313 results on '"Toshihiko Yada"'
Search Results
2. Fasting inhibits excitatory synaptic input on paraventricular oxytocin neurons via neuropeptide Y and Y1 receptor, inducing rebound hyperphagia, and weight gain
- Author
-
Lei Wang, Shigetomo Suyama, Samantha A. Lee, Yoichi Ueta, Yutaka Seino, Geoffrey W. G. Sharp, and Toshihiko Yada
- Subjects
food restriction ,neuropeptide Y ,Y1 receptor ,paraventricular nucleus ,oxytocin ,synaptic plasticity ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Fasting with varying intensities is used to treat obesity-related diseases. Re-feeding after fasting exhibits hyperphagia and often rebound weight gain. However, the mechanisms underlying the hyperphagia and rebound remain elusive. Here we show that 24 h food restriction (24 h FR) and milder 50% FR, both depress synaptic transmission in the hypothalamic paraventricular nucleus (PVN) and induce acute hyperphagia in rats. 24 h FR is followed by weight rebound but 50% FR is not. Orexigenic neuropeptide Y (NPY) via the Y1 receptor (Y1R) inhibited the miniature excitatory postsynaptic current (mEPSC) on anorexigenic oxytocin neurons in the PVN. 24 h FR and 50% FR activated this neuronal pathway to induce acute hyperphagia on Days 1–3 and Days 1–2 after FR, respectively. 24 h FR induced large mEPSC depression, recurrent hyperphagia on Days 9–12 and rebound weight gain on Days 12–17, whereas 50% FR induced moderate mEPSC depression and sustained weight reduction. Transverse data analysis on Day 1 after 24 h FR and 50% FR demonstrated saturation kinetics for the mEPSC depression-hyperphagiacurve, implying hysteresis. The results reveal FR-driven synaptic plasticity in the NPY-Y1R-oxytocin neurocircuit that drives acute hyperphagia. FR with the intensity that regulates the synapse-feeding relay without hysteresis is the key for successful dieting.
- Published
- 2022
- Full Text
- View/download PDF
3. Gastrointestinal Distension by Pectin-Containing Carbonated Solution Suppresses Food Intake and Enhances Glucose Tolerance via GLP-1 Secretion and Vagal Afferent Activation
- Author
-
Kento Ohbayashi, Yukiko Oyama, Chiharu Yamaguchi, Toshiki Asano, Toshihiko Yada, and Yusaku Iwasaki
- Subjects
gastric distension ,intestinal distension ,glucagon-like peptide-1 ,vagal afferents ,food intake ,insulin sensitivity ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Diet-induced gastrointestinal distension is known to evoke satiation and suppress postprandial hyperglycemia; however, the underlying mechanisms remain poorly understood. This study explored how gastrointestinal distension regulates energy homeostasis by using inflating stomach formulation (ISF), the carbonated solution containing pectin that forms stable gel bubbles under acidic condition in the stomach. Here we show that, in mice, oral administration of ISF induced distension of stomach and proximal intestine temporarily, stimulated intestinal glucagon-like peptide-1 (GLP-1) secretion, and activated vagal afferents and brainstem. ISF suppressed food intake and improved glucose tolerance via enhancing insulin sensitivity. The anorexigenic effect was partially inhibited, and the beneficial glycemic effect was blunted by pharmacological GLP-1 receptor blockade and chemical denervation of capsaicin-sensitive sensory nerves. In HFD-fed obese mice showing arrhythmic feeding and obesity, subchronic ISF treatment at the light period (LP) onset for 10 days attenuated LP hyperphagia and visceral fat accumulation. These results demonstrate that gastrointestinal distension by ISF stimulates GLP-1 secretion and the vagal afferent signaling to the brain, thereby regulating feeding behavior and glucose tolerance. Furthermore, subchronic ISF treatment ameliorates HFD-induced visceral obesity. We propose the diet that induces gastrointestinal distension as a novel treatment of hyperphagic obesity and diabetes.
- Published
- 2021
- Full Text
- View/download PDF
4. Ninjin'yoeito Targets Distinct Ca2+ Channels to Activate Ghrelin-Responsive vs. Unresponsive NPY Neurons in the Arcuate Nucleus
- Author
-
Chayon Goswami, Katsuya Dezaki, Lei Wang, Akio Inui, Yutaka Seino, and Toshihiko Yada
- Subjects
Ninjin-yoeito ,anorexia ,arcuate nucleus ,neuropeptide Y ,ghrelin ,N-type Ca2+ channel ,Nutrition. Foods and food supply ,TX341-641 - Abstract
Appetite loss or anorexia substantially deteriorates quality of life in various diseases, and stand upstream of frailty. Neuropeptide Y (NPY) in the hypothalamic arcuate nucleus (ARC) and ghrelin released from stomach are potent inducers of appetite. We previously reported that Ninjin'yoeito, a Japanese kampo medicine comprising twelve herbs, restores food intake, and body weight in cisplatin-treated anorectic mice. Furthermore, Ninjin'yoeito increased cytosolic Ca2+ concentration ([Ca2+]i) in not only ghrelin-responsive but ghrelin-unresponsive NPY neurons in ARC. The cellular lineage/differentiation of ghrelin-unresponsive neuron is less defined but might alter along with aging and diet. This study examined the occupancy of ghrelin-unresponsive neurons among ARC NPY neurons in adult mice fed normal chow, and explored the mechanisms underlying Ninjin'yoeito-induced [Ca2+]i increases in ghrelin-unresponsive vs. ghrelin-responsive NPY neurons. Single ARC neurons were subjected to [Ca2+]i measurement and subsequent immunostaining for NPY. Ghrelin failed to increase [Ca2+]i in 42% of ARC NPY neurons. Ninjin'yoeito (10 μg/ml)-induced increases in [Ca2+]i were abolished in Ca2+ free condition in ghrelin-responsive and ghrelin-unresponsive ARC NPY neurons. Ninjin'yoeito-induced [Ca2+]i increases were inhibited by N-type Ca2+ channel blocker ω-conotoxin in the majority (17 of 20), while by L-type Ca2+ channel blocker nitrendipine in the minority (2 of 23), of ghrelin-responsive neurons. In contrast, Ninjin'yoeito-induced [Ca2+]i increases were inhibited by nitrendipine in the majority (14 of 17), while by ω-conotoxin in the minority (8 of 24), of ghrelin-unresponsive neurons. These results indicate that ghrelin-unresponsive neurons occur substantially among NPY neurons of ARC in adult mice fed normal chow. Ninjin'yoeito preferentially target N-type and L-type Ca2+ channels in the majority of ghrelin-responsive and ghrelin-unresponsive neurons, respectively, to increase [Ca2+]i. We suggest ARC N- and L-type Ca2+ channels as potential targets for activating, respectively, ghrelin-responsive, and unresponsive NPY neurons to treat anorexia.
- Published
- 2020
- Full Text
- View/download PDF
5. Peripheral circadian rhythms in the liver and white adipose tissue of mice are attenuated by constant light and restored by time-restricted feeding.
- Author
-
Daisuke Yamamuro, Manabu Takahashi, Shuichi Nagashima, Tetsuji Wakabayashi, Hisataka Yamazaki, Akihito Takei, Shoko Takei, Kent Sakai, Ken Ebihara, Yusaku Iwasaki, Toshihiko Yada, and Shun Ishibashi
- Subjects
Medicine ,Science - Abstract
Disturbance of circadian rhythms underlies various metabolic diseases. Constant light exposure (LL) is known to disrupt both central and peripheral circadian rhythms. Here, we attempted to determine whether the effects of LL are different between various peripheral tissues and whether time-restricted feeding restores the circadian rhythms especially in white adipose tissue (WAT). Six-week-old mice were subjected to three feeding regimes: ad libitum feeding under light/dark phase (LD), ad libitum feeding under LL cycle, and restricted feeding at night-time under LL cycle with a normal chow. After 3 weeks, we compared body weight, food intake, plasma levels of lipids and glucose, and the expression patterns of the clock genes and the genes involved in lipid metabolism in the liver and WAT. The mice kept under LL with or without time-restricted feeding were 5.2% heavier (p
- Published
- 2020
- Full Text
- View/download PDF
6. Neuropeptide oxytocin enhances μ opioid receptor signaling as a positive allosteric modulator
- Author
-
Yoshiyuki Meguro, Kanako Miyano, Shigeto Hirayama, Yuki Yoshida, Naoto Ishibashi, Takumi Ogino, Yuriko Fujii, Sei Manabe, Moeko Eto, Miki Nonaka, Hideaki Fujii, Yoichi Ueta, Minoru Narita, Naohiro Sata, Toshihiko Yada, and Yasuhito Uezono
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Oxytocin (OT) is a 9-amine neuropeptide that plays an essential role in mammalian labor, lactation, maternal bonding, and social affiliation. OT has been reported to exert an analgesic effect in both humans and animals, and the results of certain animal experiments have shown that the analgesic effect of OT is partially blocked by opioid receptor antagonists. To investigate the relationship between OT and μ opioid receptor (MOR), we evaluated how OT affects MOR in vitro by performing an electrical impedance-based receptor biosensor assay (CellKey™ assay), an intracellular cAMP assay, and a competitive receptor-binding analysis by using cells stably expressing human MOR and OT receptor. In both the CellKey™ assay and the intracellular cAMP assay, OT alone exerted no direct agonistic effect on human MOR, but treatment with 10−6 M OT markedly enhanced the MOR signaling induced by 10−6 M endomorphin-1, β-endorphin, morphine, fentanyl, and DAMGO. Moreover, in the competitive receptor-binding assay, 10−6 M OT did not alter the affinity of endomorphin-1 or morphine for MOR. These results suggest that OT could function as a positive allosteric modulator that regulates the efficacy of MOR signaling, and thus OT might represent a previously unrecognized candidate analgesic agent. Keywords: Oxytocin, μ opioid receptor, Positive allosteric modulator, Analgesia, G protein-coupled receptor (GPCR)
- Published
- 2018
- Full Text
- View/download PDF
7. GLP-1 release and vagal afferent activation mediate the beneficial metabolic and chronotherapeutic effects of D-allulose
- Author
-
Yusaku Iwasaki, Mio Sendo, Katsuya Dezaki, Tohru Hira, Takehiro Sato, Masanori Nakata, Chayon Goswami, Ryohei Aoki, Takeshi Arai, Parmila Kumari, Masaki Hayakawa, Chiaki Masuda, Takashi Okada, Hiroshi Hara, Daniel J. Drucker, Yuichiro Yamada, Masaaki Tokuda, and Toshihiko Yada
- Subjects
Science - Abstract
The sweetener D-allulose has beneficial metabolic effects in animal models, but its mechanism of action was unclear. Here the authors report that D-allulose triggers GLP-1 release in the gut and GLP-1R signaling on vagal afferents, counteracting arrhythmic overeating, obesity and diabetes.
- Published
- 2018
- Full Text
- View/download PDF
8. Activation of AMPK-Regulated CRH Neurons in the PVH is Sufficient and Necessary to Induce Dietary Preference for Carbohydrate over Fat
- Author
-
Shiki Okamoto, Tatsuya Sato, Michihiro Tateyama, Haruaki Kageyama, Yuko Maejima, Masanori Nakata, Satoshi Hirako, Takashi Matsuo, Sanda Kyaw, Tetsuya Shiuchi, Chitoku Toda, Udval Sedbazar, Kumiko Saito, Nur Farehan Asgar, Boyang Zhang, Shigefumi Yokota, Kenta Kobayashi, Fabienne Foufelle, Pascal Ferré, Masamitsu Nakazato, Hiroaki Masuzaki, Seiji Shioda, Toshihiko Yada, Barbara B. Kahn, and Yasuhiko Minokoshi
- Subjects
food preference ,AMPK ,CRH ,PVH ,CPT1c ,Biology (General) ,QH301-705.5 - Abstract
Food selection is essential for metabolic homeostasis and is influenced by nutritional state, food palatability, and social factors such as stress. However, the mechanism responsible for selection between a high-carbohydrate diet (HCD) and a high-fat diet (HFD) remains unknown. Here, we show that activation of a subset of corticotropin-releasing hormone (CRH)-positive neurons in the rostral region of the paraventricular hypothalamus (PVH) induces selection of an HCD over an HFD in mice during refeeding after fasting, resulting in a rapid recovery from the change in ketone metabolism. These neurons manifest activation of AMP-activated protein kinase (AMPK) during food deprivation, and this activation is necessary and sufficient for selection of an HCD over an HFD. Furthermore, this effect is mediated by carnitine palmitoyltransferase 1c (CPT1c). Thus, our results identify the specific neurons and intracellular signaling pathway responsible for regulation of the complex behavior of selection between an HCD and an HFD.
- Published
- 2018
- Full Text
- View/download PDF
9. Inhibition of Y1 receptor signaling improves islet transplant outcome
- Author
-
Kim Loh, Yan-Chuan Shi, Stacey Walters, Mohammed Bensellam, Kailun Lee, Katsuya Dezaki, Masanori Nakata, Chi Kin Ip, Jeng Yie Chan, Esteban N. Gurzov, Helen E. Thomas, Michaela Waibel, James Cantley, Thomas W. Kay, Toshihiko Yada, D. Ross Laybutt, Shane T. Grey, and Herbert Herzog
- Subjects
Science - Abstract
Islet transplantation is considered one of the potential treatments for T1DM but limited islet survival and their impaired function pose limitations to this approach. Here Loh et al. show that the Y1 receptor is expressed in β- cells and inhibition of its signalling, both genetic and pharmacological, improves mouse and human islet function.
- Published
- 2017
- Full Text
- View/download PDF
10. PDK1-FoxO1 pathway in AgRP neurons of arcuate nucleus promotes bone formation via GHRH-GH-IGF1 axis
- Author
-
Hideyuki Sasanuma, Masanori Nakata, Kumari Parmila, Jun Nakae, and Toshihiko Yada
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: In the hypothalamic arcuate nucleus (ARC), orexigenic agouti-related peptide (AgRP) neurons regulate feeding behavior and energy homeostasis, functions connected to bone metabolism. The 3-phosphoinositide-dependent protein kinase-1 (PDK1) serves as a major signaling molecule particularly for leptin and insulin in AgRP neurons. We asked whether PDK1 in AGRP neurons also contributes to bone metabolism. Methods: We generated AgRP neuron-specific PDK1 knockout (Agrp Pdk1−/−) mice and those with additional AgRP neuron-specific expression of transactivation-defective FoxO1 (Agrp Pdk1−/− Δ256Foxo1). Bone metabolism in KO and WT mice was analyzed by quantitative computed tomography (QCT), bone histomorphometry, measurement of plasma biomarkers, and qPCR analysis of peptides. Results: In Agrp Pdk1−/− female mice aged 6 weeks, compared with Agrp Cre mice, both stature and femur length were shorter while body weight was unchanged. Cortical bone mineral density (BMD) and cancellous BMD in the femur decreased, and bone formation was delayed. Furthermore, plasma GH and IGF-1 levels were reduced in parallel with decreased mRNA expressions for GH in pituitary and GHRH in ARC. Osteoblast activity was suppressed and osteoclast activity was enhanced. These changes in stature, BMD and GH level were rescued in Agrp Pdk1−/− Δ256Foxo1 mice, suggesting that the bone abnormalities and impaired GH release were mediated by enhanced Foxo1 due to deletion of PDK1. Conclusions: This study reveals a novel role of PDK1-Foxo1 pathway of AgRP neurons in controlling bone metabolism primarily via GHRH-GH-IGF-1 axis. Keywords: AgRP, GHRH, Growth hormone, PDK1, Foxo1, Bone mineralization
- Published
- 2017
- Full Text
- View/download PDF
11. Optogenetic activation of leptin- and glucose-regulated GABAergic neurons in dorsomedial hypothalamus promotes food intake via inhibitory synaptic transmission to paraventricular nucleus of hypothalamus
- Author
-
Zesemdorj Otgon-Uul, Shigetomo Suyama, Hiroshi Onodera, and Toshihiko Yada
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: The dorsomedial hypothalamus (DMH) has been considered an orexigenic nucleus, since the DMH lesion reduced food intake and body weight and induced resistance to diet-induced obesity. The DMH expresses feeding regulatory neuropeptides and receptors including neuropeptide Y (NPY), cocaine- and amphetamine-regulated transcript (CART), cholecystokinin (CCK), leptin receptor, and melanocortin 3/4 receptors. However, the principal neurons generating the orexigenic function in the DMH remain to be defined. This study aimed to clarify the role of the DMH GABAergic neurons in feeding regulation by using optogenetics and electrophysiological techniques. Methods: We generated the mice expressing ChRFR-C167A, a bistable chimeric channelrhodopsin, selectively in GABAergic neurons of DMH via locally injected adeno-associated virus 2. Food intake after optogenetic activation of DMH GABAergic neurons was measured. Electrophysiological properties of DMH GABAergic neurons were measured using slice patch clamp. Results: Optogenetic activation of DMH GABAergic neurons promoted food intake. Leptin hyperpolarized and lowering glucose depolarized half of DMH GABAergic neurons, suggesting their orexigenic property. Optical activation of axonal terminals of DMH GABAergic neurons at the paraventricular nucleus of hypothalamus (PVN), where anorexigenic neurons are localized, increased inhibitory postsynaptic currents on PVN neurons and promoted food intake. Conclusion: DMH GABAergic neurons are regulated by metabolic signals leptin and glucose and, once activated, promote food intake via inhibitory synaptic transmission to PVN. Keywords: Dorsomedial hypothalamus, GABAergic neuron, Feeding, Leptin, Glucose, Optogenetics
- Published
- 2016
- Full Text
- View/download PDF
12. Plasticity of calcium-permeable AMPA glutamate receptors in Pro-opiomelanocortin neurons
- Author
-
Shigetomo Suyama, Alexandra Ralevski, Zhong-Wu Liu, Marcelo O Dietrich, Toshihiko Yada, Stephanie E Simonds, Michael A Cowley, Xiao-Bing Gao, Sabrina Diano, and Tamas L Horvath
- Subjects
AMPA receptors ,POMC neurons ,calcium permeability ,plasticity ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
POMC neurons integrate metabolic signals from the periphery. Here, we show in mice that food deprivation induces a linear current-voltage relationship of AMPAR-mediated excitatory postsynaptic currents (EPSCs) in POMC neurons. Inhibition of EPSCs by IEM-1460, an antagonist of calcium-permeable (Cp) AMPARs, diminished EPSC amplitude in the fed but not in the fasted state, suggesting entry of GluR2 subunits into the AMPA receptor complex during food deprivation. Accordingly, removal of extracellular calcium from ACSF decreased the amplitude of mEPSCs in the fed but not the fasted state. Ten days of high-fat diet exposure, which was accompanied by elevated leptin levels and increased POMC neuronal activity, resulted in increased expression of Cp-AMPARs on POMC neurons. Altogether, our results show that entry of calcium via Cp-AMPARs is inherent to activation of POMC neurons, which may underlie a vulnerability of these neurons to calcium overload while activated in a sustained manner during over-nutrition.
- Published
- 2017
- Full Text
- View/download PDF
13. Sweet taste receptor serves to activate glucose- and leptin-responsive neurons in the hypothalamic arcuate nucleus and participates in glucose responsiveness.
- Author
-
Daisuke Kohno, Miho Koike, Yuzo Ninomiya, Itaru Kojima, Tadahiro Kitamura, and Toshihiko Yada
- Subjects
Glucose ,Leptin ,feeding ,POMC ,Sucralose ,Sweet taste receptor ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The hypothalamic feeding center plays an important role in energy homeostasis. In the feeding center, whole-body energy signals including hormones and nutrients are sensed, processed, and integrated. As a result, food intake and energy expenditure are regulated. Two types of glucose-sensing neurons exist in the hypothalamic arcuate nucleus (ARC): glucose-excited neurons and glucose-inhibited neurons. While some molecules are known to be related to glucose sensing in the hypothalamus, the mechanism underlying glucose sensing in the hypothalamus are not fully understood. The sweet taste receptor is a heterodimer of taste type 1 receptor 2 (T1R2) and taste type 1 receptor 3 (T1R3) and senses sweet tastes. T1R2 and T1R3 receptors are distributed in multiple organs including the tongue, pancreas, adipose tissue, and hypothalamus. However, the role of sweet taste receptors in the ARC remains to be clarified. To examine the role of sweet taste receptors in the ARC, cytosolic Ca2+ concentration ([Ca2+]i) in isolated single ARC neurons were measured using Fura-2 fluorescent imaging. An artificial sweetener, sucralose at 10-5 M-10-2 M dose dependently increased [Ca2+]i in 12-16% of ARC neurons. The sucralose-induced [Ca2+]i increase was suppressed by a sweet taste receptor inhibitor, gurmarin. The sucralose-induced [Ca2+]i increase was inhibited under an extracellular Ca2+-free condition and in the presence of an L-type Ca2+ channel blocker, nitrendipine. Sucralose-responding neurons were activated by high-concentration of glucose. This response to glucose was markedly suppressed by gurmarin. More than half of sucralose-responding neurons were activated by leptin but not ghrelin. Percentage of proopiomelanocortin (POMC) neurons among sucralose-responding neurons and sweet taste receptor expressing neurons were low, suggesting that majority of sucralose-responding neurons are non-POMC neurons. These data suggest that sweet taste receptor-mediated cellular activation mainly occurs on non-POMC leptin-responding neurons and contributes to glucose responding. Endogenous sweet molecules including glucose may regulate energy homeostasis through sweet taste receptors on glucose-and leptin-responsive neurons in the ARC.
- Published
- 2016
- Full Text
- View/download PDF
14. High-Fat Diet Augments VPAC1 Receptor-Mediated PACAP Action on the Liver, Inducing LAR Expression and Insulin Resistance
- Author
-
Masanori Nakata, Boyang Zhang, Yifei Yang, Takashi Okada, Norihito Shintani, Hitoshi Hashimoto, and Toshihiko Yada
- Subjects
Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) acts on multiple processes of glucose and energy metabolism. PACAP potentiates insulin action in adipocytes and insulin release from pancreatic β-cells, thereby enhancing glucose tolerance. Contrary to these effects at organ levels, PACAP null mice exhibit hypersensitivity to insulin. However, this apparent discrepancy remains to be solved. We aimed to clarify the mechanism underlying the antidiabetic phenotype of PACAP null mice. Feeding with high-fat diet (HFD) impaired insulin sensitivity and glucose tolerance in wild type mice, whereas these changes were prevented in PACAP null mice. HFD also impaired insulin-induced Akt phosphorylation in the liver in wild type mice, but not in PACAP null mice. Using GeneFishing method, HFD increased the leukocyte common antigen-related (LAR) protein tyrosine phosphatase in the liver in wild type mice. Silencing of LAR restored the insulin signaling in the liver of HFD mice. Moreover, the increased LAR expression by HFD was prevented in PACAP null mice. HFD increased the expression of VPAC1 receptor (VPAC1-R), one of three PACAP receptors, in the liver of wild type mice. These data indicate that PACAP-VPAC1-R signaling induces LAR expression and insulin resistance in the liver of HFD mice. Antagonism of VPAC1-R may prevent progression of HFD-induced insulin resistance in the liver, providing a novel antidiabetic strategy.
- Published
- 2016
- Full Text
- View/download PDF
15. Markedly Reduced White Adipose Tissue and Increased Insulin Sensitivity in Adcyap1-Deficient Mice
- Author
-
Shuhei Tomimoto, Tatsuya Ojika, Norihito Shintani, Hitoshi Hashimoto, Ken-ichi Hamagami, Kazuya Ikeda, Masanori Nakata, Toshihiko Yada, Yusuke Sakurai, Takeshi Shimada, Yoshiko Morita, Chie Ishida, and Akemichi Baba
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide implicated in several metabolic functions, including insulin secretion and sympathoadrenal activation. To clarify the roles of PACAP in maintenance of whole-body glucose and lipid homeostasis, the impact of the deletion of PACAP on glucose homeostasis, body weight, and adipose tissue mass was examined by comparing mice lacking the Adcyap1 gene encoding PACAP (Adcyap1−/−) with wild-type littermate controls. Adcyap1−/− mice showed significant hypoinsulinemia, although being normoglycemic, and lower body weight as well as reduced food intake. They also showed greatly reduced white adipose tissue mass, in which the mRNA expression of adipocyte fatty acid-binding protein (aP2), a marker of adipocyte differentiation, was decreased. Glucose and insulin tolerance tests revealed increased insulin sensitivity in Adcyap1−/− mice. In accordance with these observations, plasma levels of resistin, an adipocytokine implicated in insulin resistance, were decreased in Adcyap1−/− mice. After a high-fat dietary challenge for six weeks, Adcyap1−/− mice still showed lower body weights and increased insulin sensitivity. These results indicate the crucial roles of PACAP in energy metabolism, including lipid metabolism, and in the regulation of body weight, raising the possibility that the PACAP-signaling pathway that favors energy storage could be a therapeutic target for obesity. Keywords:: adipose tissue, body weight, insulin sensitivity, pituitary adenylate cyclase-activating polypeptide (PACAP), resistin
- Published
- 2008
- Full Text
- View/download PDF
16. Orexin Receptor Type-1 Couples Exclusively to Pertussis Toxin-Insensitive G-Proteins, While Orexin Receptor Type-2 Couples to Both Pertussis Toxin-Sensitive and -Insensitive G-Proteins
- Author
-
Yun Zhu, Yoshihiro Miwa, Akihiro Yamanaka, Toshihiko Yada, Megumi Shibahara, Yoichiro Abe, Takeshi Sakurai, and Katsutoshi Goto
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Signal transduction pathways of orexin receptors were examined using a nerve-like cell line transfected with orexin receptor type-1 (OX1R) and orexin receptor type-2 (OX2R). Forskolin-stimulated cyclic adenosine 3,5-monophosphate (cAMP) accumulation in OX2R-expressing cells was inhibited by orexin in a dose-dependent manner, and the effect was abolished by pretreatment with pertussis toxin (PTX). The inhibitory effect of orexin on forskolin-stimulated cAMP accumulation was not observed in OX1R-expressing cells. Administration of orexin to these cells resulted in a transient increase of intracellular calcium concentration ([Ca2+]i). Orexin-stimulated increases in [Ca2+]i in OX1R- or OX2R-expressing cells were not affected by the PTX pretreatment. These observations suggest that OX1R couples exclusively to PTX-insensitive G-proteins, while OX2R couples to both PTX-sensitive and -insensitive G-proteins. To examine the relative contributions of these G-proteins in OX2R-mediated activation of neurons, we used histaminergic tuberomammillary nucleus neurons, in which OX2R is abundantly expressed. We found that a phospholipase C (PLC)-inhibitor, U73122, inhibits orexin-mediated neuronal activation, but PTX showed no effect on it. This suggests that although OX2R couples to multiple G-proteins, activation of neurons by orexins through OX2R is mediated via a PTX-insensitive, PLC dependent pathway.
- Published
- 2003
- Full Text
- View/download PDF
17. Insulin Activates Vagal Afferent Neurons Including those Innervating Pancreas via Insulin Cascade and Ca(2+) Influx: Its Dysfunction in IRS2-KO Mice with Hyperphagic Obesity.
- Author
-
Yusaku Iwasaki, Kenju Shimomura, Daisuke Kohno, Katsuya Dezaki, Enkh-Amar Ayush, Hajime Nakabayashi, Naoto Kubota, Takashi Kadowaki, Masafumi Kakei, Masanori Nakata, and Toshihiko Yada
- Subjects
Medicine ,Science - Abstract
Some of insulin's functions, including glucose/lipid metabolism, satiety and neuroprotection, involve the alteration of brain activities. Insulin could signal to the brain via penetrating through the blood-brain barrier and acting on the vagal afferents, while the latter remains unproved. This study aimed to clarify whether insulin directly regulates the nodose ganglion neurons (NGNs) of vagal afferents in mice. NGs expressed insulin receptor (IR) and insulin receptor substrate-2 (IRS2) mRNA, and some of NGNs were immunoreactive to IR. In patch-clamp and fura-2 microfluorometric studies, insulin (10(-12)∼10(-6) M) depolarized and increased cytosolic Ca(2+) concentration ([Ca(2+)]i) in single NGNs. The insulin-induced [Ca(2+)]i increases were attenuated by L- and N-type Ca(2+) channel blockers, by phosphatidylinositol 3 kinase (PI3K) inhibitor, and in NGNs from IRS2 knockout mice. Half of the insulin-responsive NGNs contained cocaine- and amphetamine-regulated transcript. Neuronal fibers expressing IRs were distributed in/around pancreatic islets. The NGNs innervating the pancreas, identified by injecting retrograde tracer into the pancreas, responded to insulin with much greater incidence than unlabeled NGNs. Insulin concentrations measured in pancreatic vein was 64-fold higher than that in circulation. Elevation of insulin to 10(-7) M recruited a remarkably greater population of NGNs to [Ca(2+)]i increases. Systemic injection of glibenclamide rapidly released insulin and phosphorylated AKT in NGs. Furthermore, in IRS2 knockout mice, insulin action to suppress [Ca(2+)]i in orexigenic ghrelin-responsive neurons in hypothalamic arcuate nucleus was intact while insulin action on NGN was markedly attenuated, suggesting a possible link between impaired insulin sensing by NGNs and hyperphagic obese phenotype in IRS2 knockout mice These data demonstrate that insulin directly activates NGNs via IR-IRS2-PI3K-AKT-cascade and depolarization-gated Ca(2+) influx. Pancreas-innervating NGNs may effectively sense dynamic changes of insulin released in response to nutritional states. These interactions could serve to convey the changes in pancreatic and systemic insulin to the brain.
- Published
- 2013
- Full Text
- View/download PDF
18. PDK1-Foxo1 in agouti-related peptide neurons regulates energy homeostasis by modulating food intake and energy expenditure.
- Author
-
Yongheng Cao, Masanori Nakata, Shiki Okamoto, Eisuke Takano, Toshihiko Yada, Yasuhiko Minokoshi, Yukio Hirata, Kazunori Nakajima, Kristy Iskandar, Yoshitake Hayashi, Wataru Ogawa, Gregory S Barsh, Hiroshi Hosoda, Kenji Kangawa, Hiroshi Itoh, Tetsuo Noda, Masato Kasuga, and Jun Nakae
- Subjects
Medicine ,Science - Abstract
Insulin and leptin intracellular signaling pathways converge and act synergistically on the hypothalamic phosphatidylinositol-3-OH kinase/3-phosphoinositide-dependent protein kinase 1 (PDK1). However, little is known about whether PDK1 in agouti-related peptide (AGRP) neurons contributes to energy homeostasis. We generated AGRP neuron-specific PDK1 knockout (AGRPPdk1(-/-)) mice and mice with selective expression of transactivation-defective Foxo1 (Δ256Foxo1(AGRP)Pdk1(-/-)). The AGRPPdk1(-/-) mice showed reductions in food intake, body length, and body weight. The Δ256Foxo1(AGRP)Pdk1(-/-) mice showed increased body weight, food intake, and reduced locomotor activity. After four weeks of calorie-restricted feeding, oxygen consumption and locomotor activity were elevated in AGRPPdk1(-/-) mice and reduced in Δ256Foxo1(AGRP)Pdk1(-/-) mice. In vitro, ghrelin-induced changes in [Ca(2+)](i) and inhibition of ghrelin by leptin were significantly attenuated in AGRPPdk1(-/-) neurons compared to control neurons. However, ghrelin-induced [Ca(2+)](i) changes and leptin inhibition were restored in Δ256Foxo1(AGRP)Pdk1(-/-) mice. These results suggested that PDK1 and Foxo1 signaling pathways play important roles in the control of energy homeostasis through AGRP-independent mechanisms.
- Published
- 2011
- Full Text
- View/download PDF
19. D-Allulose cooperates with glucagon-like peptide-1 and activates proopiomelanocortin neurons in the arcuate nucleus and central injection inhibits feeding in mice
- Author
-
Rakhat Yermek, Lei Wang, Kentaro Kaneko, Wanxin Han, Yutaka Seino, Daisuke Yabe, and Toshihiko Yada
- Subjects
Neurons ,Mice ,Pro-Opiomelanocortin ,Glucagon-Like Peptide 1 ,Arcuate Nucleus of Hypothalamus ,Biophysics ,Animals ,Fructose ,Obesity ,Cell Biology ,Molecular Biology ,Biochemistry - Abstract
A rare sugar D-Allulose has sweetness without calorie. Previous studies have shown that D-Allulose improves glucose and energy metabolism and ameliorates obesity. However, underlying mechanisms remain elusive. This study explored the effect of central injection of D-Allulose on feeding behavior in mice. We also examined direct effects of D-Allulose on the neurons in the hypothalamic arcuate nucleus (ARC) that regulate feeding, including the anorexigenic glucagon-like peptide-1 (GLP-1)-responsive neurons and proopiomelanocortin (POMC) neurons. Single neurons were isolated from ARC and cytosolic Ca
- Published
- 2022
- Full Text
- View/download PDF
20. Author response for 'Glucose‐dependent insulinotropic polypeptide counteracts diet‐induced obesity along with reduced feeding, elevated plasma leptin and activation of leptin‐responsive and proopiomelanocortin neurons in the arcuate nucleus'
- Author
-
null Wanxin Han, null Lei Wang, null Kento Ohbayashi, null Masakazu Takeuchi, null Libbey O'Farrell, null Tamer Coskun, null Yermek Rakhat, null Daisuke Yabe, null Yusaku Iwasaki, null Yutaka Seino, and null Toshihiko Yada
- Published
- 2023
- Full Text
- View/download PDF
21. d-Allulose Inhibits Ghrelin-Responsive, Glucose-Sensitive and Neuropeptide Y Neurons in the Arcuate Nucleus and Central Injection Suppresses Appetite-Associated Food Intake in Mice
- Author
-
Yermek Rakhat, Kentaro Kaneko, Lei Wang, Wanxin Han, Yutaka Seino, Daisuke Yabe, and Toshihiko Yada
- Subjects
Neurons ,Nutrition and Dietetics ,d<%2Fspan>-allulose%22">d-allulose ,ghrelin ,glucose ,arcuate nucleus ,neuropeptide Y ,cytosolic Ca2+, appetite ,hunger ,food intake ,obesity ,diabetes ,Arcuate Nucleus of Hypothalamus ,Appetite ,Fructose ,Hyperphagia ,Ghrelin ,Rats ,Rats, Sprague-Dawley ,Eating ,Mice ,Glucose ,Animals ,Neuropeptide Y ,Obesity ,Food Science - Abstract
d-allulose, a rare sugar, has sweetness with few calories. d-allulose regulates feeding and glycemia, and ameliorates hyperphagia, obesity and diabetes. All these functions involve the central nervous system. However, central mechanisms underlying these effects of d-allulose remain unknown. We recently reported that d-allulose activates the anorexigenic neurons in the hypothalamic arcuate nucleus (ARC), the neurons that respond to glucagon-like peptide-1 and that express proopiomelanocortin. However, its action on the orexigenic neurons remains unknown. This study investigated the effects of d-allulose on the ARC neurons implicated in hunger, by measuring cytosolic Ca2+ concentration ([Ca2+]i) in single neurons. d-allulose depressed the increases in [Ca2+]i induced by ghrelin and by low glucose in ARC neurons and inhibited spontaneous oscillatory [Ca2+]i increases in neuropeptide Y (NPY) neurons. d-allulose inhibited 10 of 35 (28%) ghrelin-responsive, 18 of 60 (30%) glucose-sensitive and 3 of 8 (37.5%) NPY neurons in ARC. Intracerebroventricular injection of d-allulose inhibited food intake at 20:00 and 22:00, the early dark phase when hunger is promoted. These results indicate that d-allulose suppresses hunger-associated feeding and inhibits hunger-promoting neurons in ARC. These central actions of d-allulose represent the potential of d-allulose to inhibit the hyperphagia with excessive appetite, thereby counteracting obesity and diabetes.
- Published
- 2022
- Full Text
- View/download PDF
22. 1,5-Anhydro-D-Fructose Exhibits Satiety Effects via the Activation of Oxytocin Neurons in the Paraventricular Nucleus
- Author
-
Masanori Nakata, Yuto Yamaguchi, Hikaru Monnkawa, Midori Takahashi, Boyang Zhang, Putra Santoso, Toshihiko Yada, and Ikuro Maruyama
- Subjects
1,5-Anhydro-D-fructose ,oxytocin ,feeding behavior ,PVN ,Inorganic Chemistry ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
1,5-Anhydro-D-fructose (1,5-AF) is a bioactive monosaccharide that is produced by the glycogenolysis in mammalians and is metabolized to 1,5-anhydro-D-glucitol (1,5-AG). 1,5-AG is used as a marker of glycemic control in diabetes patients. 1,5-AF has a variety of physiological activities, but its effects on energy metabolism, including feeding behavior, are unclarified. The present study examined whether 1,5-AF possesses the effect of satiety. Peroral administration of 1,5-AF, and not of 1,5-AG, suppressed daily food intake. Intracerebroventricular (ICV) administration of 1,5-AF also suppressed feeding. To investigate the neurons targeted by 1,5-AF, we investigated c-Fos expression in the hypothalamus and brain stem. ICV injection of 1,5-AF significantly increased c-Fos positive oxytocin neurons and mRNA expression of oxytocin in the paraventricular nucleus (PVN). Moreover, 1,5-AF increased cytosolic Ca2+ concentration of oxytocin neurons in the PVN. Furthermore, the satiety effect of 1,5-AF was abolished in oxytocin knockout mice. These findings reveal that 1,5-AF activates PVN oxytocin neurons to suppress feeding, indicating its potential as the energy storage monitoring messenger to the hypothalamus for integrative regulation of energy metabolism.
- Published
- 2023
- Full Text
- View/download PDF
23. TRPV1-Mediated Sensing of Sodium and Osmotic Pressure in POMC Neurons in the Arcuate Nucleus of the Hypothalamus
- Author
-
Boyang Zhang, Kazuomi Kario, Toshihiko Yada, and Masanori Nakata
- Subjects
Neurons ,endocrine system ,Nutrition and Dietetics ,Pro-Opiomelanocortin ,digestive, oral, and skin physiology ,melanocortin system ,AgRP ,POMC ,sodium sensing ,TRPV1 ,blood pressure ,Sodium ,Arcuate Nucleus of Hypothalamus ,Hypothalamus ,TRPV Cation Channels ,Mice, Inbred C57BL ,Mice ,nervous system ,Osmotic Pressure ,Animals ,Agouti-Related Protein ,hormones, hormone substitutes, and hormone antagonists ,Food Science - Abstract
The central melanocortin system conducted by anorexigenic pro-opiomelanocortin (POMC) neurons and orexigenic agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARC) not only regulates feeding behavior but also blood pressure. Excessive salt intake raises the Na+ concentration ([Na+]) in the cerebrospinal fluid (CSF) and worsens hypertension. The blood–brain barrier is immature in the ARC. Therefore, both AgRP and POMC neurons in the ARC have easy access to the electrolytes in the blood and can sense changes in their concentrations. However, the sensitivity of AgRP and POMC neurons to Na+ remains unclear. This study aimed to explore how the changes in the extracellular Na+ concentration ([Na+]) influence these neurons by measuring the cytosolic Ca2+ concentration ([Ca2+]i) in the single neurons isolated from the ARC that were subsequently immunocytochemically identified as AgRP or POMC neurons. Both AgRP and POMC neurons responded to increases in both [Na+] and osmolarity in C57BL/6 mice. In contrast, in transient receptor potential vanilloid 1 (TRPV1) knockout (KO) mice, POMC neurons failed to respond to increases in both [Na+] and osmolarity, while they responded to high glucose and angiotensin II levels with increases in [Ca2+]i. Moreover, in KO mice fed a high-salt diet, the expression of POMC was lower than that in wild-type mice. These results demonstrate that changes in [Na+] and osmolarity are sensed by the ARC POMC neurons via the TRPV1-dependent mechanism.
- Published
- 2022
24. The liver–brain–gut neural arc maintains the Treg cell niche in the gut
- Author
-
Po Sung Chu, Masayuki Inoue, Yuya Hagihara, Takaharu Okada, Akihiko Yoshimura, Mamoru Tanida, Yosuke Harada, Harumichi Ishigame, Minoru Matsui, Toshihiko Yada, Yusaku Iwasaki, Kentaro Miyamoto, Hideyuki Okano, Takanori Kanai, Wataru Suda, Nobuhito Taniki, Shinsuke Shibata, Nobuhiro Nakamoto, Yuko Kitagawa, Yohei Mikami, Masahira Hattori, Toshiaki Teratani, Tomohisa Sujino, Keita Kohno, Takahiro Suzuki, Koji Okabayashi, and Makoto Tsuda
- Subjects
0301 basic medicine ,Multidisciplinary ,Liver cytology ,Reflex arc ,digestive, oral, and skin physiology ,Central nervous system ,Biology ,Gut flora ,biology.organism_classification ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Muscarinic acetylcholine receptor ,medicine ,Brainstem ,Receptor ,030217 neurology & neurosurgery ,Homeostasis - Abstract
Recent clinical and experimental evidence has evoked the concept of the gut–brain axis to explain mutual interactions between the central nervous system and gut microbiota that are closely associated with the bidirectional effects of inflammatory bowel disease and central nervous system disorders1–4. Despite recent advances in our understanding of neuroimmune interactions, it remains unclear how the gut and brain communicate to maintain gut immune homeostasis, including in the induction and maintenance of peripheral regulatory T cells (pTreg cells), and what environmental cues prompt the host to protect itself from development of inflammatory bowel diseases. Here we report a liver–brain–gut neural arc that ensures the proper differentiation and maintenance of pTreg cells in the gut. The hepatic vagal sensory afferent nerves are responsible for indirectly sensing the gut microenvironment and relaying the sensory inputs to the nucleus tractus solitarius of the brainstem, and ultimately to the vagal parasympathetic nerves and enteric neurons. Surgical and chemical perturbation of the vagal sensory afferents at the hepatic afferent level reduced the abundance of colonic pTreg cells; this was attributed to decreased aldehyde dehydrogenase (ALDH) expression and retinoic acid synthesis by intestinal antigen-presenting cells. Activation of muscarinic acetylcholine receptors directly induced ALDH gene expression in both human and mouse colonic antigen-presenting cells, whereas genetic ablation of these receptors abolished the stimulation of antigen-presenting cells in vitro. Disruption of left vagal sensory afferents from the liver to the brainstem in mouse models of colitis reduced the colonic pTreg cell pool, resulting in increased susceptibility to colitis. These results demonstrate that the novel vago-vagal liver–brain–gut reflex arc controls the number of pTreg cells and maintains gut homeostasis. Intervention in this autonomic feedback feedforward system could help in the development of therapeutic strategies to treat or prevent immunological disorders of the gut. A liver–brain–gut neural circuit responds to the gut microenvironment and regulates the activity of peripheral regulatory T cells in the colon by controlling intestinal antigen-presenting cells in a muscarinic signalling-dependent manner.
- Published
- 2020
- Full Text
- View/download PDF
25. Circadian Clock Component BMAL1 in the Paraventricular Nucleus Regulates Glucose Metabolism
- Author
-
Masanori Nakata, Parmila Kumari, Rika Kita, Nanako Katsui, Yuriko Takeuchi, Tomoki Kawaguchi, Toshiya Yamazaki, Boyang Zhang, Shigeki Shimba, and Toshihiko Yada
- Subjects
endocrine system ,vasopressin ,glucose metabolism ,Article ,Mice ,Circadian Clocks ,Glucose Intolerance ,Animals ,Insulin ,insulin release ,TX341-641 ,RNA, Messenger ,Mice, Knockout ,Neurons ,Nutrition and Dietetics ,Nutrition. Foods and food supply ,urogenital system ,paraventricular nucleus ,circadian ,BMAL1 ,digestive, oral, and skin physiology ,ARNTL Transcription Factors ,Arginine Vasopressin ,Glucose ,Gene Expression Regulation ,nervous system ,hormones, hormone substitutes, and hormone antagonists ,Food Science ,Paraventricular Hypothalamic Nucleus - Abstract
It is suggested that clock genes link the circadian rhythm to glucose and lipid metabolism. In this study, we explored the role of the clock gene Bmal1 in the hypothalamic paraventricular nucleus (PVN) in glucose metabolism. The Sim1-Cre-mediated deletion of Bmal1 markedly reduced insulin secretion, resulting in impaired glucose tolerance. The pancreatic islets’ responses to glucose, sulfonylureas (SUs) and arginine vasopressin (AVP) were well maintained. To specify the PVN neuron subpopulation targeted by Bmal1, the expression of neuropeptides was examined. In these knockout (KO) mice, the mRNA expression of Avp in the PVN was selectively decreased, and the plasma AVP concentration was also decreased. However, fasting suppressed Avp expression in both KO and Cre mice. These results demonstrate that PVN BMAL1 maintains Avp expression in the PVN and release to the circulation, possibly providing islet β-cells with more AVP. This action helps enhance insulin release and, consequently, glucose tolerance. In contrast, the circadian variation of Avp expression is regulated by feeding, but not by PVN BMAL1.
- Published
- 2021
- Full Text
- View/download PDF
26. Coexistence of sensory qualities and value representations in human orbitofrontal cortex
- Author
-
Takaaki Yoshimoto, Shuntaro Okazaki, Motofumi Sumiya, Haruka K. Takahashi, Eri Nakagawa, Takahiko Koike, Ryo Kitada, Shiki Okamoto, Masanori Nakata, Toshihiko Yada, Hirotaka Kosaka, Norihiro Sadato, and Junichi Chikazoe
- Subjects
Reward ,General Neuroscience ,Humans ,Prefrontal Cortex ,General Medicine ,Magnetic Resonance Imaging - Abstract
Despite the multiple regions and neural networks associated with value-based decision-making, the orbitofrontal cortex (OFC) is possible a particularly important one. Although the role of the OFC in reinforcer devaluation tasks, which assess the ability to represent identity, sensory qualities, and subjective values of the expected outcomes, has been established, the specific aspect represented in this area remains unclear. In this study, using functional magnetic resonance imaging, wherein participants rated the palatability of 128 food items using photographs, we investigated whether the human OFC represents object identity, sensory qualities, or value. Employing many items helped us dissociate object identity from sensory qualities and values; the inferred sensory qualities of identical items were manipulated by a change in metabolic state. Moreover, value differences between items were analytically controlled by employing a technique similar to age adjustment. The palatability ratings for food items significantly decreased after a meal. Using representational similarity analysis, we confirmed that the OFC represents value. Moreover, identical items were represented similarly in the lateral OFC in a given metabolic state; however, these representations were altered post-feeding. Importantly, this change was not explained by subjective value, suggesting that the OFC represents sensory quality and value, but not object identity.
- Published
- 2021
27. Status of ghrelin as an islet hormone and paracrine/autocrine regulator of insulin secretion
- Author
-
Toshihiko Yada and Katsuya Dezaki
- Subjects
endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Physiology ,medicine.medical_treatment ,Regulator ,Biochemistry ,Cellular and Molecular Neuroscience ,Paracrine signalling ,Islets of Langerhans ,Endocrinology ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Humans ,Insulin ,Autocrine signalling ,Receptors, Ghrelin ,geography ,geography.geographical_feature_category ,Chemistry ,Pancreatic islets ,digestive, oral, and skin physiology ,Islet ,Ghrelin ,medicine.anatomical_structure ,hormones, hormone substitutes, and hormone antagonists ,Hormone - Abstract
Ghrelin is expressed in the pancreatic islet cells as well as the stomach. In the perfused pancreas and isolated islets, GHS-R antagonism, ghrelin immunoneutralization and ghrelin-knockout (Ghr-KO) all increase glucose-induced insulin release. Thus, pharmacological, immunological and genetic blockades of ghrelin in the pancreatic islets all markedly augment glucose-induced insulin release, showing that islet-derived ghrelin physiologically restricts insulin release in rodents. In this review, we focus on the current understanding of the following key questions: 1) from which islet cells ghrelin is released, 2) on which islet cells ghrelin acts, and 3) mechanisms by which the islet-derived ghrelin inhibits insulin secretion.
- Published
- 2021
28. Central Glucagon-like Peptide-1 Receptor Signaling via Brainstem Catecholamine Neurons Counteracts Hypertension in Spontaneously Hypertensive Rats
- Author
-
Toshihiko Yada, Yuko Maejima, Neeru M. Sharma, Boyang Zhang, Masanori Nakata, Kazuomi Kario, Kenichi Katsurada, Toshinobu Saito, Kaushik K. Patel, and Shyam Sundar Nandi
- Subjects
Agonist ,Male ,medicine.medical_specialty ,Saporin ,medicine.drug_class ,lcsh:Medicine ,Dopamine beta-Hydroxylase ,030204 cardiovascular system & hematology ,Rats, Inbred WKY ,Glucagon-Like Peptide-1 Receptor ,Article ,03 medical and health sciences ,Norepinephrine ,0302 clinical medicine ,Dopamine ,Internal medicine ,Rats, Inbred SHR ,medicine ,Animals ,lcsh:Science ,Multidisciplinary ,biology ,Liraglutide ,business.industry ,Dopaminergic Neurons ,Area postrema ,lcsh:R ,Glucagon-like peptide-1 ,Metabolic syndrome ,Rats ,Endocrinology ,nervous system ,Hypertension ,Catecholamine ,biology.protein ,lcsh:Q ,business ,030217 neurology & neurosurgery ,medicine.drug ,Brain Stem ,Signal Transduction ,Neuroscience ,circulatory and respiratory physiology - Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists, widely used to treat type 2 diabetes, reduce blood pressure (BP) in hypertensive patients. Whether this action involves central mechanisms is unknown. We here report that repeated lateral ventricular (LV) injection of GLP-1R agonist, liraglutide, once daily for 15 days counteracted the development of hypertension in spontaneously hypertensive rats (SHR). In parallel, it suppressed urinary norepinephrine excretion, and induced c-Fos expressions in the area postrema (AP) and nucleus tractus solitarius (NTS) of brainstem including the NTS neurons immunoreactive to dopamine beta-hydroxylase (DBH). Acute administration of liraglutide into fourth ventricle, the area with easy access to the AP and NTS, transiently decreased BP in SHR and this effect was attenuated after lesion of NTS DBH neurons with anti-DBH conjugated to saporin (anti-DBH-SAP). In anti-DBH-SAP injected SHR, the antihypertensive effect of repeated LV injection of liraglutide for 14 days was also attenuated. These findings demonstrate that the central GLP-1R signaling via NTS DBH neurons counteracts the development of hypertension in SHR, accompanied by attenuated sympathetic nerve activity.
- Published
- 2019
- Full Text
- View/download PDF
29. Ninjin-yoeito activates ghrelin-responsive and unresponsive NPY neurons in the arcuate nucleus and counteracts cisplatin-induced anorexia
- Author
-
Katsuya Dezaki, Akio Inui, Yutaka Seino, Toshihiko Yada, Lei Wang, and Chayon Goswami
- Subjects
Male ,Modern medicine ,medicine.medical_specialty ,media_common.quotation_subject ,Antineoplastic Agents ,030209 endocrinology & metabolism ,Anorexia ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Endocrinology ,Arcuate nucleus ,Internal medicine ,medicine ,Animals ,Neuropeptide Y ,media_common ,Neurons ,Arc (protein) ,Endocrine and Autonomic Systems ,business.industry ,Arcuate Nucleus of Hypothalamus ,Appetite ,General Medicine ,Neuropeptide Y receptor ,Ghrelin ,Neurology ,Anorectic ,Cisplatin ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Drugs, Chinese Herbal - Abstract
Reduced appetite or anorexia substantially deteriorates quality of life in various diseases including cancer, depression and heart failure. Furthermore, reduced appetite may stand upstream of sarcopenia and frailty. All these diseases are heavy burdens in the modern medicine and society. Therefore, the means that counteracts reduced appetite has been awaited, however, effective and well evidenced substance is not currently available. Ninjin-yoeito, a Japanese kampo medicine comprising twelve herbs has been used to treat anorexia. However, underlying mechanism is little known. Neuropeptide Y (NPY) and ghrelin are the most potent central and peripheral inducers of appetite, respectively. This study sought to determine whether Ninjin-yoeito influences NPY and/or ghrelin-responsive neurons in the hypothalamic arcuate nucleus (ARC), a feeding center. We isolated single neurons from ARC of mice and measured cytosolic Ca2+ concentration ([Ca2+]i) with fura-2 fluorescence imaging, followed by immunocytochemical identification of NPY neurons. Ninjin-yoeito (1–10 μg/ml) increased [Ca2+]i in ARC neurons, the majority (80%) of which was immunoreactive to NPY. One fraction of these Ninjin-yoeito-responsive NPY neurons also responded to ghrelin, while another fraction did not. Furthermore, oral administration of Ninjin-yoeito (1 g/kg/day) counteracted the reductions in food intake and body weight by cisplatin, an anti-cancer drug, in mice. These results demonstrate that Ninjin-yoeito directly targets both ghrelin-responsive and unresponsive NPY neurons in ARC and preserves food intake and body weight in cisplatin-treated anorectic mice. Ninjin-yoeito's signaling through ghrelin-responsive and ghrelin-unresponsive NPY pathways may provide strong mechanistic basis for this medicine for treating anorectic conditions associated with cancer, depression, heart failure, sarcopenia, frailty and aging.
- Published
- 2019
- Full Text
- View/download PDF
30. Onion component, isoalliin, stimulates feeding and activates the arcuate nucleus neuropeptide Y, ghrelin- and Ninjin'yoeito-responsive neurons
- Author
-
Toshihiko Yada, Yusaku Iwasaki, Lei Wang, Wanxin Han, Geoffrey W. G. Sharp, Rakhat Yermek, and Yutaka Seino
- Subjects
medicine.medical_specialty ,media_common.quotation_subject ,Anorexia ,Cellular and Molecular Neuroscience ,Eating ,Mice ,Endocrinology ,Arcuate nucleus ,Orexigenic ,Internal medicine ,medicine ,Animals ,Neuropeptide Y ,Cysteine ,media_common ,Neurons ,Arc (protein) ,Dose-Response Relationship, Drug ,Endocrine and Autonomic Systems ,Chemistry ,Arcuate Nucleus of Hypothalamus ,Appetite ,General Medicine ,Neuropeptide Y receptor ,Ghrelin ,medicine.anatomical_structure ,Neurology ,Calcium ,Neuron ,medicine.symptom ,medicine.drug ,Drugs, Chinese Herbal - Abstract
Appetite loss or anorexia substantially decreases the quality of life in patients with cancer, depression and gastrointestinal disorders, and can lead to sarcopenia and frailty. Foods that restore appetite have been sought-for but are not currently available. Historically, onion intake was adopted to treat a variety of diseases with reduced appetite including cancer and gastrointestinal disturbances. While isoalliin is a core component of onion, the effects of isoalliin on feeding behavior and feeding centers remain unknown. Neuropeptide Y (NPY) and ghrelin are the most potent central and peripheral inducers of appetite. A Japanese kampo medicine Ninjin'yoeito activates ghrelin-responsive NPY neurons in the hypothalamic arcuate nucleus (ARC) and counteracts anorexia induced by an anti-cancer drug cisplatin. This study explored the effects of isoalliin on feeding behavior and activities of ARC neurons in mice. Isoalliin, injected intraperitoneally, dose-dependently increased food intake during dark phase (DP) and daily without altering light phase (LP) food intake. We measured cytosolic Ca2+ concentration ([Ca2+]i) in single ARC neurons including NPY neurons identified by GFP fluorescence. Isoalliin increased [Ca2+]i in 10 of 18 (55.6%) NPY neurons, a majority of which also responded to ghrelin with [Ca2+]i increases, indicating that the ARC ghrelin-responsive NPY neuron is the major target of isoalliin. Isoalliin also increased [Ca2+]i in the ARC neurons that responded to Ninjin'yoeito. These results indicate that isoalliin enhances feeding at the active period and activates ARC ghrelin-responsive NPY neurons and Ninjin'yoeito-responsive neurons. These abilities of isoalliin to stimulate DP feeding and activate ARC orexigenic neurons provide scientific evidence for the health beneficial effects of onion experienced historically and globally.
- Published
- 2021
31. Peripheral circadian rhythms in the liver and white adipose tissue of mice are attenuated by constant light and restored by time-restricted feeding
- Author
-
Toshihiko Yada, Manabu Takahashi, Yusaku Iwasaki, Shoko Takei, Tetsuji Wakabayashi, Ken Ebihara, Daisuke Yamamuro, Kent Sakai, Shun Ishibashi, Akihito Takei, Shuichi Nagashima, and Hisataka Yamazaki
- Subjects
0301 basic medicine ,Male ,Light ,Physiology ,Adipose tissue ,Gene Expression ,White adipose tissue ,Biochemistry ,Mice ,0302 clinical medicine ,Gene expression ,Medicine and Health Sciences ,photoperiodism ,Multidisciplinary ,Circadian Rhythm Signaling Peptides and Proteins ,Liver Diseases ,Fatty liver ,Fatty Acids ,Fasting ,Lipids ,Circadian Rhythm ,CLOCK ,Circadian Rhythms ,Circadian Oscillators ,Physiological Parameters ,Liver ,Models, Animal ,Medicine ,Genetic Oscillators ,Research Article ,medicine.medical_specialty ,Adipose Tissue, White ,Photoperiod ,Science ,Gastroenterology and Hepatology ,Biology ,03 medical and health sciences ,Internal medicine ,medicine ,Genetics ,Animals ,Circadian rhythm ,Body Weight ,Biology and Life Sciences ,Lipid metabolism ,medicine.disease ,Lipid Metabolism ,Fatty Liver ,030104 developmental biology ,Endocrinology ,Metabolism ,Gene Expression Regulation ,Chronobiology ,030217 neurology & neurosurgery - Abstract
Disturbance of circadian rhythms underlies various metabolic diseases. Constant light exposure (LL) is known to disrupt both central and peripheral circadian rhythms. Here, we attempted to determine whether the effects of LL are different between various peripheral tissues and whether time-restricted feeding restores the circadian rhythms especially in white adipose tissue (WAT). Six-week-old mice were subjected to three feeding regimes: ad libitum feeding under light/dark phase (LD), ad libitum feeding under LL cycle, and restricted feeding at night-time under LL cycle with a normal chow. After 3 weeks, we compared body weight, food intake, plasma levels of lipids and glucose, and the expression patterns of the clock genes and the genes involved in lipid metabolism in the liver and WAT. The mice kept under LL with or without time-restricted feeding were 5.2% heavier (p
- Published
- 2020
32. Suprachiasmatic vasopressin to paraventricular oxytocin neurocircuit in the hypothalamus relays light reception to inhibit feeding behavior
- Author
-
Toshihiko Yada, Masanori Nakata, Yoichi Ueta, and Putra Santoso
- Subjects
Male ,0301 basic medicine ,endocrine system ,Vasopressin ,medicine.medical_specialty ,Light ,Physiology ,Endocrinology, Diabetes and Metabolism ,Hypothalamus ,Paraventricular hypothalamic nucleus ,Biology ,Oxytocin ,03 medical and health sciences ,0302 clinical medicine ,Feeding behavior ,Physiology (medical) ,Internal medicine ,Neural Pathways ,medicine ,Animals ,Calcium Signaling ,Circadian rhythm ,Rats, Wistar ,Calcium signaling ,Neurons ,Suprachiasmatic nucleus ,digestive, oral, and skin physiology ,Feeding Behavior ,Circadian Rhythm ,Rats ,Arginine Vasopressin ,Inhibition, Psychological ,030104 developmental biology ,Endocrinology ,nervous system ,Receptors, Oxytocin ,Suprachiasmatic Nucleus ,Proto-Oncogene Proteins c-fos ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,Paraventricular Hypothalamic Nucleus ,medicine.drug - Abstract
Light synchronizes the body’s circadian rhythms by modulating the master clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. In modern lifestyles that run counter to normal circadian rhythms, the extended and/or irregular light exposure impairs circadian rhythms and, consequently, promotes feeding and metabolic disorders. However, the neuronal pathway through which light is coupled to feeding behavior is less elucidated. The present study employed the light exposure during the dark phase of the day in rats and observed its effect on neuronal activity and feeding behavior. Light exposure acutely suppressed food intake and elevated c-Fos expression in the AVP neurons of SCN and the oxytocin (Oxt) neurons of paraventricular nucleus (PVN) in the hypothalamus. The light-induced suppression of food intake was abolished by blockade of the Oxt receptor in the brain. Retrograde tracer analysis demonstrated the projection of SCN AVP neurons to the PVN. Furthermore, intracerebroventricular injection of AVP suppressed food intake and increased c-Fos in PVN Oxt neurons. Intra-PVN injection of AVP exerted a stronger anorexigenic effect than intracerebroventriclar injection. AVP also induced intracellular Ca2+ signaling and increased firing frequency in Oxt neurons in PVN slices. These results reveal the novel neurocircuit from SCN AVP to PVN Oxt that relays light reception to inhibition of feeding behavior. This light-induced neurocircuit may serve as a pathway for forming the circadian feeding rhythm and linking irregular light exposure to arrhythmic feeding and, consequently, obesity and metabolic diseases.
- Published
- 2018
- Full Text
- View/download PDF
33. Neuropeptide oxytocin enhances μ opioid receptor signaling as a positive allosteric modulator
- Author
-
Yuki Yoshida, Yuriko Fujii, Toshihiko Yada, Naoto Ishibashi, Shigeto Hirayama, Takumi Ogino, Hideaki Fujii, Sei Manabe, Yoshiyuki Meguro, Moeko Eto, Miki Nonaka, Minoru Narita, Yasuhito Uezono, Naohiro Sata, Kanako Miyano, and Yoichi Ueta
- Subjects
0301 basic medicine ,Allosteric modulator ,medicine.drug_class ,Receptors, Opioid, mu ,Neuropeptide ,Pharmacology ,Oxytocin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Allosteric Regulation ,Opioid receptor ,Cyclic AMP ,medicine ,Animals ,Humans ,Receptor ,Analgesics ,Chemistry ,Neuropeptides ,lcsh:RM1-950 ,Stimulation, Chemical ,DAMGO ,HEK293 Cells ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Morphine ,Molecular Medicine ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction ,medicine.drug - Abstract
Oxytocin (OT) is a 9-amine neuropeptide that plays an essential role in mammalian labor, lactation, maternal bonding, and social affiliation. OT has been reported to exert an analgesic effect in both humans and animals, and the results of certain animal experiments have shown that the analgesic effect of OT is partially blocked by opioid receptor antagonists. To investigate the relationship between OT and μ opioid receptor (MOR), we evaluated how OT affects MOR in vitro by performing an electrical impedance-based receptor biosensor assay (CellKey™ assay), an intracellular cAMP assay, and a competitive receptor-binding analysis by using cells stably expressing human MOR and OT receptor. In both the CellKey™ assay and the intracellular cAMP assay, OT alone exerted no direct agonistic effect on human MOR, but treatment with 10−6 M OT markedly enhanced the MOR signaling induced by 10−6 M endomorphin-1, β-endorphin, morphine, fentanyl, and DAMGO. Moreover, in the competitive receptor-binding assay, 10−6 M OT did not alter the affinity of endomorphin-1 or morphine for MOR. These results suggest that OT could function as a positive allosteric modulator that regulates the efficacy of MOR signaling, and thus OT might represent a previously unrecognized candidate analgesic agent. Keywords: Oxytocin, μ opioid receptor, Positive allosteric modulator, Analgesia, G protein-coupled receptor (GPCR)
- Published
- 2018
- Full Text
- View/download PDF
34. GLP-1 receptor agonist liraglutide exerts central action to induce β-cell proliferation through medulla to vagal pathway in mice
- Author
-
Zesemdorj Otgon-Uul, Bo Yang Zhang, Masanori Nakata, Toshihiko Yada, and Parmila Kumari
- Subjects
Atropine ,Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Injections, Subcutaneous ,Hypothalamus ,Biophysics ,030209 endocrinology & metabolism ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Insulin-Secreting Cells ,Internal medicine ,Muscarinic acetylcholine receptor ,medicine ,Animals ,Receptor ,Molecular Biology ,Glucagon-like peptide 1 receptor ,Cell Proliferation ,Injections, Intraventricular ,Medulla Oblongata ,Chemistry ,Liraglutide ,digestive, oral, and skin physiology ,Area postrema ,Vagus Nerve ,Feeding Behavior ,Cell Biology ,Glucagon-like peptide-1 ,Vagus nerve ,Mice, Inbred C57BL ,Glucose ,030104 developmental biology ,Endocrinology ,Dorsal motor nucleus ,Proto-Oncogene Proteins c-fos ,hormones, hormone substitutes, and hormone antagonists ,Brain Stem ,medicine.drug - Abstract
Endogenous GLP-1 and GLP-1 receptor agonists (GLP-1RAs) regulate glucose metabolism via common and distinct mechanisms. Postprandial release of GLP-1 is modest and it is degraded by DPP-4 within 2 min, and hence it cannot enter the brain in substantial amount. In contrast, DPP-4-resistant GLP-1RAs are administered at 10 times higher concentration than endogenous GLP-1 level, which enables them to reach several brain regions including ARC and AP, the areas implicated in glucose metabolism. Hence, some of the effects of GLP-1RAs observed clinically and experimentally, including pancreatic β-cell proliferation, are thought to involve the brain. However, the effects of centrally acting GLP-1/GLP-1RAs on glucose metabolism and underlying neural mechanism are unclear. This study aimed to establish the link of central GLP-1/GLP-1RA action to pancreatic β-cell proliferation. Both subcutaneous (SC) and intracerebroventricular (ICV) injections of liraglutide increased the number of pancreatic β-cells expressing Ki67 and PCNA, proliferation markers, in C57BL/6J mice. This effect was induced by single ICV administration of liraglutide at relatively low dose that was incapable of suppressing food intake. These SC and ICV liraglutide-induced effects were inhibited by 50% and 70%, respectively, by pretreatment with atropine, a muscarinic receptor blocker. ICV liraglutide induced c-Fos expression in the area postrema (AP), nucleus tractus solitaries (NTS), and dorsal motor nucleus of the vagus (DMX) of the brain stem. These results demonstrate that central action of liraglutide induces pancreatic β-cell proliferation via the pathway involving the brain stem AP/NTS/DMX area and vagus nerve. This route is highly sensitive to GLP-1/GLP-1RA. Hence, this brain-pancreatic β-cell pathway may operate in type 2 diabetic patients treated with GLP-RAs and serve to counteract the reduction of β-cell mass.
- Published
- 2018
- Full Text
- View/download PDF
35. Weaning stage hyperglycemia induces glucose-insensitivity in arcuate POMC neurons and hyperphagia in type 2 diabetic GK rats
- Author
-
Shun Ishibashi, Fumihiko Maekawa, Masanori Nakata, Katsuya Dezaki, Akihiko Ando, Darambazar Gantulga, and Toshihiko Yada
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Pro-Opiomelanocortin ,Weaning ,Type 2 diabetes ,Hyperphagia ,Eating ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Endocrinology ,Arcuate nucleus ,Diabetes mellitus ,Internal medicine ,medicine ,Animals ,Premovement neuronal activity ,Rats, Wistar ,Neurons ,Arc (protein) ,Endocrine and Autonomic Systems ,business.industry ,Glucokinase ,Miglitol ,digestive, oral, and skin physiology ,Arcuate Nucleus of Hypothalamus ,nutritional and metabolic diseases ,General Medicine ,medicine.disease ,Glucose ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,Neurology ,Hyperglycemia ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
s Hyperphagia triggers and accelerates diabetes, and prevents proper dietary control of glycemia. Inversely, the impact of hyperglycemia on hyperphagia and possible mechanistic cause common for these two metabolic disorders in type 2 diabetes are less defined. The present study examined the precise developmental process of hyperglycemia and hyperphagia and explored the alterations in the hypothalamic arcuate nucleus (ARC), the primary feeding and metabolic center, in Goto-Kakizaki (GK) rats with type 2 diabetes and nearly normal body weight. At mid 3 to 4 weeks of age, GK rats first exhibited hyperglycemia, and then hyperphagia and reduced mRNA expressions for anorexigenic pro-opiomelanocortin (POMC) and glucokinase in ARC. Furthermore, [Ca2+]i responses to high glucose in ARC POMC neurons were impaired in GK rats at 4 weeks. Treating GK rats from early 3 to mid 6 weeks of age with an anti-diabetic medicine miglitol not only suppressed hyperglycemia but ameliorated hyperphagia and restored POMC mRNA expression in ARC. These results suggest that the early hyperglycemia occurring in weaning period may lead to impaired glucose sensing and neuronal activity of POMC neurons, and thereby induce hyperphagia in GK rats. Correction of hyperglycemia in the early period may prevent and/or ameliorate the progression of hyperphagia in type 2 diabetes.
- Published
- 2018
- Full Text
- View/download PDF
36. Inhibition of Y1 receptor signaling improves islet transplant outcome
- Author
-
Jeng Yie Chan, Thomas W.H. Kay, Katsuya Dezaki, Kim Loh, James Cantley, Esteban Nicolas Gurzov, Stacey N. Walters, Kailun Lee, Chi Kin Ip, Shane St Grey, Ross Laybutt, Toshihiko Yada, Herbert Herzog, Yan-Chuan Shi, Helen E. Thomas, Michaela Waibel, Masanori Nakata, and Mohammed Bensellam
- Subjects
0301 basic medicine ,medicine.medical_specialty ,endocrine system ,endocrine system diseases ,medicine.medical_treatment ,Science ,Islets of Langerhans Transplantation ,General Physics and Astronomy ,Biology ,Arginine ,General Biochemistry, Genetics and Molecular Biology ,Article ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Cyclic AMP ,Animals ,Humans ,Insulin ,Receptor ,lcsh:Science ,Islet cell transplantation ,geography ,Type 1 diabetes ,Multidisciplinary ,geography.geographical_feature_category ,Pancreatic islets ,General Chemistry ,Sciences bio-médicales et agricoles ,Islet ,medicine.disease ,3. Good health ,Receptors, Neuropeptide Y ,Transplantation ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cancer research ,lcsh:Q ,Signal transduction ,Signal Transduction - Abstract
Failure to secrete sufficient quantities of insulin is a pathological feature of type-1 and type-2 diabetes, and also reduces the success of islet cell transplantation. Here we demonstrate that Y1 receptor signaling inhibits insulin release in β-cells, and show that this can be pharmacologically exploited to boost insulin secretion. Transplanting islets with Y1 receptor deficiency accelerates the normalization of hyperglycemia in chemically induced diabetic recipient mice, which can also be achieved by short-term pharmacological blockade of Y1 receptors in transplanted mouse and human islets. Furthermore, treatment of non-obese diabetic mice with a Y1 receptor antagonist delays the onset of diabetes. Mechanistically, Y1 receptor signaling inhibits the production of cAMP in islets, which via CREB mediated pathways results in the down-regulation of several key enzymes in glycolysis and ATP production. Thus, manipulating Y1 receptor signaling in β-cells offers a unique therapeutic opportunity for correcting insulin deficiency as it occurs in the pathological state of type-1 diabetes as well as during islet transplantation.Islet transplantation is considered one of the potential treatments for T1DM but limited islet survival and their impaired function pose limitations to this approach. Here Loh et al. show that the Y1 receptor is expressed in β- cells and inhibition of its signalling, both genetic and pharmacological, improves mouse and human islet function., info:eu-repo/semantics/published
- Published
- 2017
37. Complexity of Stomach–Brain Interaction Induced by Molecular Hydrogen in Parkinson’s Disease Model Mice
- Author
-
Mami Noda, Yusaku Nakabeppu, Toshihiko Yada, Yusaku Iwasaki, Yuya Uemura, Yusuke Yoshii, and Taikai Inoue
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,medicine.drug_class ,medicine.medical_treatment ,Substantia nigra ,Stimulation ,Vagotomy ,Biochemistry ,Neuroprotection ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Parkinsonian Disorders ,Internal medicine ,medicine ,Animals ,Mice, Knockout ,Chemistry ,Stomach ,digestive, oral, and skin physiology ,Brain ,Vagus Nerve ,General Medicine ,Deuterium ,Receptor antagonist ,medicine.disease ,Ghrelin ,Vagus nerve ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Gastric Mucosa ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
Molecular hydrogen (H2), as a new medical gas, has protective effects in neurological disorders including Parkinson's disease (PD). In our previous report, the neuroprotective effect of drinking water with saturated H2 (H2 water) in PD mice might be due to stomach-brain interaction via release of gastric hormone, ghrelin. In the present study, we assessed the effect of H2-induced ghrelin more precisely. To confirm the contribution of ghrelin in H2 water-drinking PD model mice, ghrelin-knock out (KO) mice were used. Despite the speculation, the effect of H2 water was still observed in ghrelin-KO PD model mice. To further check the involvement of ghrelin, possible contribution of ghrelin-induced vagal afferent effect was tested by performing subdiaphragmatic vagotomy before treating with H2 water and administration of MPTP (1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine). The protective effect of H2 water was still observed in the vagotomized mice in substantia nigra, suggesting that stimulation of vagal afferent nerves is not involved in H2-induced neuroprotection. Other neuroprotective substitutes in ghrelin-KO mice were speculated because H2-induced neuroprotection was not cancelled by ghrelin receptor antagonist, D-Lys3 GHRP-6, in ghrelin-KO PD model mice, unlike in wild-type PD model mice. Our results indicate that ghrelin may not be the only factor for H2-induced neuroprotection and other factors can substitute the role of ghrelin when ghrelin is absent, raising intriguing options of research for H2-responsive factors.
- Published
- 2017
- Full Text
- View/download PDF
38. The liver-brain-gut neural arc maintains the T
- Author
-
Toshiaki, Teratani, Yohei, Mikami, Nobuhiro, Nakamoto, Takahiro, Suzuki, Yosuke, Harada, Koji, Okabayashi, Yuya, Hagihara, Nobuhito, Taniki, Keita, Kohno, Shinsuke, Shibata, Kentaro, Miyamoto, Harumichi, Ishigame, Po-Sung, Chu, Tomohisa, Sujino, Wataru, Suda, Masahira, Hattori, Minoru, Matsui, Takaharu, Okada, Hideyuki, Okano, Masayuki, Inoue, Toshihiko, Yada, Yuko, Kitagawa, Akihiko, Yoshimura, Mamoru, Tanida, Makoto, Tsuda, Yusaku, Iwasaki, and Takanori, Kanai
- Subjects
Male ,Neurons ,Afferent Pathways ,digestive, oral, and skin physiology ,Antigen-Presenting Cells ,Brain ,Vagus Nerve ,Colitis ,Receptors, Muscarinic ,T-Lymphocytes, Regulatory ,Article ,Rats ,Intestines ,Mice ,Liver ,Animals ,Homeostasis ,Humans ,Spleen - Abstract
Vagus nerve stimulation has shown promise in treating inflammatory bowel disease. A new study identifies a hepatic vagal branch pathway necessary to maintain colonic immune cell homeostasis in experimental colitis, challenging the dogma that an anti-inflammatory reflex requires an intact spleen and that α7-nicotinic receptor agonists are a panacea for inflammatory conditions.
- Published
- 2019
39. Relay of peripheral oxytocin to central oxytocin neurons via vagal afferents for regulating feeding
- Author
-
Toshihiko Yada, Lei Wang, Yusaku Iwasaki, Parmila Kumari, Shizu Hidema, and Katsuhiko Nishimori
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,medicine.drug_class ,medicine.medical_treatment ,Biophysics ,Vagotomy ,Body weight ,Oxytocin ,Biochemistry ,Supraoptic nucleus ,Uterine contraction ,03 medical and health sciences ,Eating ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Melanocyte-Stimulating Hormones ,Molecular Biology ,Mice, Knockout ,Neurons ,business.industry ,digestive, oral, and skin physiology ,Cell Biology ,Receptor antagonist ,Peripheral ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Receptors, Oxytocin ,030220 oncology & carcinogenesis ,medicine.symptom ,business ,Nucleus ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Oxytocin (Oxt), a neurohormone synthesized in the neurons of hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus induces milk-ejection and uterine contraction and regulates social behavior, stress responses, memory and food intake. Peripheral (intraperitoneal and subcutaneous) infusion of Oxt decreases food intake and body weight in obese animals via mechanisms involving vagal afferent nerves and in obese subjects when administered nasally. Peripherally injected and intracerebroventricularly injected Oxt inhibit food intake to similar extent and with similar time course. Thus, peripheral Oxt mimics the effects of central Oxt, however, underlying mechanisms are unclear. In the present study we explored whether intraperitoneal Oxt activates Oxt neurons in PVN via vagal afferents and whether this pathway is linked to inhibition of feeding. We here show that intraperitoneal Oxt injection induces c-Fos expression in PVN largely in Oxt neurons and inhibits food intake, and these effects are blunted by subdiaphragmatic vagotomy. The intraperitoneal Oxt-induced inhibition of food intake was blunted in Oxt KO mice, by intracerebroventricular injection of Oxt receptor antagonist, and by vagotomy. These results demonstrate that intraperitoneal Oxt injection activates PVN Oxt neurons via vagal afferent nerves, thereby inhibiting food intake. This vagal afferents-mediated Oxt's peripheral-to-central coupling may serve to promote satiety and possibly a series of neural functions of Oxt and to treat their disorders.
- Published
- 2019
40. Immunohistochemical localization and possible functions of nesfatin-1 in the testis of mice during pubertal development and sexual maturation
- Author
-
Mayank Choubey, Ashutosh Ranjan, Amitabh Krishna, and Toshihiko Yada
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Histology ,GLUT8 ,Physiology ,medicine.medical_treatment ,03 medical and health sciences ,Mice ,Organ Culture Techniques ,Western blot ,In vivo ,Internal medicine ,Testis ,medicine ,Sexual maturity ,Animals ,Nucleobindins ,Testosterone ,Sexual Maturation ,Gonadal Steroid Hormones ,Spermatogenesis ,030102 biochemistry & molecular biology ,biology ,medicine.diagnostic_test ,Leydig Cells ,Cell Biology ,General Medicine ,Phosphoproteins ,Immunohistochemistry ,Receptor, Insulin ,Insulin receptor ,Steroid hormone ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,biology.protein ,Lactates ,Immunostaining - Abstract
The study was aimed to address the role of nesfatin-1 on the sexual maturation of testis during the pubertal transition. The immunostaining of testis suggested nesfatin-1 is expressed in Leydig cells with pubertal maturation. The pre-pubertal mice for in vivo study were randomly divided in three groups; (a) control-saline (b) treated with low (0.25 nM) dose of nesfatin-1/gbw/day and (c) treated with high (1.25 nM) dose nesfatin-1/gbw/day. Histological analysis showed that nesfatin-1 loaded mice showed facilitated maturation of testis. Western blot analysis on various protein expressions upon injection of nesfatin-1 into pre-pubertal mice suggested that expressions of proteins involving steroid hormone production, spermatogenic markers (PCNA, Bcl2, AR), glucose uptake-related proteins (GLUT8 and insulin receptor) and GnRH-R and GPR-54 proteins were facilitated. Both of lactose dehydrogenase activity and lactate levels were increased. The treatment with nesfatin-1 also reduced oxidative stress, which further facilitates testicular functions during puberty. The treatment of nesfatin-1 on cultured testis also supports in vivo findings as evident by the increased testosterone production and StAR protein expression as well as increased glucose and lactate production. In sum, our data report for the first time the accelerative role of nesfatin-1 on spermatogenesis and steroidogenesis of pre-pubertal male mice by directly acting on the testis coupled with the advancement of puberty.
- Published
- 2019
41. Nesfatin-1 ameliorates type-2 diabetes-associated reproductive dysfunction in male mice
- Author
-
Mayank Choubey, Ashutosh Ranjan, Toshihiko Yada, and Amitabh Krishna
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Type 2 diabetes ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,Diabetes mellitus ,Testis ,medicine ,Animals ,Insulin ,Nucleobindins ,Prohibitin ,Spermatogenesis ,Testosterone ,biology ,business.industry ,Reproduction ,nutritional and metabolic diseases ,medicine.disease ,Streptozotocin ,Receptor, Insulin ,Up-Regulation ,Insulin receptor ,Diabetes Mellitus, Type 2 ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,biology.protein ,Insulin Resistance ,business ,medicine.drug - Abstract
The present study was aimed to demonstrate the recuperative effect of nesfatin-1 on testicular dysfunction in the high-fat diet (HFD)/streptozotocin (STZ)-induced type-2 diabetes mellitus (T2DM) mice. Three experimental groups were formed: (1) vehicle control (VC), (2) T2DM mice, (3) T2DM + nesf-1. The mice with blood glucose level higher than 300 mg/dL following HFD and a single dose of STZ were used for the experiment. The T2DM mice showed increases in body mass, blood glucose and insulin levels, reductions in spermatogenesis and steroidogenesis, production of antioxidative enzymes, and disturbed lipid profile. These alterations were all ameliorated by administration of nesfatin-1 at 20 μg/Kg BW for 15 days. Nesfatin-1 treatment also increased the production of testosterone (T), improved insulin sensitivity, and effectively ameliorated the testicular aberrations, and increased spermatogenesis and steroidogenesis. In addition, nesfatin-1 treatment upregulated the PCNA and Bcl2 expression and inhibited the caspase-3 and prohibitin expression in T2DM mice. Nesfatin-1 increased insulin receptor (IR) and GLUT8 expressions, and lactate production, the changes that further substantiate the increase of energy influx to the testis. Altogether, the results suggest the ameliorative effect of nesfatin-1 against T2DM-associated testicular dysfunctions and improved insulin sensitivity along with promoting T production and fertility in T2DM mice.
- Published
- 2019
42. Islet β-cell-produced NUCB2/nesfatin-1 maintains insulin secretion and glycemia along with suppressing UCP-2 in β-cells
- Author
-
Boyang Zhang, Jun Nakae, Yifei Yang, Masanori Nakata, Toshihiko Yada, and Masatomo Mori
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,endocrine system ,medicine.medical_specialty ,Physiology ,medicine.medical_treatment ,Cell ,Carbohydrate metabolism ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Animals ,Insulin ,Nucleobindins ,Uncoupling Protein 2 ,Mice, Knockout ,Glucose tolerance test ,Gene knockdown ,geography ,geography.geographical_feature_category ,medicine.diagnostic_test ,Chemistry ,Pancreatic islets ,Glucose Tolerance Test ,Islet ,Nucleobindin 2 ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Glucose ,Glycemic Index ,030217 neurology & neurosurgery - Abstract
Nesfatin-1 is a hypothalamic anorexigenic peptide processed from nucleobindin 2 (NUCB2). Central and peripheral administration of NUCB2/nesfatin-1 enhances glucose metabolism and insulin release. NUCB2/nesfatin-1 is also localized in pancreatic islets, while its function remains unknown. To explore the role of pancreatic β-cell-produced NUCB2/nesfatin-1, we developed pancreatic β-cell-specific NUCB2 knockout (βNUCB2 KO) mice and NUCB2 gene knockdown (shNUCB2) MIN6 β-cell line. In βNUCB2 KO mice, casual blood glucose was elevated from 12 weeks of age. In a glucose tolerance test at 12 weeks, insulin secretion at 15 min was reduced and blood glucose at 2 h increased in βNUCB2 KO mice fasted 8 h. In islets isolated from βNUCB2 KO mice, high glucose-stimulated insulin secretion (GSIS) was impaired. In shNUCB2 MIN6 cells, GSIS was reduced and UCP-2 mRNA expression was elevated. These results show impaired GSIS possibly associated with UCP-2 overexpression in NUCB2-silenced β-cells, suggesting that β-cell-produced NUCB2/nesfatin-1 maintains GSIS and thereby glycemia.
- Published
- 2019
43. Helicobacter pylori Vacuolating Cytotoxin A Causes Anorexia and Anxiety via Hypothalamic Urocortin 1 in Mice
- Author
-
Haruki Iwai, Toshihiko Yada, Akio Inui, Akihiro Asakawa, Takakazu Yagi, Yoshito Yokoyama, Masayasu Kojima, Kinnosuke Yahiro, Toshiya Hirayama, Koji Ataka, Kai-Chun Cheng, Norifumi Nakamura, Takeshi Arai, Hajime Suzuki, Katsuhiro Yamamoto, and Miharu Ushikai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Hypothalamus ,lcsh:Medicine ,Anorexia ,Anxiety ,Article ,Corticotropin-releasing hormone receptor 1 ,Pathogenesis ,03 medical and health sciences ,Eating ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,RNA, Messenger ,lcsh:Science ,Receptor ,Protein kinase C ,Urocortins ,Multidisciplinary ,Phospholipase C ,Helicobacter pylori ,Chemistry ,Cytotoxins ,lcsh:R ,digestive, oral, and skin physiology ,Body Weight ,bacterial infections and mycoses ,digestive system diseases ,030104 developmental biology ,Endocrinology ,Anxiogenic ,nervous system ,Vacuoles ,bacteria ,lcsh:Q ,medicine.symptom ,Gerbillinae ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Helicobacter pylori (Hp) infection is related to the pathogenesis of chronic gastric disorders and extragastric diseases. Here, we examined the anorexigenic and anxiogenic effects of Hp vacuolating cytotoxin A (VacA) through activation of hypothalamic urocortin1 (Ucn1). VacA was detected in the hypothalamus after peripheral administration and increased Ucn1 mRNA expression and c-Fos-positive cells in the hypothalamus but not in the nucleus tractus solitarius. c-Fos and Ucn1-double positive cells were detected. CRF1 and CRF2 receptor antagonists suppressed VacA-induced anxiety and anorexia, respectively. VacA activated single paraventricular nucleus neurons and A7r5 cells; this activation was inhibited by phospholipase C (PLC) and protein kinase C (PKC) inhibitors. VacA causes anorexia and anxiety through the intracellular PLC-PKC pathway, migrates across the blood-brain barrier, and activates the Ucn1-CRF receptor axis.
- Published
- 2019
44. YIL counteracts ghrelin-inhibited insulin release in pancreatic islets of langerhans
- Author
-
Toshihiko Yada, Rauza Sukma Rita, and Katsuya Dezaki
- Subjects
medicine.medical_specialty ,Endocrinology ,medicine.anatomical_structure ,business.industry ,Pancreatic islets ,Internal medicine ,Insulin ,medicine.medical_treatment ,Medicine ,Ghrelin ,business - Abstract
Ghrelin is a peptide hormone that is produced mainly from the stomach. Ghrelin is reported to have many biological functions, such as modulating feeding behavior, energy balance, and glucose homeostasis. This study aimed to examine whether YIL, a ghrelin receptor antagonist, could counteract the effect of ghrelin-inhibited insulin release in the pancreatic islet of Langerhans. This study is experimental research using wild-type C57BL/6J mice [8-10 weeks old]. Islet of Langerhans was isolated by collagenase digestion and the insulin release [ng/islet/h] from the islet is examined by the ELISA method. Data represent means ± SEM and is analyzed by one-way ANOVA. The result showed that 8.3 mM glucose concentration increase insulin release compared to 2.8 mM glucose, respectively [0,393 ± 0,025 vs 0,219 ± 0,022 ng/islet/h]. In the presence of 8.3 mM glucose, ghrelin 1 nM showed a decrease in insulin release significantly compared to 8.3 mM glucose only [0,283 ± 0,001 vs 0,393 ± 0,025, p < 0,01]. In contrast, in the presence of 8.3 mM glucose and ghrelin 1 nM, YIL 1 μM induced insulin secretion [0,386 ± 0,012 vs 0,283 ± 0,001, p < 0,01]. In conclusion, YIL is significantly counteracted ghrelin-inhibited insulin release in pancreatic islets of Langerhans. Furthermore, YIL is one of the candidates for the treatment of type 2 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
45. Endogenous α2A-Adrenoceptor–Operated Sympathoadrenergic Tones Attenuate Insulin Secretion via cAMP/TRPM2 Signaling
- Author
-
Masashi Yoshida, Toshihiko Yada, Kazuo Hara, Masafumi Kakei, Kaoru Tabei, Yoshiyuki Morishita, Rina Miura, Katsuya Dezaki, Kiyonori Ito, Masanobu Kawakami, Rauza Sukma Rita, Hodaka Yamada, and Susumu Ookawara
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Patch-Clamp Techniques ,Sympathetic Nervous System ,Epinephrine ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,TRPM Cation Channels ,Adrenergic ,Incretins ,Islets of Langerhans ,Mice ,03 medical and health sciences ,Transient receptor potential channel ,Receptors, Adrenergic, alpha-2 ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,Cyclic AMP ,Internal Medicine ,medicine ,Animals ,Insulin ,TRPM2 ,Patch clamp ,Sympathomimetics ,Mice, Knockout ,Glucose tolerance test ,medicine.diagnostic_test ,Chemistry ,Glucose Tolerance Test ,Yohimbine ,Glucose ,030104 developmental biology ,Endocrinology ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
In pancreatic β-cells, pharmacological concentrations of catecholamines, including adrenaline, have been used to inhibit insulin release and explore the multiple mechanisms involved. However, the significance of these signaling pathways for physiological adrenergic functions in β-cells is largely unknown. In the process of glucose-induced insulin secretion, opening of background current through nonselective cation channels (NSCCs) might facilitate membrane depolarization by closure of the ATP-sensitive K+ channels. Here, we examined whether physiological insulinostatic adrenaline action is mediated via the transient receptor potential melastatin 2 (TRPM2) channel, a type of NSCC, in β-cells. Results showed that physiological concentrations of adrenaline strongly suppressed glucose-induced and incretin-potentiated cAMP production and insulin secretion and inhibited NSCCs current and membrane excitability via the α2A-adrenoceptor in wild-type mice; however, insulin secretion was not attenuated in TRPM2-knockout (KO) mice. Administration of yohimbine, an α2-adrenoceptor antagonist, failed to affect glucose tolerance in TRPM2-KO mice, in contrast to an improved glucose tolerance in wild-type mice receiving the antagonist. The current study demonstrated that a physiological concentration of adrenaline attenuates insulin release via coupling of α2A-adrenoceptor to cAMP/TRPM2 signaling, thereby providing a potential therapeutic tool to treat patients with type 2 diabetes.
- Published
- 2016
- Full Text
- View/download PDF
46. Caspase-1 deficiency promotes high-fat diet-induced adipose tissue inflammation and the development of obesity
- Author
-
Hiroko Tsutsui, Toshihiko Yada, Yuka Endo, Yusaku Iwasaki, Ai Sadatomo, Jun Nakamura, Akira Kawashima, Masafumi Takahashi, Motoi Kobayashi, Tadayoshi Karasawa, Tadashi Kasahara, Fumitake Usui, and Hiroaki Kimura
- Subjects
Blood Glucose ,Leptin ,Male ,0301 basic medicine ,Physiology ,Endocrinology, Diabetes and Metabolism ,Interleukin-1beta ,Adipose tissue ,Mice ,0302 clinical medicine ,Adipocytes ,Insulin ,Chemokine CCL2 ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Caspase 1 ,Interleukin-18 ,Inflammasome ,Stromal vascular fraction ,Flow Cytometry ,Interleukin-12 ,Cholesterol ,Adipose Tissue ,030220 oncology & carcinogenesis ,Body Composition ,Adiponectin ,medicine.symptom ,medicine.drug ,medicine.medical_specialty ,Adipose tissue macrophages ,Inflammation ,CCL2 ,Biology ,Diet, High-Fat ,Real-Time Polymerase Chain Reaction ,Proinflammatory cytokine ,Interferon-gamma ,03 medical and health sciences ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Obesity ,Triglycerides ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Macrophages ,X-Ray Microtomography ,Glucose Tolerance Test ,030104 developmental biology ,Endocrinology - Abstract
Caspase-1 is a cysteine protease responsible for the processing of the proinflammatory cytokine interleukin-1β and activated by the formation of inflammasome complexes. Although several investigations have found a link between diet-induced obesity and caspase-1, the relationship remains controversial. Here, we found that mice deficient in caspase-1 were susceptible to high-fat diet-induced obesity with increased adiposity as well as normal lipid and glucose metabolism. Caspase-1 deficiency clearly promoted the infiltration of inflammatory macrophages and increased the production of C-C motif chemokine ligand 2 (CCL2) in the adipose tissue. The dominant cellular source of CCL2 was stromal vascular fraction rather than adipocytes in the adipose tissue. These findings demonstrate a critical role of caspase-1 in macrophage-driven inflammation in the adipose tissue and the development of obesity. These data provide novel insights into the mechanisms underlying inflammation in the pathophysiology of obesity.
- Published
- 2016
- Full Text
- View/download PDF
47. Paraventricular NUCB2/Nesfatin-1 Supports Oxytocin and Vasopressin Neurons to Control Feeding Behavior and Fluid Balance in Male Mice
- Author
-
Putra Santoso, Darambazar Gantulga, Takashi Okada, Boyang Zhang, Chiaki Masuda, Masatomo Mori, Masanori Nakata, and Toshihiko Yada
- Subjects
Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Vasopressin ,Arginine ,Vasopressins ,Blotting, Western ,Nerve Tissue Proteins ,Endogeny ,Calorimetry ,Biology ,Oxytocin ,Energy homeostasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Insulin resistance ,Internal medicine ,medicine ,Animals ,Nucleobindins ,RNA, Small Interfering ,Adiposity ,Mice, Knockout ,Neurons ,Gene knockdown ,Calcium-Binding Proteins ,digestive, oral, and skin physiology ,Feeding Behavior ,medicine.disease ,Immunohistochemistry ,DNA-Binding Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,Hypothalamus ,Insulin Resistance ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,Paraventricular Hypothalamic Nucleus ,medicine.drug - Abstract
Nesfatin-1, derived from nucleobindin-2 (NUCB2), is expressed in the hypothalamus, including the paraventricular nucleus (PVN), an integrative center for energy homeostasis. However, precise role of the NUCB2/nesfatin-1 in PVN remains less defined. The present study aimed to clarify physiological and/or pathophysiological roles of endogenous NUCB2/nesfatin-1 in PVN by using adeno-associated virus vectors encoding short hairpin RNAs targeting NUCB2 in mice. PVN-specific NUCB2 knockdown primarily increased food intake and decreased plasma oxytocin level specifically in light phase, leading to increased body weight gain without affecting energy expenditure. Furthermore, high-salt diet increased the systolic blood pressure, plasma arginine vasopressin (AVP) and AVP mRNA expression in PVN, and all these changes were blunted by PVN-specific NUCB2 knockdown. These results reveal that the endogenous NUCB2/nesfatin-1 in PVN regulates PVN AVP and oxytocin and consequently the fluid and energy balance.
- Published
- 2016
- Full Text
- View/download PDF
48. Neural effects of gut‐ and brain‐derived glucagon‐like peptide‐1 and its receptor agonist
- Author
-
Kenichi Katsurada and Toshihiko Yada
- Subjects
0301 basic medicine ,Agonist ,medicine.medical_specialty ,endocrine system ,medicine.drug_class ,Proceedings of INCRETIN 2015, A Symposium Celebrating the 45th Anniversary of the Discovery of GIP, 29–31 July 2015, Vancouver, Canada. This publication has been supported by: The Local Organizing Committee of INCRETIN 2015 ,Endocrinology, Diabetes and Metabolism ,Mini Review ,Enteroendocrine Cells ,Central nervous system ,Incretin ,Enteroendocrine cell ,Biology ,Cardiovascular ,Brain stem ,Glucagon-Like Peptide-1 Receptor ,Cardiovascular Physiological Phenomena ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Glucagon-Like Peptide 1 ,Internal medicine ,Internal Medicine ,medicine ,Solitary Nucleus ,Animals ,Humans ,Neurotransmitter ,Receptor ,Review Articles ,Solitary nucleus ,Feeding ,digestive, oral, and skin physiology ,General Medicine ,Feeding Behavior ,Glucagon-like peptide-1 ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,hormones, hormone substitutes, and hormone antagonists - Abstract
Glucagon‐like peptide‐1 (GLP‐1) is derived from both the enteroendocrine L cells and preproglucagon‐expressing neurons in the nucleus tractus solitarius (NTS) of the brain stem. As GLP‐1 is cleaved by dipeptidyl peptidase‐4 yielding a half‐life of less than 2 min, it is plausible that the gut‐derived GLP‐1, released postprandially, exerts its effects on the brain mainly by interacting with vagal afferent neurons located at the intestinal or hepatic portal area. GLP‐1 neurons in the NTS widely project in the central nervous system and act as a neurotransmitter. One of the physiological roles of brain‐derived GLP‐1 is restriction of feeding. GLP‐1 receptor agonists have recently been used to treat type 2 diabetic patients, and have been shown to exhibit pleiotropic effects beyond incretin action, which involve brain functions. GLP‐1 receptor agonist administered in the periphery is stable because of its resistance to dipeptidyl peptidase‐4, and is highly likely to act on the brain by passing through the blood–brain barrier (BBB), as well as interacting with vagal afferent nerves. Central actions of GLP‐1 have various roles including regulation of feeding, weight, glucose and lipid metabolism, cardiovascular functions, cognitive functions, and stress and emotional responses. In the present review, we focus on the source of GLP‐1 and the pathway by which peripheral GLP‐1 informs the brain, and then discuss recent findings on the central effects of GLP‐1 and GLP‐1 receptor agonists.
- Published
- 2016
49. Betatrophin expression is promoted in obese hyperinsulinemic type 2 but not type 1 diabetic mice
- Author
-
Toshihiko Yada, Masanori Nakata, Yifei Yang, Boyang Zhang, EnXu Li, and Atsumi Shinozaki
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Betatrophin ,Adipose Tissue, White ,Peptide Hormones ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Mice, Obese ,Adipose tissue ,Type 2 diabetes ,White adipose tissue ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,Endocrinology ,Adipose Tissue, Brown ,Angiopoietin-Like Protein 8 ,Hyperinsulinism ,Internal medicine ,Brown adipose tissue ,medicine ,Animals ,Obesity ,Type 1 diabetes ,business.industry ,Insulin ,nutritional and metabolic diseases ,Streptozotocin ,medicine.disease ,Mice, Inbred C57BL ,Angiopoietin-like Proteins ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,Liver ,business ,medicine.drug - Abstract
Regeneration of pancreatic β-cell mass benefits both type 1 and type 2 diabetic patients. A recent study identified betatrophin as a β-cell proliferation factor. However, the expressional regulation of betatrophin remains less defined. In this study, we aimed to clarify the regulation of betatrophin expression in obese type 2 vs. type 1 diabetes model animals. We experimented type 2 diabetes models, diet-induced-obesity (DIO) mice and db/db mice, and type 1 diabetes models, C57B6 mice receiving streptozotocin (STZ) or 70% pancreatectomy to destroy or remove β-cells. Serum betatrophin levels and betatrophin mRNA expressions in the liver, white adipose tissue (WAT) and brown adipose tissue (BAT) were measured. In DIO mice and db/db mice, serum betatrophin and betatrophin mRNA expressions in the liver, WAT and BAT were elevated in parallel with increases in body weight and plasma insulin. These elevated betatrophin mRNA expressions were not altered by treatment with SGLT2 inhibitor that ameliorated hyperglycemia. In pancreatectomized mice, betatrophin expression in WAT decreased in parallel with reductions in weight and insulin. In STZ-treated mice, betatrophin expressions in the liver, WAT and BAT were reduced. However, when the mouse liver slices were cultured with STZ, betatrophin expression was significantly reduced, indicating a direct action of STZ on the liver. These results indicate that the expression of betatrophin is upregulated in the liver, WAT and BAT in obese hyperinsulinemic type 2 diabetes but decreased in WAT in hypoinsulinemic type 1 diabetes, suggesting its positive correlation with body weight and plasma insulin but not blood glucose.
- Published
- 2016
- Full Text
- View/download PDF
50. Direct effects of neuropeptide nesfatin-1 on testicular spermatogenesis and steroidogenesis of the adult mice
- Author
-
Mayank Choubey, Amitabh Krishna, Ashutosh Ranjan, and Toshihiko Yada
- Subjects
Male ,medicine.medical_specialty ,Aging ,GLUT8 ,Neuropeptide ,030209 endocrinology & metabolism ,Nerve Tissue Proteins ,medicine.disease_cause ,Antioxidants ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,Paracrine signalling ,Mice ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Proliferating Cell Nuclear Antigen ,Testis ,medicine ,Animals ,Nucleobindins ,Testosterone ,RNA, Messenger ,Spermatogenesis ,030304 developmental biology ,0303 health sciences ,biology ,Estradiol ,Calcium-Binding Proteins ,Neuropeptides ,Spermatozoa ,DNA-Binding Proteins ,Insulin receptor ,Oxidative Stress ,chemistry ,biology.protein ,Animal Science and Zoology ,Steroids ,Oxidative stress ,Biomarkers ,Signal Transduction - Abstract
Recent studies have revealed nesfatin-1 as a hypothalamic neuropeptide, regulating food intake, energy expenditure and reproduction primarily by acting on the hypothalamic-pituitary-gonadal axis. Nesfatin-1 is also localized in several peripheral tissues including testes. However, functional significance of nesfatin-1 in testicular activities is not yet well documented in mammals. Therefore, this study was aimed to elucidate the direct effects of nesfatin-1 on testicular markers for steroid productions, spermatogenesis, metabolic changes and oxidative stress. The results revealed the expression of both protein and mRNA of nesfatin-1 in the testes of adult mice. The testes treated in vitro with nesfatin-1 showed significant increase in testosterone production, which correlated significantly with increased expression of steroidogenic markers and insulin receptor proteins in the testes. Furthermore, the in vitro treatment with nesfatin-1 showed stimulatory effects on spermatogenesis by promoting cell proliferation (PCNA) and survival (Bcl2), while inhibiting apoptosis (caspase-3) in the testes. The nesfatin-1 treatment in vitro further increased the expression of insulin receptor and GLUT8 proteins, in parallel with increase in the intra-testicular transport of glucose and production of lactate. This nesfatin-1 induced enhanced transport of energy substrate (glucose and lactate) may be responsible for promoting spermatogenesis and steroidogenesis. Nesfatin-1 significantly reduced oxidative stress and nitric oxide, which may also be responsible for stimulatory effects on testicular activities. The present finding suggests that nesfatin-1 acts via paracrine manner to increase sperm count and fertility, thus promoting the testicular function.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.