. Twenty-four rats divided into three groups: sham-operated group, vehicle-treated I/R and nesfatin-1-treated I/R. Nesfatin-1 was intraperitoneally injected 30 min before renal ischemia. We harvested serum and kidneys at 24 h after reperfusion. Renal function and histological changes were assessed. Marker of oxidative stress and cells in kidney were also evaluated. RESULTS: The animals with nesfatin-1 significantly improved renal functional and histologic lesions induced by I/R injury. The malondialdehyde (MDA) level decreased, whereas superoxide dismutase (SOD) and catalase (CAT) activities were significantly increased. Moreover, nesfatin-1-treated rats had a markedly decrease in apoptotic tubular cells, as well as a decrease in caspase-3 activity and an increase in the bcl-2/Bax ratio. CONCLUSIONS: This is the first evidence that nesfatin-1 treatment ameliorates acute renal I/R injury by suppressing oxidative stress and cell apoptosis. Therefore, it is promising as a potential therapeutic agent for renal IR injury.


Subject(s)
Acute Kidney Injury/pathology , Antioxidants/administration & dosage , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Kidney/pathology , Nerve Tissue Proteins/administration & dosage , Oxidative Stress/drug effects , Reperfusion Injury/pathology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Male , Malondialdehyde/metabolism , Nucleobindins , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL