Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Pineal Res ; 67(3): e12594, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31286565

ABSTRACT

Astronauts experience osteoporosis-like loss of bone mass because of microgravity conditions during space flight. To prevent bone loss, they need a riskless and antiresorptive drug. Melatonin is reported to suppress osteoclast function. However, no studies have examined the effects of melatonin on bone metabolism under microgravity conditions. We used goldfish scales as a bone model of coexisting osteoclasts and osteoblasts and demonstrated that mRNA expression level of acetylserotonin O-methyltransferase, an enzyme essential for melatonin synthesis, decreased significantly under microgravity. During space flight, microgravity stimulated osteoclastic activity and significantly increased gene expression for osteoclast differentiation and activation. Melatonin treatment significantly stimulated Calcitonin (an osteoclast-inhibiting hormone) mRNA expression and decreased the mRNA expression of receptor activator of nuclear factor κB ligand (a promoter of osteoclastogenesis), which coincided with suppressed gene expression levels for osteoclast functions. This is the first study to report the inhibitory effect of melatonin on osteoclastic activation by microgravity. We also observed a novel action pathway of melatonin on osteoclasts via an increase in CALCITONIN secretion. Melatonin could be the source of a potential novel drug to prevent bone loss during space flight.


Subject(s)
Bone Resorption/prevention & control , Melatonin/therapeutic use , Space Flight , Animals , Bone Density/drug effects , Calcitonin/metabolism , Cell Differentiation/drug effects , Goldfish , Immunohistochemistry , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Weightlessness/adverse effects
2.
Article in English | MEDLINE | ID: mdl-28614698

ABSTRACT

The nucleotide sequence of a sardine preprocalcitonin precursor has been determined from their ultimobranchial glands in the present study. From our analysis of this sequence, we found that sardine procalcitonin was composed of procalcitonin amino-terminal cleavage peptide (N-proCT) (53 amino acids), CT (32 amino acids), and procalcitonin carboxyl-terminal cleavage peptide (C-proCT) (18 amino acids). As compared with C-proCT, N-proCT has been highly conserved among teleosts, reptiles, and birds, which suggests that N-proCT has some bioactivities. Therefore, both sardine N-proCT and sardine CT were synthesized, and their bioactivities for osteoblasts and osteoclasts were examined using our assay system with goldfish scales that consisted of osteoblasts and osteoclasts. As a result, sardine N-proCT (10-7M) activated osteoblastic marker enzyme activity, while sardine CT did not change. On the other hand, sardine CT (10-9 to 10-7M) suppressed osteoclastic marker enzyme activity, although sardine N-proCT did not influence enzyme activity. Furthermore, the mRNA expressions of osteoblastic markers such as type 1 collagen and osteocalcin were also promoted by sardine N-proCT (10-7M) treatment; however, sardine CT did not influence their expressions. The osteoblastic effects of N-proCT lack agreement. In the present study, we can evaluate exactly the action for osteoblasts because our scale assay system is very sensitive and it is a co-culture system for osteoblasts and osteoclasts with calcified bone matrix. Both CT and N-proCT seem to influence osteoblasts and osteoclasts and promote bone formation by different actions in teleosts.


Subject(s)
Calcitonin/analogs & derivatives , Calcitonin/pharmacology , Osteoblasts/drug effects , Amino Acid Sequence , Animals , Base Sequence , Calcitonin/genetics , Goldfish , Phylogeny , Sequence Homology, Amino Acid
3.
Zoolog Sci ; 30(3): 217-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23480382

ABSTRACT

Fish scales are a form of calcified tissue similar to that found in human bone. In medaka scales, we detected both osteoblasts and osteoclasts and subsequently developed a new scale assay system. Using this system, we analyzed the osteoblastic and osteoclastic responses under 2-, 3-, and 4-gravity (G) loading by both centrifugation and vibration. After loading for 10 min, the scales from centrifugal and vibration loading were incubated for 6 and 24 hrs, respectively, after which the osteoblastic and osteoclastic activities were measured. Osteoblastic activity significantly increased under 2- to 4-G loading by both centrifugation and vibration. In contrast, we found that osteoclastic activity significantly decreased under 2- and 3-G loading in response to both centrifugation and vibration. Under 4-G loading, osteoclastic activity also decreased on centrifugation, but significantly increased under 4-G loading by vibration, concomitant with markedly increased osteoblastic activity. Expression of the receptor activator of the NF-κB ligand (RANKL), an activation factor of osteoclasts expressed in osteoblasts, increased significantly under 4-G loading by vibration but was unchanged by centrifugal loading. A protein sequence similar to osteoprotegerin (OPG), which is known as an osteoclastogenesis inhibitory factor, was found in medaka using our sequence analysis. The ratio of RANKL/OPG-like mRNAs in the vibration-loaded scales was significantly higher than that in the control scales, although there was no difference between centrifugal loaded scales and the control scales. Accordingly, medaka scales provide a useful model by which to analyze bone metabolism in response to physical strain.


Subject(s)
Hypergravity , Oryzias/anatomy & histology , Osteoblasts/physiology , Osteoclasts/physiology , Amino Acid Sequence , Animals , Biomechanical Phenomena , Gene Expression Regulation/physiology , Osteoblasts/cytology , Osteoclasts/cytology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism
4.
Zoolog Sci ; 29(8): 499-504, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22873807

ABSTRACT

Using our original in vitro assay system with goldfish scales, we examined the direct effect of prostaglandin E2 (PGE2) on osteoclasts and osteoblasts in teleosts. In this assay system, we measured the activity of alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) as respective indicators of each activity in osteoblasts and osteoclasts. ALP activity in scales significantly increased following treatment at high concentration of PGE2(10⁻7 and 10⁻6 M) over 6 hrs of incubation. At 18 hrs of incubation, ALP activity also significantly increased in the PGE2 (10⁻9 to 10⁻6 M)-treated scale. In the case of osteoclasts, TRAP activity tended to increase at 6 hrs of incubation, and then significantly increased at 18 hrs of incubation by PGE2 (10(-7) to 10⁻6 M) treatment. At 18 hrs of incubation, the mRNA expression of osteoclastic markers (TRAP and cathepsin K) and receptor activator of the NF-κB ligand (RANKL), an activating factor of osteoclasts expressed in osteoblasts, increased in PGE2 treated-scales. Thus, PGE2 acts on osteoblasts, and then increases the osteoclastic activity in the scales of goldfish as it does in the bone of mammals. In an in vivo experiment, plasma calcium levels and scale TRAP and ALP activities in the PGE2-injencted goldfish increased significantly. We conclude that, in teleosts, PGE2 activates both osteoblasts and osteoclasts and participates in calcium metabolism.


Subject(s)
Calcium/physiology , Dinoprostone/pharmacology , Goldfish/physiology , Osteoblasts/drug effects , Osteoclasts/drug effects , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Cathepsin K/genetics , Cathepsin K/metabolism , Gene Expression Regulation/physiology , Integumentary System/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Osteoblasts/physiology , Osteoclasts/physiology , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tartrate-Resistant Acid Phosphatase , Tissue Culture Techniques
5.
J Comp Neurol ; 520(18): 4168-83, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22605619

ABSTRACT

The central melanocortin system regulates body energy homeostasis including the melanocortin-4 receptor (MC4R). The lateral hypothalamic area (LHA) receives dense melanocortinergic inputs from the arcuate nucleus of the hypothalamus and regulates multiple processes including food intake, reward behaviors, and autonomic function. By using a mouse line in which green fluorescent protein (GFP) is expressed under control of the MC4R gene promoter, we systemically investigated MC4R signaling in the LHA by combining double immunohistochemistry, electrophysiology, and retrograde tracing techniques. We found that LHA MC4R-GFP neurons coexpress neurotensin as well as the leptin receptor but do not coexpress other peptide neurotransmitters found in the LHA including orexin, melanin-concentrating hormone, and nesfatin-1. Furthermore, electrophysiological recording demonstrated that leptin, but not the MC4R agonist melanotan II, hyperpolarizes the majority of LHA MC4R-GFP neurons in an ATP- sensitive potassium channel-dependent manner. Retrograde tracing revealed that LHA MC4R-GFP neurons do not project to the ventral tegmental area, dorsal raphe nucleus, nucleus accumbens, and spinal cord, and only limited number of neurons project to the nucleus of the solitary tract and parabrachial nucleus. Our findings provide new insights into MC4R signaling in the LHA and its potential implications in homeostatic regulation of body energy balance.


Subject(s)
Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Neural Pathways/physiology , Neuroanatomy , Neurons/physiology , Receptor, Melanocortin, Type 4/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Count , Cholera Toxin/metabolism , Circadian Rhythm/physiology , DNA-Binding Proteins/metabolism , Green Fluorescent Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leptin/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neuropeptides/metabolism , Nucleobindins , Orexins , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 4/genetics , STAT3 Transcription Factor/metabolism , Stilbamidines/metabolism
6.
Int J Dev Neurosci ; 30(6): 499-505, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22627026

ABSTRACT

The ventroposterior thalamus and the habenular nuclei of the epithalamus are relevant to the monoaminergic system functionally and anatomically. The glia-derived S100B protein plays a critical role in the development of the nervous system including the monoaminergic systems. In this study, we performed an immunohistochemical study of glia-related proteins including S100B, serotonin transporter, and microtubule-associated protein 2, as well as cytochrome oxidase histochemistry in neonatal rats. Results showed the same findings for S100B immunohistochemistry between the ventroposterior thalamus and the lateral habenula at postnatal day 7: intense staining in cell bodies of astrocytes, diffusely spread immunoproduct in the intercellular space, and S100B-free areas as well as a strong reaction to cytochrome oxidase histochemistry. Further common features were the scarcity of glial fibrillary acidic protein-positive astrocytes and the few apoptotic cells observed. The results of the cytochrome oxidase reaction suggested that S100B is released actively into intercellular areas in restricted brain regions showing high neuronal activity at postnatal day 7. Pathology of the ventroposterior thalamus and the habenula is suggested in mental disorders, and S100B might be a key factor for investigations in these areas.


Subject(s)
Electron Transport Complex IV/metabolism , Habenula/growth & development , Habenula/metabolism , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Ventral Thalamic Nuclei/growth & development , Ventral Thalamic Nuclei/metabolism , Age Factors , Animals , Animals, Newborn , Glial Fibrillary Acidic Protein/metabolism , Habenula/cytology , Microtubule-Associated Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , S100 Calcium Binding Protein beta Subunit , Serotonin Plasma Membrane Transport Proteins/metabolism , Ventral Thalamic Nuclei/cytology
7.
Gene ; 499(2): 326-31, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22429839

ABSTRACT

In cartilaginous fish, two cDNAs encoding calcitonin-family receptors were isolated for the first time from the stingray brain. The open reading frame of one receptor cDNA coded a 525-amino acid protein. The amino acid identity of this receptor to human calcitonin-receptor-like receptor (CRLR) is 64.5%, frog CRLR is 64.7%, and flounder CRLR is 61.2% and this was higher than to human calcitonin receptor (CTR) (46.1%), frog CTR (54.7%), and flounder CTR (48.9%). We strongly suggested that this receptor is a ray CRLR based on phylogenetic analysis. In case of the second receptor, amino acid identity among CRLRs (human 50.5%, frog 50.7%, flounder 48.0%) and CTRs (human 43.2%, frog 49.1%, flounder 41.8%) was similar. From phylogenetic analysis of both CRLRs and CTRs, we believe that this receptor is ray CTR. The expression of ray CRLR mRNA was predominantly detected in the nervous system (brain) and vascular system (atrium, ventricle, and gill), which reflects the similar localization of CGRP in the nervous and vascular systems as mammals. It was observed that the second receptor was expressed in several tissues, namely cartilage, brain, pituitary gland, gill, atrium, ventricle, pancreas, spleen, liver, gall bladder, intestine, rectal gland, kidney, testis and ovary. This localization pattern was very similar to flounder CTR. Both receptor mRNAs were strongly expressed in the gill. This suggests that the calcitonin-family members are involved in the osmoregulation of stingray as this fish is known to be euryhaline. When a stingray was transferred to diluted seawater (20% seawater), the expression of both receptors significantly decreased in the gill. Similar results were obtained in the kidney of the stingray. Thus, our cloning and isolation of both receptors in the stingray will be helpful for elucidation of their physiological role(s) such as osmoregulation including calcium metabolism of cartilaginous fish.


Subject(s)
Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Skates, Fish/genetics , Animals , Brain/metabolism , Cloning, Molecular , Female , Gills/metabolism , Kidney/metabolism , Male , Seawater , Skates, Fish/metabolism , Water-Electrolyte Balance
8.
J Comp Neurol ; 518(1): 6-24, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19882715

ABSTRACT

Vagal afferents regulate energy balance by providing a link between the brain and postprandial signals originating from the gut. In the current study, we investigated melanocortin-4 receptor (MC4R) expression in the nodose ganglion, where the cell bodies of vagal sensory afferents reside. By using a line of mice expressing green fluorescent protein (GFP) under the control of the MC4R promoter, we found GFP expression in approximately one-third of nodose ganglion neurons. By using immunohistochemistry combined with in situ hybridization, we also demonstrated that approximately 20% of GFP-positive neurons coexpressed cholecystokinin receptor A. In addition, we found that the GFP is transported to peripheral tissues by both vagal sensory afferents and motor efferents, which allowed us to assess the sites innervated by MC4R-GFP neurons. GFP-positive efferents that co-expressed choline acetyltransferase specifically terminated in the hepatic artery and the myenteric plexus of the stomach and duodenum. In contrast, GFP-positive afferents that did not express cholinergic or sympathetic markers terminated in the submucosal plexus and mucosa of the duodenum. Retrograde tracing experiments confirmed the innervation of the duodenum by GFP-positive neurons located in the nodose ganglion. Our findings support the hypothesis that MC4R signaling in vagal afferents may modulate the activity of fibers sensitive to satiety signals such as cholecystokinin, and that MC4R signaling in vagal efferents may contribute to the control of the liver and gastrointestinal tract.


Subject(s)
Neural Pathways , Nodose Ganglion , Postprandial Period/physiology , Receptor, Cholecystokinin A/metabolism , Receptor, Melanocortin, Type 4/metabolism , Vagus Nerve , Animals , Gastrointestinal Tract/innervation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Liver/innervation , Male , Mice , Mice, Transgenic , Neural Pathways/anatomy & histology , Neural Pathways/physiology , Nodose Ganglion/cytology , Nodose Ganglion/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Melanocortin, Type 4/genetics , Vagus Nerve/cytology , Vagus Nerve/metabolism
9.
Cell ; 123(3): 493-505, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16269339

ABSTRACT

Activation of melanocortin-4-receptors (MC4Rs) reduces body fat stores by decreasing food intake and increasing energy expenditure. MC4Rs are expressed in multiple CNS sites, any number of which could mediate these effects. To identify the functionally relevant sites of MC4R expression, we generated a loxP-modified, null Mc4r allele (loxTB Mc4r) that can be reactivated by Cre-recombinase. Mice homozygous for the loxTB Mc4r allele do not express MC4Rs and are markedly obese. Restoration of MC4R expression in the paraventricular hypothalamus (PVH) and a subpopulation of amygdala neurons, using Sim1-Cre transgenic mice, prevented 60% of the obesity. Of note, increased food intake, typical of Mc4r null mice, was completely rescued while reduced energy expenditure was unaffected. These findings demonstrate that MC4Rs in the PVH and/or the amygdala control food intake but that MC4Rs elsewhere control energy expenditure. Disassociation of food intake and energy expenditure reveals unexpected divergence in melanocortin pathways controlling energy balance.


Subject(s)
Eating/physiology , Energy Metabolism/physiology , Receptor, Melanocortin, Type 4/biosynthesis , Amygdala/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Integrases/genetics , Mice , Mice, Knockout , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 4/genetics , Repressor Proteins/genetics
10.
Neuroreport ; 16(9): 897-901, 2005 Jun 21.
Article in English | MEDLINE | ID: mdl-15931058

ABSTRACT

Gamma aminobutyric acid (GABA) is localized in neuropeptide Y (NPY) neurons of the hypothalamic arcuate nucleus (ARC). We examined regulation of ARC NPY neurons by GABA. Light and electron microscopic immunohistochemistry confirmed that GABA-containing nerve terminals contacted NPY-containing neurons in the ARC. Lowering glucose (1 mM) increased cytosolic Ca2+ concentration ([Ca2+]i) in isolated ARC neurons that were immunoreactive to NPY. The [Ca2+]i increases were inhibited by GABA, the gamma-aminobutyric acid type A receptor (GABAA) agonist muscimol and the gamma-aminobutyric acid type B receptor (GABAB) agonist baclofen. Neither the GABAA antagonist bicuculline nor the GABAB antagonist CGP35348 counteracted the GABA inhibition when applied alone, but did so when applied together. These results indicate that GABA regulates ARC glucose-sensitive NPY neurons via GABAA and GABAB receptors, which could function to attenuate the orexigenic NPY pathway when it is not beneficial.


Subject(s)
Arcuate Nucleus of Hypothalamus/cytology , Glucose/pharmacology , Neurons/drug effects , Neuropeptide Y/metabolism , Receptors, GABA/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Baclofen/pharmacology , Bicuculline/pharmacology , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , GABA Agonists/pharmacology , GABA Antagonists , Immunohistochemistry/methods , Male , Microscopy, Immunoelectron/methods , Muscimol/pharmacology , Neurons/metabolism , Neurons/ultrastructure , Organophosphorus Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA/classification , Time Factors
11.
Neurosci Lett ; 385(2): 110-3, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-15961227

ABSTRACT

Salusins are two newly discovered TOR-related peptides consisting of 28 and 20 amino acids and designated salusin-alpha and salusin-beta, respectively. Using immunohistochemistry techniques, salusin-like immunoreactivity was detected in the rat hypothalamo-neurohypophyseal tract and immunopositive cells were distributed in the suprachiasmatic, supraoptic and paraventricular nucleus. In the paraventricular nucleus, salusin-like immunoreactivity was observed both in parvocellular and magnocellular neurons. Many salusin-positive nerve fibers and their terminals were identified in the internal layer of the median eminence and posterior pituitary. Less intense salusin-positive staining of fibers and terminals was found in the suprachiasmatic nucleus and external layer of the median eminence. Dual immunostaining was performed to determine if salusin coexisted with vasopressin or oxytocin in the hypothalamus. Most of the salusin-like immunoreactivity was detected in vasopressin- but not in oxytocin-containing neurons in these nuclei. The functional significance of the coexistence of salusin with vasopressin is discussed, including the possibility that salusin participates in the regulation of blood pressure.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Vasopressins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Blood Pressure/physiology , Hypothalamo-Hypophyseal System/anatomy & histology , Hypothalamus/anatomy & histology , Immunohistochemistry , Male , Median Eminence/cytology , Median Eminence/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland, Posterior/cytology , Pituitary Gland, Posterior/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Suprachiasmatic Nucleus/metabolism , Supraoptic Nucleus/metabolism , Vasoconstriction/physiology
12.
Regul Pept ; 126(1-2): 49-54, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15620413

ABSTRACT

Orexins are neuropeptides that have a range of physiological effects including the regulation of feeding behavior and the sleep-wakefulness cycle. Recently, we reported that level of orexin A in spinal fluid was decreased in the patients of some neurodegenerative diseases and it is considered that orexin A and the receptors might be related to central nervous system disorders. However, the expression and localization of orexin receptors is not elicited well. Therefore, the purpose of this study is to investigate the time-dependent changes and the cellular localization of orexin receptor focusing on orexin-1 receptor (OX1R) in the mouse brain after transient common carotid artery occlusion (tCCAO) model by using immunohistochemical techniques. OX1R immunoreactivity dramatically increased and peaked in the hippocampus and cortex 2 days after tCCAO, but remained unchanged in the hypothalamus. Using double-immunohistochemistry, the OX1R immunopositive cells at 2 days after tCCAO were co-localized not only with neuronal marker, NeuN-immunoreactivity but also with astroglial and oligodendroglial markers, GFAP- and CNPase-immunoreactivities, respectively. These results suggested that OX1R is induced other cells in addition to the neurons during stress such as ischemia and orexins and its receptor might play an important role for ischemic insult.


Subject(s)
Brain Ischemia/metabolism , Cerebellar Cortex/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, Neuropeptide/biosynthesis , Animals , Brain Ischemia/pathology , Cerebellar Cortex/pathology , Hippocampus/pathology , Male , Mice , Neurons/pathology , Orexin Receptors , Stroke/metabolism , Stroke/pathology , Stroke/physiopathology
13.
Regul Pept ; 126(1-2): 67-71, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15620416

ABSTRACT

Ghrelin, a novel peptide isolated from stomach tissue of rats and humans, has been identified as the endogenous ligand for the growth hormone secretagogue receptor (GHS-R). In addition to its secretion from the stomach, ghrelin is also expressed in the hypothalamic arcuate nucleus, intestine, kidney, placenta, and pancreas. GHS-R mRNA, on the other hand, is expressed in the hypothalamus, pituitary, heart, lung, liver, pancreas, stomach, intestine, and adipose tissue. Ghrelin is considered to have important roles in feeding regulation and energy metabolism as well as in the release of growth hormone (GH). Recent physiological experiments on the pancreas have shown that ghrelin regulates insulin secretion. However, sites of action of ghrelin in the pancreas are yet to be identified. In this study, to gain insight into the role of ghrelin in rat pancreatic islets, we used immunohistochemistry to determine the localization of ghrelin and GHS-R in islet cells. Double fluorescence immunohistochemistry revealed that weak GHS-R-like immunoreactivity was found in B cells containing insulin. GHS-R immunoreactivity overlapped that of glucagon-like immunoreactive cells. Moreover, both ghrelin and GHS-R-like immunoreactivities were detected mostly in the same cells in the periphery of the islets of Langerhans. These observations suggest that ghrelin is synthesized and secreted from A cells, and acts back on A cells in an autocrine and/or paracrine manner. In addition, ghrelin may act on B cells via GHS-R to regulate insulin secretion.


Subject(s)
Islets of Langerhans/metabolism , Peptide Hormones/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Animals , Ghrelin , Growth Hormone/metabolism , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Male , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin
14.
Regul Pept ; 126(1-2): 79-83, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15620418

ABSTRACT

Galanin-like peptide (GALP) is a novel orexigenic neuropeptide that is recently isolated from the porcine hypothalamus. GALP-containing neurons predominantly locate in the hypothalamic arcuate nucleus (ARC). The expression of GALP mRNA within the ARC is increased after the administration of leptin. GALP-containing neurons express leptin receptor and contain alpha-melanocyte-stimulating hormone. We have recently reported that neuropeptide Y (NPY)- and orexin-containing axon terminals are in close apposition with GALP-containing neurons in the ARC. In addition, GALP-containing neurons express orexin-1 receptor (OX1-R). Thus, GALP may function under the influence of leptin and orexin. However, the target neurons of GALP have not yet been clarified. To clarify the neuronal interaction between GALP-containing and other feeding regulating neurons, double-immunostaining method using antibodies against GALP- and orexin- or melanin-concentrating hormone (MCH) was performed in the rat lateral hypothalamus (LH). GALP-immunoreactive fibers appeared to project to the LH around the fornix. They were also found from the rostral to the caudal part of the ARC, paraventricular nucleus (PVH), stria terminalis (BST), medial preoptic area (MPA), and lateral septal nucleus (LSV). Moreover, GALP-like immunoreactive nerve fibers were directly contacted with orexin- and melanin-concentrating hormone (MCH)-like immunoreactive neurons in the LH. Our findings strongly suggest that GALP-containing neurons interact with orexin- and/or MCH-containing neurons in the lateral hypothalamus and that it participates in the regulation of feeding behavior in harmony with other feeding-regulating neurons in the hypothalamus.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Cell Communication/physiology , Galanin-Like Peptide/biosynthesis , Hypothalamic Hormones/biosynthesis , Melanins/biosynthesis , Neuropeptides/biosynthesis , Pituitary Hormones/biosynthesis , Animals , Axons/physiology , Eating/physiology , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins , Male , Orexins , Rats , Rats, Sprague-Dawley
15.
Regul Pept ; 126(1-2): 137-43, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15620427

ABSTRACT

Cholecystokinin (CCK) plays a major role in the regulation of pancreatic enzyme secretion based on its binding to the CCK-A receptor (CCK-AR). While CCK-AR is known to be expressed in rat islet B cells, the localization of CCK-AR in rat pancreatic A and D cells remains poorly understood. The aim of this study was to identify the localization of CCK-AR in rat pancreatic islets by means of double immunofluorescence straining with antibodies against CCK-AR, glucagon, insulin and somatostatin and with in situ hybridization to detect its transcript. CCK-AR-like immunoreactive cells were found to overlap both with glucagon-like immunoreactive cells and insulin-like immunoreactive cells but not with somatostatin-like immunoreactive cells. An in situ hybridization study using a cRNA probe for CCK-AR revealed that CCK-AR mRNA was expressed in the center and periphery of the pancreatic islets. Further to this, immunofluorecsence staining using anti-glucagon antibody was carried out after in situ hybridization using the CCK-AR cRNA probe in order to identify CCK-AR mRNA expressing cells. CCK-AR mRNA exhibited a distribution pattern almost identical to that of glucagon-like immunoreactive cells. These results show clearly that CCK-AR exists not only in B but also in A cells of the rat pancreas, suggesting that CCK regulates the secretion of insulin and glucagon at least partly via CCK-AR.


Subject(s)
Islets of Langerhans/physiology , Receptor, Cholecystokinin A/biosynthesis , Animals , Cholecystokinin/metabolism , Glucagon/metabolism , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Male , Rats , Rats, Sprague-Dawley
16.
Regul Pept ; 123(1-3): 167-72, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15518908

ABSTRACT

In the present work, PAC1-R (G-protein-coupled receptor specific for PACAP) was detected on cells in the normal thymus. Immunohistochemically PAC1-R was expressed strongly in stromal cells of the thymic medulla. Positive cells were also observed in the thymus of fetal and old adult rats. After 8 Gy irradiation to 9-week-old rats, PAC1-R expressions in the thymus decreased and almost recovered by day 21. The expression of PAC1-R mRNA was weak in the thymus and decreased further after irradiation. The expression almost recovered by day 28. Hip and hip/hop variants, which were not expressed in the normal thymus, were expressed in the thymus on days 3, 5 and 21 after irradiation. The expressions of IL-6 and IL-10 tended to increase initially after irradiation then decreased. Histologically, the thymic structures were destroyed on day 3 after irradiation and the thymus almost recovered by day 21. Thus PACAP is thought to be one of the important factors for cross-talk between cells involved in thymic regeneration.


Subject(s)
Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Thymus Gland/metabolism , Thymus Gland/radiation effects , Animals , Base Sequence , DNA, Complementary/genetics , Female , Gene Expression/radiation effects , Immunohistochemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/pathology
17.
Eur J Neurosci ; 19(6): 1524-34, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15066149

ABSTRACT

Orexin-A and -B (hypocretin-1 and -2) have been implicated in the stimulation of feeding. Here we show the effector neurons and signaling mechanisms for the orexigenic action of orexins in rats. Immunohistochemical methods showed that orexin axon terminals contact with neuropeptide Y (NPY)- and proopiomelanocortin (POMC)-positive neurons in the arcuate nucleus (ARC) of the rats. Microinjection of orexins into the ARC markedly increased food intake. Orexins increased cytosolic Ca(2+) concentration ([Ca(2+)](i)) in the isolated neurons from the ARC, which were subsequently shown to be immunoreactive for NPY. The increases in [Ca(2+)](i) were inhibited by blockers of phospholipase C (PLC), protein kinase C (PKC) and Ca(2+) uptake into endoplasmic reticulum. The stimulation of food intake and increases in [Ca(2+)](i) in NPY neurons were greater with orexin-A than with orexin-B, indicative of involvement of the orexin-1 receptor (OX(1)R). In contrast, orexin-A and -B equipotently attenuated [Ca(2+)](i) oscillations and decreased [Ca(2+)](i) levels in POMC-containing neurons. These effects were counteracted by pertussis toxin, suggesting involvement of the orexin-2 receptor and Gi/Go subtypes of GTP-binding proteins. Orexins also decreased [Ca(2+)](i) levels in glucose-responsive neurons in the ventromedial hypothalamus (VMH), a satiety center. Leptin exerted opposite effects on these three classes of neurons. These results demonstrate that orexins directly regulate NPY, POMC and glucose-responsive neurons in the ARC and VMH, in a manner reciprocal to leptin. Orexin-A evokes Ca(2+) signaling in NPY neurons via OX(1)R-PLC-PKC and IP(3) pathways. These neural pathways and intracellular signaling mechanisms may play key roles in the orexigenic action of orexins.


Subject(s)
Calcium Signaling/drug effects , Carrier Proteins/pharmacology , Glucose/metabolism , Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins , Leptin/pharmacology , Neuropeptide Y/metabolism , Neuropeptides/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Dose-Response Relationship, Drug , Drug Interactions , Eating/drug effects , Enzyme Inhibitors , Fura-2/metabolism , Immunohistochemistry , Models, Neurological , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Orexins , Pertussis Toxin/pharmacology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
18.
Endocrine ; 23(1): 59-75, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15034198

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is found not only in the brain, but is also abundantly expressed in the testicular germ cells. However, the physiological role of testicular PACAP remains unknown. Autoradiographic studies showed a considerable number of PACAP-specific binding sites in the seminiferous tubules. Immunohistochemistry demonstrated PAC1-receptor (R)-like immunoreactivity (li) in the cytoplasm of round spermatids, aggregated in the acrosome and coexpressed with PACAP-li. Spermatid-enriched fractions were examined for the subcellular localization of PACAP binding sites and PAC1-R-li. The highest levels of PACAP binding sites and PAC1-R-li were found in the cytosolic, followed by the nuclear, and the lowest levels in the membrane fraction. The testicular cytosolic PAC1-R-like protein showed a specific competitive inhibition in the radio-receptor assay for PACAP38 and 27, with a Ki of 0.069 nM and 0.179 nM, respectively. The addition of PACAP to the cytosol of spermatids only slightly activated adenylate cyclase, while it markedly stimulated the expression and activation of ERK-type mitogen-activated protein kinase (MAPK). In the PAC1-R-like protein-depleted cytosol, a PAC1-R-specific agonist, maxadilan, did not activate MAPK, but PACAP and VIP still did. Because VPAC2-R, which binds both PACAP and VIP, is expressed in the testis, the findings suggest that cytosolic VPAC2-R-like proteins are also present and coupled to MAPK. The MAPK activation does not seem to require a heterotrimeric G-protein. Because PACAP and its receptors are coexpressed in the cytoplasm of spermatids, endogenous PACAP may directly interact with the cytosolic PAC1-R-like protein without the ligand being released into the extracellular space. This possibility is supported by the observation that cytosolic endogenous PACAP in spermatids was co-immunoprecipitated with the cytosolic PAC1-R. This mechanism may be called "intracrine," and its physiological significance is discussed.


Subject(s)
Neuropeptides/physiology , Receptors, Pituitary Hormone/physiology , Spermatids/physiology , Testis/physiology , Adenylyl Cyclases/metabolism , Animals , Binding Sites/physiology , Binding, Competitive/physiology , Blotting, Western , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Immunohistochemistry , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide, Type II , Signal Transduction/physiology , Subcellular Fractions/physiology , Testis/cytology , Vasoactive Intestinal Peptide/pharmacology
19.
J Neurosci Res ; 75(3): 384-90, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14743451

ABSTRACT

Glycoproteins from mammalian brain tissues contain unique N-linked oligosaccharides terminating with beta-N-acetylglucosamine residues. Lectin blot analysis of membrane glycoprotein samples from human neuroblastoma SH-SY5Y cells showed that several protein bands bind to Psathylera velutina lectin (PVL), which interacts with beta-N-acetylglucosamine-terminating oligosaccharides. No lectin positive bands were detected by digestion with jack bean beta-N-acetyl-hexosaminidase or N-glycanase before incubation with the lectin, indicating that the cells contain beta-N-acetylglucosamine-terminating N-linked oligosaccharides. When cells were cultured in dishes with different concentrations of PVL, the cell proliferation was inhibited in a dose-dependent manner. Similarly, the neurite extension, which was stimulated with nerve growth factor, was also inhibited in a manner dependent on the lectin dose. Cell proliferation and neurite extension were recovered by the addition of 10 mM N-acetylglucosamine into the medium. Immunoblot analysis of the activation of mitogen-activated protein (MAP) kinases and protein kinase C revealed that phosphorylation of 42-kDa and 44-kDa MAP kinases and 80-kDa protein kinase C are inhibited when SH-SY5Y cells are cultured in PVL-coated dishes, but are restored by the addition of the haptenic sugar into the medium, indicating that MAP kinase and protein kinase C pathways are inhibited by interaction with immobilized PVL. These results indicate that beta-N-acetylglucosamine-terminating N-linked oligosaccharides expressed on neural cells can induce intracellular signals upon binding to extracellular receptors, and are important for growth regulation of neural cells.


Subject(s)
Acetylglucosamine/metabolism , Lectins , Membrane Glycoproteins/metabolism , Neurites/physiology , Oligosaccharides/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/pharmacology , Agaricales/chemistry , Animals , Cell Culture Techniques , Cell Division/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Immunoblotting , Lectins/pharmacology , Membrane Glycoproteins/chemistry , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , Neurites/drug effects , Oligosaccharides/chemistry , Phosphorylation , Protein Kinase C/metabolism
20.
Neurosci Res ; 47(4): 451-4, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14630350

ABSTRACT

We have generated a transgenic mouse model in which astrocytes express an enhanced green fluorescent protein (EGFP) under the control of the mouse glial fibrillary acidic protein (GFAP) promoter. EGFP, which is characteristically found throughout the cell, was expressed in these animals even in astrocytic fine processes, and EGFP expressing cells demonstrated morphological characters of protoplasmic, fibrous, or reactive astrocytes. In contrast, GFAP immunoreactivity was found only in the perinuclear region and in the main processes. The transgenic mouse model therefore provides a valuable tool for the detailed morphological investigation of astrocytes.


Subject(s)
Astrocytes/cytology , Brain/cytology , Mice, Transgenic/anatomy & histology , Animals , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Organ Culture Techniques , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...