Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cytotherapy ; 25(7): 683-698, 2023 07.
Article in English | MEDLINE | ID: mdl-37097265

ABSTRACT

The 5th Asia Partnership Conference of Regenerative Medicine (APACRM) was held online on April 7, 2022 to promote regulatory harmonization of regenerative medicine products throughout Asia. The recognition of domestic regulatory guidelines within each country and region and the underpinning rationales are important initial steps toward the harmonization of regulations. The 5th APACRM featured open dialog regarding non-clinical, quality and environmental impact assessment settings for cell and gene therapy products through presentations from the industry and panel discussions with regulatory agencies. The latest updates on regenerative medicine fields in each country and region were also introduced. This paper summarizes the proceedings of the 5th APACRM for public dissemination to foster future discussion.


Subject(s)
Environment , Regenerative Medicine , Asia , Genetic Therapy/adverse effects
2.
Cytotherapy ; 24(9): 892-904, 2022 09.
Article in English | MEDLINE | ID: mdl-35193824

ABSTRACT

The 4th Asia Partnership Conference of Regenerative Medicine (APACRM) was held online on April 15, 2021, to promote regulatory harmonization of regenerative medicine products throughout Asia. Recognizing domestic regulatory guidelines within each country and region, and their underpinning rationales, is an important initial step toward a convergence of regulations. The 4th APACRM consisted of an open dialog with regulatory agencies regarding nonclinical and quality settings for cell therapy products (CTPs) through industry presentations and panel discussions with regulatory agencies. The latest updates on regenerative medicine fields in each country and region, and specific regulatory schematics in Japan, were also introduced. The objective of this paper is to summarize the proceedings of the 4th APACRM for public dissemination and to foster further discussion in the future.


Subject(s)
Cell- and Tissue-Based Therapy , Regenerative Medicine , Asia , Japan
3.
Commun Biol ; 4(1): 994, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417555

ABSTRACT

Reduced glucose uptake into the skeletal muscle is an important pathophysiological abnormality in type 2 diabetes, and is caused by impaired translocation of glucose transporter 4 (GLUT4) to the skeletal muscle cell surface. Here, we show a xanthene derivative, DS20060511, induces GLUT4 translocation to the skeletal muscle cell surface, thereby stimulating glucose uptake into the tissue. DS20060511 induced GLUT4 translocation and stimulated glucose uptake into differentiated L6-myotubes and into the skeletal muscles in mice. These effects were completely abolished in GLUT4 knockout mice. Induction of GLUT4 translocation by DS20060511 was independent of the insulin signaling pathways including IRS1-Akt-AS160 phosphorylation and IRS1-Rac1-actin polymerization, eNOS pathway, and AMPK pathway. Acute and chronic DS20060511 treatment attenuated the glucose intolerance in obese diabetic mice. Taken together, DS20060511 acts as a skeletal muscle-specific GLUT4 translocation enhancer to facilitate glucose uptake. Further studies of DS20060511 may pave the way for the development of novel antidiabetic medicines.


Subject(s)
Glucose Intolerance/genetics , Glucose Transporter Type 4/genetics , Muscle, Skeletal/metabolism , Translocation, Genetic , Xanthenes/metabolism , Animals , Glucose Intolerance/metabolism , Glucose Transporter Type 4/metabolism , Mice
4.
Cytotherapy ; 23(10): 874-885, 2021 10.
Article in English | MEDLINE | ID: mdl-34116946

ABSTRACT

BACKGROUND AIMS: Cell-based regenerative medicine is an innovative field that can potentially alter the overall survival and quality of life of patients with devastating diseases. Several cell therapy products (CTPs) have been approved within the last two decades, and more are under development. The establishment of an effective developmental strategy in accordance with the regulatory bodies of each country/region is crucial for fast delivery of each respective CTP. In particular, facilitating investigational new drug (IND) approval is important for accelerating the transition from non-clinical to clinical research/trial phases. METHODS: Here the authors compared the non-clinical prerequisites for initiating clinical studies in five Asian countries/regions (India, China, Korea, Taiwan and Japan) from an industry viewpoint. The authors first identified the differences and tried to clarify the perspectives/considerations underpinning the different requirements. RESULTS: The authors' findings revealed that differences in regulations and development experiences, especially with CTPs, have led to clear differences in the non-clinical study package and its corresponding study design. CONCLUSIONS: By sharing experiences of the research and development of CTPs among Asian countries/regions and including not only industry but also regulatory authorities, we will be able to expedite cross-border IND approval and eventually contribute to the early delivery of innovative CTPs to many Asian patients.


Subject(s)
Cell- and Tissue-Based Therapy , Quality of Life , Asia , China , Humans , Japan
5.
EMBO Rep ; 21(9): e49828, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32672883

ABSTRACT

While brown adipose tissue (BAT) is well-recognized for its ability to dissipate energy in the form of heat, recent studies suggest multifaced roles of BAT in the regulation of glucose and lipid homeostasis beyond stimulating thermogenesis. One of the functions involves interorgan communication with metabolic organs, such as the liver, through BAT-derived secretory factors, a.k.a., batokine. However, the identity and the roles of such mediators remain insufficiently understood. Here, we employed proteomics and transcriptomics in human thermogenic adipocytes and identified previously unappreciated batokines, including phospholipid transfer protein (PLTP). We found that increased circulating levels of PLTP, via systemic or BAT-specific overexpression, significantly improve glucose tolerance and insulin sensitivity, increased energy expenditure, and decrease the circulating levels of cholesterol, phospholipids, and sphingolipids. Such changes were accompanied by increased bile acids in the circulation, which in turn enhances glucose uptake and thermogenesis in BAT. Our data suggest that PLTP is a batokine that contributes to the regulation of systemic glucose and lipid homeostasis as a mediator of BAT-liver interorgan communication.


Subject(s)
Adipose Tissue, Brown , Glucose , Adipose Tissue, Brown/metabolism , Energy Metabolism , Glucose/metabolism , Homeostasis , Humans , Lipids , Liver , Thermogenesis
6.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G610-G622, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29420068

ABSTRACT

We investigated whether vasoactive intestinal peptide (VIP) and/or prostaglandins contribute to peripheral corticotropin-releasing factor (CRF)-induced CRF1 receptor-mediated stimulation of colonic motor function and diarrhea in rats. The VIP antagonist, [4Cl-D-Phe6, Leu17]VIP injected intraperitoneally completely prevented CRF (10 µg/kg ip)-induced fecal output and diarrhea occurring within the first hour after injection, whereas pretreatment with the prostaglandins synthesis inhibitor, indomethacin, had no effect. In submucosal plexus neurons, CRF induced significant c-Fos expression most prominently in the terminal ileum compared with duodenum and jejunum, whereas no c-Fos was observed in the proximal colon. c-Fos expression in ileal submucosa was colocalized in 93.4% of VIP-positive neurons and 31.1% of non-VIP-labeled neurons. CRF1 receptor immunoreactivity was found on the VIP neurons. In myenteric neurons, CRF induced only a few c-Fos-positive neurons in the ileum and a robust expression in the proximal colon (17.5 ± 2.4 vs. 0.4 ± 0.3 cells/ganglion in vehicle). The VIP antagonist prevented intraperitoneal CRF-induced c-Fos induction in the ileal submucosal plexus and proximal colon myenteric plexus. At 60 min after injection, CRF decreased VIP levels in the terminal ileum compared with saline (0.8 ± 0.3 vs. 2.5 ± 0.7 ng/g), whereas VIP mRNA level detected by qPCR was not changed. These data indicate that intraperitoneal CRF activates intestinal submucosal VIP neurons most prominently in the ileum and myenteric neurons in the colon. It also implicates VIP signaling as part of underlying mechanisms driving the acute colonic secretomotor response to a peripheral injection of CRF, whereas prostaglandins do not play a role. NEW & NOTEWORTHY Corticotropin-releasing factor (CRF) in the gut plays a physiological role in the stimulation of lower gut secretomotor function induced by stress. We showed that vasoactive intestinal peptide (VIP)-immunoreactive neurons in the ileal submucosal plexus expressed CRF1 receptor and were prominently activated by CRF, unlike colonic submucosal neurons. VIP antagonist abrogated CRF-induced ileal submucosal and colonic myenteric activation along with functional responses (defecation and diarrhea). These data point to VIP signaling in ileum and colon as downstream effectors of CRF.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Diarrhea , Gastrointestinal Motility , Myenteric Plexus , Vasoactive Intestinal Peptide , Animals , Colon/metabolism , Colon/physiopathology , Defecation/drug effects , Defecation/physiology , Diarrhea/metabolism , Diarrhea/physiopathology , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Genes, fos/physiology , Ileum/metabolism , Ileum/physiopathology , Intestinal Mucosa/metabolism , Male , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Neuroprotective Agents/metabolism , Rats , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/metabolism
7.
J Med Chem ; 59(3): 854-66, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26789203

ABSTRACT

CRF mediates numerous stress-related endocrine, autonomic, metabolic, and behavioral responses. We present the synthesis and chemical and biological properties of astressin B analogues {cyclo(30-33)[D-Phe(12),Nle(21,38),C(α)MeLeu(27,40),Glu(30),Lys(33)]-acetyl-h/r-CRF(9-41)}. Out of 37 novel peptides, 17 (2, 4, 6-8, 10, 11, 16, 17, 27, 29, 30, 32-36) and 16 (3, 5, 9, 12-15, 18, 19, 22-26, 28, 31) had k(i) to CRF receptors in the high picomolar and low nanomole ranges, respectively. Peptides 1, 2, and 11 inhibited h/rCRF and urocortin 1-induced cAMP release from AtT20 and A7r5 cells. When Astressin C 2 was administered to adrenalectomized rats at 1.0 mg subcutaneously, it inhibited ACTH release for >7 d. Additional rat data based on the inhibitory effect of (2) on h/rCRF-induced stimulation of colonic secretory motor activity and urocortin 2-induced delayed gastric emptying also indicate a safe and long-lasting antagonistic effect. The overall properties of selected analogues may fulfill the criteria expected from clinical candidates.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Peptide Fragments/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/chemistry , Cyclic AMP/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Rats , Structure-Activity Relationship , Urocortins/antagonists & inhibitors
8.
PLoS One ; 10(9): e0139325, 2015.
Article in English | MEDLINE | ID: mdl-26421719

ABSTRACT

Abdominal surgery inhibits food intake and induces c-Fos expression in the hypothalamic and medullary nuclei in rats. Rikkunshito (RKT), a Kampo medicine improves anorexia. We assessed the alterations in meal microstructure and c-Fos expression in brain nuclei induced by abdominal surgery and the modulation by RKT in mice. RKT or vehicle was gavaged daily for 1 week. On day 8 mice had no access to food for 6-7 h and were treated twice with RKT or vehicle. Abdominal surgery (laparotomy-cecum palpation) was performed 1-2 h before the dark phase. The food intake and meal structures were monitored using an automated monitoring system for mice. Brain sections were processed for c-Fos immunoreactivity (ir) 2-h after abdominal surgery. Abdominal surgery significantly reduced bouts, meal frequency, size and duration, and time spent on meals, and increased inter-meal interval and satiety ratio resulting in 92-86% suppression of food intake at 2-24 h post-surgery compared with control group (no surgery). RKT significantly increased bouts, meal duration and the cumulative 12-h food intake by 11%. Abdominal surgery increased c-Fos in the prelimbic, cingulate and insular cortexes, and autonomic nuclei, such as the bed nucleus of the stria terminalis, central amygdala, hypothalamic supraoptic (SON), paraventricular and arcuate nuclei, Edinger-Westphal nucleus (E-W), lateral periaqueduct gray (PAG), lateral parabrachial nucleus, locus coeruleus, ventrolateral medulla and nucleus tractus solitarius (NTS). RKT induced a small increase in c-Fos-ir neurons in the SON and E-W of control mice, and in mice with surgery there was an increase in the lateral PAG and a decrease in the NTS. These findings indicate that abdominal surgery inhibits food intake by increasing both satiation (meal duration) and satiety (meal interval) and activates brain circuits involved in pain, feeding behavior and stress that may underlie the alterations of meal pattern and food intake inhibition. RKT improves food consumption post-surgically that may involve modulation of pain pathway.


Subject(s)
Abdomen/surgery , Brain/drug effects , Brain/metabolism , Drugs, Chinese Herbal/administration & dosage , Eating/drug effects , Plant Extracts/administration & dosage , Abdomen/pathology , Administration, Oral , Animals , Anorexia/drug therapy , Brain/cytology , Eating/physiology , Eating/psychology , Ghrelin/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy , Plants, Medicinal , Postoperative Complications/drug therapy , Postoperative Complications/pathology , Proto-Oncogene Proteins c-fos/immunology , Proto-Oncogene Proteins c-fos/metabolism , Satiation/drug effects , Satiety Response/drug effects , Tyrosine 3-Monooxygenase/metabolism
9.
Horm Behav ; 73: 15-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26026616

ABSTRACT

Somatostatin was discovered four decades ago as hypothalamic factor inhibiting growth hormone release. Subsequently, somatostatin was found to be widely distributed throughout the brain and to exert pleiotropic actions via interaction with five somatostatin receptors (sst1-5) that are also widely expressed throughout the brain. Interestingly, in contrast to the predominantly inhibitory actions of peripheral somatostatin, the activation of brain sst2 signaling by intracerebroventricular injection of stable somatostatin agonists potently stimulates food intake and independently, drinking behavior in rodents. The orexigenic response involves downstream orexin-1, neuropeptide Y1 and µ receptor signaling while the dipsogenic effect is mediated through the activation of the brain angiotensin 1 receptor. Brain sst2 activation is part of mechanisms underlying the stimulation of feeding and more prominently water intake in the dark phase and is able to counteract the anorexic response to visceral stressors.


Subject(s)
Appetite Regulation/genetics , Brain/physiology , Drinking Behavior/physiology , Feeding Behavior/physiology , Receptors, Somatostatin/physiology , Animals , Drinking/genetics , Eating/physiology , Humans , Receptors, Somatostatin/genetics , Rodentia , Somatostatin/physiology
10.
Am J Physiol Regul Integr Comp Physiol ; 307(7): R793-801, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25031229

ABSTRACT

Intracerebroventricular injection of stable somatostatin (SST) agonists stimulates food and water intake in rats. We investigated the receptor subtype(s) involved in the dipsogenic effect of intracerebroventricular injection of SST agonists, mechanisms of action, and role. In nonfasted and non-water-deprived male rats with chronic intracerebroventricular cannula, intake of water without food or food without water was monitored separately to avoid any interactions compared with intracerebroventricular vehicle. SST-14 and cortistatin (CST-14) (1 µg/rat icv) increased water intake by 3.1- and 2.7-fold, respectively, while both peptides did not alter food intake at 1 h postinjection in the light phase. By contrast, the stable pan-somatostatin agonist ODT8-SST (1 µg/rat icv) increased both water and food intake by 4.9- and 3.7-fold, respectively. S-346-011, a selective receptor 2 (sst2) agonist (1 µg/rat icv) induced water ingestion, while sst1 or sst4 agonist, injected under the same conditions, did not. The sst2 antagonist S-406-028 (1 µg/rat icv) prevented the 1-h water intake induced by intracerebroventricular ODT8-SST and CST-14. Losartan (100 µg/rat icv), an angiotensin receptor 1 (AT1) antagonist, completely blocked the water consumption induced by intracerebroventricular ODT8-SST, whereas intracerebroventricular injection of S-406-028 did not modify the intracerebroventricular ANG II-induced dipsogenic response. The sst2 antagonist reduced by 40% the increase of the 3-h water intake in the early dark phase. These data indicate that SST-14 and CST-14 interact with sst2 to exert a potent dipsogenic effect, which is mediated downstream by angiotensin-AT1 signaling. These data also indicate that sst2 activation by brain SST-14 and/or CST-14 may play an important role in the regulation of drinking behavior.


Subject(s)
Brain/drug effects , Drinking Behavior/drug effects , Eating/drug effects , Neuropeptides/pharmacology , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology , Animals , Behavior, Animal/drug effects , Brain/metabolism , Drinking/drug effects , Drinking/physiology , Eating/physiology , Injections, Intraventricular/methods , Male , Neuropeptides/administration & dosage , Rats, Sprague-Dawley , Somatostatin/administration & dosage
11.
Neurosci Lett ; 576: 88-92, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24915296

ABSTRACT

Intracerebroventricular (icv) injection of the stable somatostatin pan-agonist, ODT8-SST induces a somatostatin 2 receptor (sst2) mediated robust feeding response that involves neuropeptide Y and opioid systems in rats. We investigated whether the orexigenic system driven by orexin also plays a role. Food and water intake after icv injection was measured concomitantly in non-fasted and non-water deprived rats during the light phase. In vehicle treated rats (100% DMSO, icv), ODT8-SST (1µg/rat, icv) significantly increased the 2-h food and water intake compared to icv vehicle plus saline (5.1±1.0g vs. 1.2±0.4g and 11.3±1.9mL vs. 2.5±1.2mL, respectively). The orexin-1 receptor antagonist, SB-334867 (16µg/rat, icv) completely inhibited the 2-h food and water intake induced by icv ODT8-SST. In contrast, the icv pretreatment with the selective somatostatin sst2 antagonist, S-406-028, established to block the orexigenic effect of icv ODT8-SST, did not modify the increased food and water intake induced by icv orexin-A (10.7µg/rat). These data indicate that orexin-1 receptor signaling system is part of the brain neurocircuitry contributing to the orexigenic and dipsogenic responses induced by icv ODT8-SST and that orexin-A stimulates food intake independently from brain sst2 activation.


Subject(s)
Drinking/drug effects , Eating/drug effects , Peptide Fragments/pharmacology , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Animals , Benzoxazoles/pharmacology , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Naphthyridines , Neuropeptides/pharmacology , Octreotide/pharmacology , Orexin Receptor Antagonists , Orexin Receptors , Orexins , Rats, Sprague-Dawley , Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
12.
Peptides ; 55: 136-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24631952

ABSTRACT

We previously reported that ghrelin prevented l-dopa (LD)-induced inhibition of gastric emptying (GE) of a non-nutrient solution in rats. Parkinson's disease treatment involves the combined administration of l-dopa with the enzyme l-amino acid decarboxylase inhibitor, carbidopa (CD) to reduce peripheral formation of dopamine. We investigated the effect LD/CD given orogastrically (og) on GE of a non-nutrient or nutrient meal and whether og pretreatment with rikkunshito, a kampo medicine clinically used to treat gastroparesis, influenced LD/CD effect on GE and postprandial antral and duodenal motility in conscious rats. LD/CD (20/2 mgkg(-1)) decreased significantly GE to 26.3 ± 6.0% compared to 61.2 ± 3.2% in og vehicle monitored 20-min after a non-nutrient meal and to 41.9 ± 5.8% compared to 72.9 ± 5.2% in og vehicle monitored 60 min after a nutrient meal. Rikkunshito (0.5 or 1.0 g kg(-1)) reduced the LD/CD (20/2 mg kg(-1)) inhibition of GE of non-nutrient meal (36.9 ± 7.4% and 46.6 ± 4.8% respectively vs. 12.1 ± 7.4% in og vehicle plus LD/CD) while having no effect alone (56.6 ± 8.5%). The ghrelin antagonist, [d-Lys(3)]-GHRP-6 (1 mg kg(-1)) injected intraperitoneally partially reversed rikkunshito preventive effect on LD/CD-inhibited GE. Rikkunshito (1.0 g kg(-1)) blocked LD/CD (20/2 mg kg(-1))-induced delayed GE of a nutrient meal and the reduction of postprandial antral motility. In 6-hydroxydopamine-induced Parkinson's disease rat model, rikkunshito (1.0 g kg(-1), og) also prevented LD/CD-inhibited gastric emptying of a nutrient meal and enhanced fasting plasma levels of acylated ghrelin. These data indicate that oral rikkunshito alleviates the delayed GE induced by LD/CD in naïve and PD rat model in part through ghrelin-related mechanisms.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gastric Emptying/drug effects , Levodopa/pharmacology , Parkinson Disease, Secondary/physiopathology , Animals , Carbidopa/pharmacology , Ghrelin/blood , Male , Oligopeptides/pharmacology , Parkinson Disease, Secondary/blood , Parkinson Disease, Secondary/drug therapy , Postprandial Period , Rats, Sprague-Dawley , Receptors, Ghrelin/antagonists & inhibitors
13.
Am J Physiol Regul Integr Comp Physiol ; 305(6): R582-91, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23883680

ABSTRACT

Obesity is an increasing health problem. Because drug treatments are limited, diets remain popular. High-protein diets (HPD) reduce body weight (BW), although the mechanisms are unclear. We investigated physiological mechanisms altered by switching diet induced obesity (DIO) rats from Western-type diet (WTD) to HPD. Male rats were fed standard (SD) or WTD (45% calories from fat). After developing DIO (50% of rats), they were switched to SD (15% calories from protein) or HPD (52% calories from protein) for up to 4 weeks. Food intake (FI), BW, body composition, glucose tolerance, insulin sensitivity, and intestinal hormone plasma levels were monitored. Rats fed WTD showed an increased FI and had a 25% greater BW gain after 9 wk compared with SD (P < 0.05). Diet-induced obese rats switched from WTD to HPD reduced daily FI by 30% on day 1, which lasted to day 9 (-9%) and decreased BW during the 2-wk period compared with SD/SD (P < 0.05). During these 2 wk, WTD/HPD rats lost 72% more fat mass than WTD/SD (P < 0.05), whereas lean mass was unaltered. WTD/HPD rats had lower blood glucose than WTD/SD at 30 min postglucose gavage (P < 0.05). The increase of pancreatic polypeptide and peptide YY during the 2-h dark-phase feeding was higher in WTD/HPD compared with WTD/SD (P < 0.05). These data indicate that HPD reduces BW in WTD rats, which may be related to decreased FI and the selective reduction of fat mass accompanied by improved glucose tolerance, suggesting relevant benefits of HPD in the treatment of obesity.


Subject(s)
Adiposity/drug effects , Dietary Proteins/administration & dosage , Dietary Proteins/metabolism , Glucose Tolerance Test , Glucose/metabolism , Obesity/diet therapy , Obesity/metabolism , Animals , Body Weight , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome
14.
Obesity (Silver Spring) ; 19(3): 514-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20706204

ABSTRACT

An obesity-induced diabetes model using genetically normal mouse strains would be invaluable but remains to be established. One reason is that several normal mouse strains are resistant to high-fat diet-induced obesity. In the present study, we show the effectiveness of gold thioglucose (GTG) in inducing hyperphagia and severe obesity in mice, and demonstrate the development of obesity-induced diabetes in genetically normal mouse strains. GTG treated DBA/2, C57BLKs, and BDF1 mice gained weight rapidly and exhibited significant increases in nonfasting plasma glucose levels 8-12 weeks after GTG treatment. These mice showed significantly impaired insulin secretion, particularly in the early phase after glucose load, and reduced insulin content in pancreatic islets. Interestingly, GTG treated C57BL/6 mice did not become diabetic and retained normal early insulin secretion and islet insulin content despite being as severely obese and insulin resistant as the other mice. These results suggest that the pathogenesis of obesity-induced diabetes in GTG-treated mice is attributable to the inability of their pancreatic ß-cells to secrete enough insulin to compensate for insulin resistance. Mice developing obesity-induced diabetes after GTG treatment might be a valuable tool for investigating obesity-induced diabetes. Furthermore, comparing the genetic backgrounds of mice with different susceptibilities to diabetes may lead to the identification of novel genetic factors influencing the ability of pancreatic ß-cells to secrete insulin.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Disease Models, Animal , Hyperphagia/chemically induced , Insulin-Secreting Cells/physiology , Insulin/metabolism , Obesity, Morbid/complications , Animals , Aurothioglucose , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/physiopathology , Genetic Predisposition to Disease , Glucose Intolerance , Insulin Resistance/genetics , Insulin Resistance/physiology , Insulin Secretion , Male , Mice , Mice, Inbred Strains , Obesity, Morbid/chemically induced , Obesity, Morbid/physiopathology , Species Specificity
15.
Biochem Biophys Res Commun ; 401(1): 79-84, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20833146

ABSTRACT

We have previously reported that obesity-induced diabetes developed in high-fat diet (HFD)-fed BDF1 mice. This is caused by insufficient insulin response to an excess glucose load. In this study, we have shown that the enhanced expression of retinaldehyde dehydrogenase 3 (Raldh3) causes functional disorders of pancreatic islets in diabetic mouse models. In the pancreatic islets of HFD-induced diabetic BDF1 mice and spontaneously diabetic C57BL/KsJ(db/db) mice, gene expression analysis with oligonucleotide microarray revealed a significant increase in Raldh3 expression. Exposure to a culture medium containing a higher glucose concentration (25 mM) significantly increased Raldh3 expression in murine MIN6 and alphaTC1 clone 9 cells, which derived from the α and ß-cells of pancreatic islets, respectively. Overexpression of Raldh3 reduced the insulin secretion in MIN6 cells, and surprisingly, increased the glucagon secretion in alphaTC1 clone 9 cells. Furthermore, the knockdown of Raldh3 expression with siRNA decreased the glucagon secretion in alphaTC1 clone 9 cells. Raldh3 catalyzes the conversion of 13-cis retinal to 13-cis retinoic acid and we revealed that 13-cis retinoic acid significantly reduces cell viability in MIN6 and alphaTC1 clone 9 cells, but not in cells of H4IIEC3, 3T3-L1, and COS-1 cell lines. These findings suggest that an increasing expression of Raldh3 deregulates the balanced mechanisms of insulin and glucagon secretion in the pancreatic islets and may induce ß-cell dysfunction leading to the development of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Glucagon/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Retinal Dehydrogenase/biosynthesis , 3T3-L1 Cells , Animals , COS Cells , Chlorocebus aethiops , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Isotretinoin/pharmacology , Mice , Mice, Inbred C57BL , Retinal Dehydrogenase/genetics
16.
J Occup Health ; 52(5): 278-86, 2010.
Article in English | MEDLINE | ID: mdl-20697184

ABSTRACT

OBJECTIVE: To assess the usefulness of the Laboratory of Physical Science (LOPS) protocol for detecting exaggerated blood pressure (BP) response as a risk factor for future hypertension when controlling for work and personal factors. METHODS: Subjects were 815 healthy normotensive men (mean age, 43.1 ± 6.76 yr; range, 29-64 yr) who participated in the LOPS protocol, a graded 4-stage exercise test undertaken for the measurement of 40-70% of maximum oxygen consumption. A hypertensive response was defined as systolic blood pressure (SBP) of 250 mmHg or diastolic blood pressure (DBP) of 120 mmHg during the exercise test. RESULTS: New-onset hypertension or the initiation of antihypertensive drug treatment had occurred in 108 men (13.3%) after 7 yr of follow-up. Cox proportional survival analysis revealed significantly increased risks of developing hypertension were associated with exaggerated BP response to exercise (hazard ratio, 2.3; 95% confidence interval, 1.4-3.7) and higher frequency of business trips (hazard ratio, 1.7; 95% confidence interval, 1.2-2.5) after multivariable adjustments for work and personal-related risk factors. CONCLUSIONS: These results suggest that the LOPS protocol is effective for detecting exaggerated BP response as a risk factor for future hypertension when controlling for work and personal-related risk factors. Exaggerated BP response to exercise and higher frequency of business trips are risk factors for developing future hypertension.


Subject(s)
Blood Pressure/physiology , Hypertension/epidemiology , Adult , Employment , Exercise Test , Follow-Up Studies , Humans , Male , Middle Aged , Predictive Value of Tests , Risk Factors , Survival Analysis , Travel
17.
Metabolism ; 58(3): 296-303, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19217442

ABSTRACT

For experimental research on type 2 diabetes mellitus, a diet-induced obesity-dependent diabetes model developed using genetically normal animals is essential. However, attempts at feeding a high-fat diet (HFD) to major inbred strains of mice have not resulted in the establishment of an ideal model. Here, we show that BDF1 mice, the F(1) hybrids of C57BL/6 and DBA/2 normal strains, develop HFD-induced obesity-dependent diabetes. BDF1 mice fed a HFD gained weight rapidly and developed severe diabetes characterized by hyperglycemia, glucosuria, and elevation of hemoglobin A(1C) levels in 3 to 4 months. The glucose tolerance of the diabetic mice was significantly impaired, and the elevation of plasma insulin after a glucose load was significantly reduced. Isolated pancreatic islets of HFD-fed BDF1 mice showed decreased insulin content and a reduced insulin secretory response to higher concentrations of glucose. Immunohistochemical analysis of the pancreas showed reduced staining intensity to insulin and aberrant distribution of glucagon-positive cells in diabetic BDF1 mice. These observations suggest the cause of the diabetes in HFD-fed BDF1 mice to be dysfunction of the pancreatic beta-cells, which do not produce or secrete enough insulin to compensate for insulin resistance. BDF1 mice fed a HFD showing obesity-dependent diabetes are suggested to be an appropriate animal model of type 2 diabetes mellitus. This model would be useful for exploring the mechanism of obesity-dependent type 2 diabetes mellitus and evaluating antiobesity and antidiabetic drugs.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/etiology , Dietary Fats , Obesity/complications , Weight Gain , Animals , Cholesterol/blood , Fatty Acids, Nonesterified/blood , Glucose Tolerance Test , Glycosuria/urine , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Models, Biological , Triglycerides/blood
18.
Nihon Hoshasen Gijutsu Gakkai Zasshi ; 60(1): 111-7, 2004 Jan.
Article in Japanese | MEDLINE | ID: mdl-15041913

ABSTRACT

We examined ROC curve fitting with continuous rating data by using a corrected mean value and corrected standard deviation as binormal ROC parameters. Curve fitting was performed by using corrected categorical data in the same way as the continuous data. A large residual of curve fitting was found in cases of difference between means of positive and negative signal data greater than three times the standard deviation, or with a ratio of the standard deviation of positive and negative signal data above six. We conclude that our proposed method performed well for ROC curve fitting except in some special cases.


Subject(s)
Data Interpretation, Statistical , ROC Curve , Statistical Distributions
19.
Eur J Pharmacol ; 468(1): 15-9, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12729838

ABSTRACT

Motor dysfunction associated with dyskinesia can be caused by imbalance between dopaminergic and cholinergic actions. Antimuscarinic agents are used to treat extrapyramidal symptoms in Parkinson's disease and extrapyramidal side effects of antipsychotics. These therapeutic effects are mediated by blockade of the striatal muscarinic receptors, which comprise five distinct subtypes (M(1-5)). To evaluate the role of muscarinic M(4) receptors, we have generated mutant mice lacking this subtype (muscarinic M(4) receptor-knockout mice) and analyzed their cataleptic responses induced by haloperidol (an animal model of extrapyramidal side effects). While the muscarinic M(4) receptor-knockout mice developed the cataleptic response normally, systemic administration of scopolamine could not suppress the cataleptic response. These results suggest that acute, but not chronic, blockade of muscarinic M(4) receptors plays important roles in the therapeutic effects of antimuscarinic agents.


Subject(s)
Behavior, Animal/drug effects , Catalepsy/physiopathology , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/deficiency , Scopolamine/pharmacology , Animals , Blotting, Northern , Catalepsy/chemically induced , Catalepsy/prevention & control , Haloperidol/pharmacology , Mice , Mice, Knockout , Receptor, Muscarinic M4
SELECTION OF CITATIONS
SEARCH DETAIL
...