1
|
Ortiz-Guzman J, Swanson JL, Tantry EK, Kochukov M, Ung K, Addison AP, Srivastava S, Belfort BD, Ji E, Dooling SW, Chen SA, Tong Q, Arenkiel BR. Cholinergic Basal Forebrain Connectivity to the Basolateral Amygdala Modulates Food Intake. eNeuro 2024; 11:ENEURO.0369-23.2024. [PMID: 38383587 PMCID: PMC10915460 DOI: 10.1523/eneuro.0369-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
Obesity results from excessive caloric input associated with overeating and presents a major public health challenge. The hypothalamus has received significant attention for its role in governing feeding behavior and body weight homeostasis. However, extrahypothalamic brain circuits also regulate appetite and consumption by altering sensory perception, motivation, and reward. We recently discovered a population of basal forebrain cholinergic (BFc) neurons that regulate appetite suppression. Through viral tracing methods in the mouse model, we found that BFc neurons densely innervate the basolateral amygdala (BLA), a limbic structure involved in motivated behaviors. Using channelrhodopsin-assisted circuit mapping, we identified cholinergic responses in BLA neurons following BFc circuit manipulations. Furthermore, in vivo acetylcholine sensor and genetically encoded calcium indicator imaging within the BLA (using GACh3 and GCaMP, respectively) revealed selective response patterns of activity during feeding. Finally, through optogenetic manipulations in vivo, we found that increased cholinergic signaling from the BFc to the BLA suppresses appetite and food intake. Together, these data support a model in which cholinergic signaling from the BFc to the BLA directly influences appetite and feeding behavior.
Collapse
Affiliation(s)
- Joshua Ortiz-Guzman
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Jessica L Swanson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Evelyne K Tantry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Mikhail Kochukov
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Kevin Ung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Angela P Addison
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Snigdha Srivastava
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Benjamin D Belfort
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Emily Ji
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Sean W Dooling
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Sarah A Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Qingchun Tong
- Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
2
|
Bahn YJ, Yadav H, Piaggi P, Abel BS, Gavrilova O, Springer DA, Papazoglou I, Zerfas PM, Skarulis MC, McPherron AC, Rane SG. CDK4-E2F3 signals enhance oxidative skeletal muscle fiber numbers and function to affect myogenesis and metabolism. J Clin Invest 2023; 133:e162479. [PMID: 37395281 PMCID: PMC10313363 DOI: 10.1172/jci162479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/19/2023] [Indexed: 07/04/2023] Open
Abstract
Understanding how skeletal muscle fiber proportions are regulated is vital to understanding muscle function. Oxidative and glycolytic skeletal muscle fibers differ in their contractile ability, mitochondrial activity, and metabolic properties. Fiber-type proportions vary in normal physiology and disease states, although the underlying mechanisms are unclear. In human skeletal muscle, we observed that markers of oxidative fibers and mitochondria correlated positively with expression levels of PPARGC1A and CDK4 and negatively with expression levels of CDKN2A, a locus significantly associated with type 2 diabetes. Mice expressing a constitutively active Cdk4 that cannot bind its inhibitor p16INK4a, a product of the CDKN2A locus, were protected from obesity and diabetes. Their muscles exhibited increased oxidative fibers, improved mitochondrial properties, and enhanced glucose uptake. In contrast, loss of Cdk4 or skeletal muscle-specific deletion of Cdk4's target, E2F3, depleted oxidative myofibers, deteriorated mitochondrial function, and reduced exercise capacity, while increasing diabetes susceptibility. E2F3 activated the mitochondrial sensor PPARGC1A in a Cdk4-dependent manner. CDK4, E2F3, and PPARGC1A levels correlated positively with exercise and fitness and negatively with adiposity, insulin resistance, and lipid accumulation in human and rodent muscle. All together, these findings provide mechanistic insight into regulation of skeletal muscle fiber-specification that is of relevance to metabolic and muscular diseases.
Collapse
Affiliation(s)
- Young Jae Bahn
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hariom Yadav
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona
| | - Brent S. Abel
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases
| | | | - Ioannis Papazoglou
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Monica C. Skarulis
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Alexandra C. McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Sushil G. Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Tang Y, Du X, Sun S, Shi W, Han Y, Zhou W, Zhang J, Teng S, Ren P, Liu G. Circadian Rhythm and Neurotransmitters Are Potential Pathways through Which Ocean Acidification and Warming Affect the Metabolism of Thick-Shell Mussels. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:4324-4335. [PMID: 35293730 DOI: 10.1021/acs.est.1c06735] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although the impacts of ocean acidification and warming on marine organisms have been increasingly documented, little is known about the affecting mechanism underpinning their interactive impacts on physiological processes such as metabolism. Therefore, the effects of these two stressors on metabolism were investigated in thick-shell mussel Mytilus coruscus in this study. In addition, because metabolism is primarily regulated by circadian rhythm and neurotransmitters, the impacts of acidification and warming on these two regulatory processes were also analyzed. The data obtained demonstrated that the metabolism of mussels (indicated by the clearance rate, oxygen consumption rate, ammonia excretion rate, O:N ratio, ATP content, activity of pyruvate kinase, and expression of metabolism-related genes) were significantly affected by acidification and warming, resulting in a shortage of energy supply (indicated by the in vivo content of ATP). In addition, exposure to acidification and warming led to evident disruption in circadian rhythm (indicated by the heartrate and the expression rhythm of Per2, Cry, and BMAL1) and neurotransmitters (indicated by the activity of acetyl cholinesterase and in vivo contents of ACh, GABA, and DA). These findings suggest that circadian rhythms and neurotransmitters might be potential routes through which acidification and warming interactively affect the metabolism of mussels.
Collapse
Affiliation(s)
- Yu Tang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Xueying Du
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Shuge Sun
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yu Han
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Jiongming Zhang
- Zhejiang Mariculture Research Institute, Wenzhou 325005, P.R. China
| | | | - Peng Ren
- Zhejiang Mariculture Research Institute, Wenzhou 325005, P.R. China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| |
Collapse
|
4
|
Neuro-immune-metabolism: The tripod system of homeostasis. Immunol Lett 2021; 240:77-97. [PMID: 34655659 DOI: 10.1016/j.imlet.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 11/20/2022]
Abstract
Homeostatic regulation of cellular and molecular processes is essential for the efficient physiological functioning of body organs. It requires an intricate balance of several networks throughout the body, most notable being the nervous, immune and metabolic systems. Several studies have reported the interactions between neuro-immune, immune-metabolic and neuro-metabolic pathways. Current review aims to integrate the information and show that neuro, immune and metabolic systems form the triumvirate of homeostasis. It focuses on the cellular and molecular interactions occurring in the extremities and intestine, which are innervated by the peripheral nervous system and for the intestine in particular the enteric nervous system. While the interdependence of neuro-immune-metabolic pathways provides a fallback mechanism in case of disruption of homeostasis, in chronic pathologies of continued disequilibrium, the collapse of one system spreads to the other interacting networks as well. Current review illustrates this domino-effect using diabetes as the main example. Together, this review attempts to provide a holistic picture of the integrated network of neuro-immune-metabolism and attempts to broaden the outlook when devising a scientific study or a treatment strategy.
Collapse
|
5
|
Takahashi T, Shiraishi A, Murata J, Matsubara S, Nakaoka S, Kirimoto S, Osawa M. Muscarinic receptor M3 contributes to intestinal stem cell maintenance via EphB/ephrin-B signaling. Life Sci Alliance 2021; 4:4/9/e202000962. [PMID: 34244422 PMCID: PMC8321669 DOI: 10.26508/lsa.202000962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/31/2022] Open
Abstract
Acetylcholine (ACh) signaling through activation of nicotinic and muscarinic ACh receptors regulates expression of specific genes that mediate and sustain proliferation, differentiation, and homeostasis in the intestinal crypts. This signaling plays a pivotal role in the regulation of intestinal stem cell function, but the details have not been clarified. Here, we performed experiments using type 3 muscarinic acetylcholine receptor (M3) knockout mice and their intestinal organoids and report that endogenous ACh affects the size of the intestinal stem niche via M3 signaling. RNA sequencing of crypts identified up-regulation of the EphB/ephrin-B signaling pathway. Furthermore, using an MEK inhibitor (U0126), we found that mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, which is downstream of EphB/ephrin-B signaling, is activated in M3-deficient crypts. Collectively, M3, EphB/ephrin-B, and the MAPK/ERK signaling cascade work together to maintain the homeostasis of intestinal epithelial cell growth and differentiation following modifications of the cholinergic intestinal niche.
Collapse
Affiliation(s)
- Toshio Takahashi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Akira Shiraishi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Jun Murata
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Shin Matsubara
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | | | | | - Masatake Osawa
- Department of Regenerative Medicine and Applied Biomedical Sciences, Graduate School of Medicine, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu, Japan
| |
Collapse
|
6
|
Carli M, Kolachalam S, Longoni B, Pintaudi A, Baldini M, Aringhieri S, Fasciani I, Annibale P, Maggio R, Scarselli M. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals (Basel) 2021; 14:238. [PMID: 33800403 PMCID: PMC8001502 DOI: 10.3390/ph14030238] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Atypical antipsychotics (AAPs) are commonly prescribed medications to treat schizophrenia, bipolar disorders and other psychotic disorders. However, they might cause metabolic syndrome (MetS) in terms of weight gain, dyslipidemia, type 2 diabetes (T2D), and high blood pressure, which are responsible for reduced life expectancy and poor adherence. Importantly, there is clear evidence that early metabolic disturbances can precede weight gain, even if the latter still remains the hallmark of AAPs use. In fact, AAPs interfere profoundly with glucose and lipid homeostasis acting mostly on hypothalamus, liver, pancreatic β-cells, adipose tissue, and skeletal muscle. Their actions on hypothalamic centers via dopamine, serotonin, acetylcholine, and histamine receptors affect neuropeptides and 5'AMP-activated protein kinase (AMPK) activity, thus producing a supraphysiological sympathetic outflow augmenting levels of glucagon and hepatic glucose production. In addition, altered insulin secretion, dyslipidemia, fat deposition in the liver and adipose tissues, and insulin resistance become aggravating factors for MetS. In clinical practice, among AAPs, olanzapine and clozapine are associated with the highest risk of MetS, whereas quetiapine, risperidone, asenapine and amisulpride cause moderate alterations. The new AAPs such as ziprasidone, lurasidone and the partial agonist aripiprazole seem more tolerable on the metabolic profile. However, these aspects must be considered together with the differences among AAPs in terms of their efficacy, where clozapine still remains the most effective. Intriguingly, there seems to be a correlation between AAP's higher clinical efficacy and increase risk of metabolic alterations. Finally, a multidisciplinary approach combining psychoeducation and therapeutic drug monitoring (TDM) is proposed as a first-line strategy to avoid the MetS. In addition, pharmacological treatments are discussed as well.
Collapse
Affiliation(s)
- Marco Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Anna Pintaudi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Marco Baldini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Stefano Aringhieri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.F.); (R.M.)
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany;
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.F.); (R.M.)
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| |
Collapse
|
7
|
Intestinal Electrical Stimulation Enhances Release of Postprandial Incretin Hormones Via Cholinergic Mechanisms. Obes Surg 2021; 31:1957-1966. [PMID: 33469859 DOI: 10.1007/s11695-021-05228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Intestinal electrical stimulation (IES) has been reported to reduce body weight and improve glucose tolerance in obese and diabetic rats. Our study aimed to investigate possible IES mechanisms involving incretin hormones using intraduodenal glucose infusion in rats. We hypothesized that the enhanced release of postprandial glucagon-like peptide-1 (GLP-1) at early phase by IES was mediated through neuro/paracrine mechanisms involving the vagal nerve and glucose-dependent insulinotropic peptide (GIP). METHODS Fifteen normal male Sprague-Dawley rats chronically implanted with duodenal electrodes for IES, and an intra-duodenum catheter for the infusion of glucose were studied in a series of sessions with IES of different parameters with and without atropine and M3 receptor antagonist. Blood samples were collected via the tail vein for the measurement of blood glucose, and plasma GLP-1, and GIP. RESULTS (1) Compared to sham-IES, IES of 0.3 ms reduced blood glucose by 16.5-28.4% between 30 and 120 min (all time points p < 0.05), and IES of 3-ms reduced blood glucose at 60 (12.6%) and 90 min (11.8%). IES of 0.3 ms showed a greater hypoglycemic effect than 3 ms (p = 0.024) at 30 min. (2) IES elevated plasma GLP-1 with 0.3 ms (p = 0.001) and with 3 ms p = 0.03). (3) IES substantially elevated plasma GIP with 0.3 ms (p = 0.002) and with 3 ms (p < 0.001). (4) Pretreatment of atropine and the M3 receptor antagonist 4-DAMP blocked the effects of IES on GLP-1, GIP, and blood glucose. CONCLUSIONS IES reduces postprandial blood glucose by enhancing the release of GLP-1 and GIP mediated via the cholinergic mechanism.
Collapse
|
8
|
Vincent SG, Fisher JT. In vivo cardiopulmonary impact of skeletal M 3Dq DREADD expression: a pilot study. J Comp Physiol B 2021; 191:1059-1070. [PMID: 34272586 PMCID: PMC8572194 DOI: 10.1007/s00360-021-01387-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/23/2021] [Accepted: 07/06/2021] [Indexed: 01/07/2023]
Abstract
The muscarinic M3 receptor (M3R) is implicated in cardiopulmonary control and many other peripheral physiologic functions. Previous observations report mortality in mice expressing a Gq-linked designer G-protein coupled receptor (Dq) selectively in striated muscle, while M3Dq DREADD (Designer Receptor Exclusively Activated by Designer Drug), selectively expressed in skeletal muscle (SKM) impacts glucose metabolism. We investigated whether activation of SKM M3Dq impacts cardiopulmonary function. Heart rate (HR), body temperature (Tb) and locomotor activity (ACT) were measured in 4 conscious, chronically instrumented M3Dq DREADD mice and 4 wildtype controls. Circadian values of HR, BT and ACT were not different between genotypes (p > 0.05). Activation of the M3Dq DREADD by clozapine N-oxide (CNO; 0.1 mg/kg) resulted in: a significant drop in heart rate, 2 h after injection, compared with a time-matched baseline control period from the same animals (460 ± 28 vs. 532 ± 6, p < 0.05), significantly lower ACT compared to the baseline control (p < 0.05) and reduced pulmonary minute ventilation compared to pre-CNO control (p < 0.05). M3Dq DREADD activation did not cause bronchoconstriction (separate protocol), however, there was a concomitant reduction in HR, Tb and ventilation, accompanied by cardiac arrhythmias. We speculate that reductions in Tb, HR and ventilation reflect a mechanistic link between SKM Gq signaling and the metabolic responses associated with the initiation of torpor. Supported by the Canadian Institutes of Health Research (CIHR MOP-81211).
Collapse
Affiliation(s)
- Sandra G. Vincent
- Department of Biomedical and Molecular Sciences and Division of Respirology, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - John T. Fisher
- Department of Biomedical and Molecular Sciences and Division of Respirology, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6 Canada
| |
Collapse
|
9
|
Saiyasit N, Chunchai T, Apaijai N, Pratchayasakul W, Sripetchwandee J, Chattipakorn N, Chattipakorn SC. Chronic high-fat diet consumption induces an alteration in plasma/brain neurotensin signaling, metabolic disturbance, systemic inflammation/oxidative stress, brain apoptosis, and dendritic spine loss. Neuropeptides 2020; 82:102047. [PMID: 32327191 DOI: 10.1016/j.npep.2020.102047] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Chronic high-fat diet (HFD) consumption caused not only negative effects on obesity and metabolic disturbance, but also instigated several brain pathologies, including dendritic spine loss. In addition, alterations in plasma/brain neurotensin (NT) levels and NT signaling were observed in obesity. However, the mechanistic link between the NT levels in plasma and brain, NT signaling, and peripheral/brain pathologies following prolonged HFD consumption still needs to be elucidated. We hypothesized that an increase in peripheral/brain NT signaling were associated with peripheral/brain pathologies after prolonged HFD consumption. Male Wistar rats (n = 24) were given either a normal diet (ND) or a HFD for 12 and 40 weeks. At the end of each time course, metabolic parameters and plasma NT levels were measured. Rats were then decapitated and the brains were examined the levels of brain NT, hippocampal reactive oxygen species, the number of Iba-1 positive cells, the dendritic spine densities, and the expression of NT-, mitophagy-, autophagy-, and apoptotic-related proteins. The findings showed an increase in the level of plasma NT with dyslipidemia, metabolic disturbances, systemic inflammation/oxidative stress, and hippocampal pathologies in rats fed HFD for 12 and 40 weeks. The expression of brain NT signaling and brain apoptosis were markedly increased after 40 weeks of HFD feeding. These results indicated that the alteration in the level of circulating/brain NT and its downstream signaling were associated with central and peripheral pathologies after long-term HFD intake. Therefore, these alterations in NT level or its signaling could be considered as a therapeutic target in treating obesity.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirapas Sripetchwandee
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
10
|
Adipocyte G i signaling is essential for maintaining whole-body glucose homeostasis and insulin sensitivity. Nat Commun 2020; 11:2995. [PMID: 32532984 PMCID: PMC7293267 DOI: 10.1038/s41467-020-16756-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Adipocyte dysfunction links obesity to insulin resistance and type 2 diabetes. Adipocyte function is regulated by receptor-mediated activation of heterotrimeric G proteins. Little is known about the potential in vivo metabolic roles of Gi-type G proteins expressed by adipocytes, primarily due to the lack of suitable animal models. To address this question, we generated mice lacking functional Gi proteins selectively in adipocytes. Here we report that these mutant mice displayed significantly impaired glucose tolerance and reduced insulin sensitivity when maintained on an obesogenic diet. In contrast, using a chemogenetic strategy, we demonstrated that activation of Gi signaling selectively in adipocytes greatly improved glucose homeostasis and insulin signaling. We also elucidated the cellular mechanisms underlying the observed metabolic phenotypes. Our data support the concept that adipocyte Gi signaling is essential for maintaining euglycemia. Drug-mediated activation of adipocyte Gi signaling may prove beneficial for restoring proper glucose homeostasis in type 2 diabetes. Gs-coupled receptor signaling is well known to modulate adipocyte metabolism, but the role of Gi-coupled receptors in adipose tissue is less well understood. Here the authors show that signaling via Gi-type G proteins expressed by adipocytes is essential for maintaining proper blood glucose homeostasis.
Collapse
|
11
|
Rodriguez-Diaz R, Tamayo A, Hara M, Caicedo A. The Local Paracrine Actions of the Pancreatic α-Cell. Diabetes 2020; 69:550-558. [PMID: 31882565 PMCID: PMC7085245 DOI: 10.2337/dbi19-0002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Secretion of glucagon from the pancreatic α-cells is conventionally seen as the first and most important defense against hypoglycemia. Recent findings, however, show that α-cell signals stimulate insulin secretion from the neighboring β-cell. This article focuses on these seemingly counterintuitive local actions of α-cells and describes how they impact islet biology and glucose metabolism. It is mostly based on studies published in the last decade on the physiology of α-cells in human islets and incorporates results from rodents where appropriate. As this and the accompanying articles show, the emerging picture of α-cell function is one of increased complexity that needs to be considered when developing new therapies aimed at promoting islet function in the context of diabetes.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Tamayo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, IL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
12
|
Lian J, Deng C. The dosage-dependent effects of cevimeline in preventing olanzapine-induced metabolic side-effects in female rats. Pharmacol Biochem Behav 2020; 191:172878. [PMID: 32112786 DOI: 10.1016/j.pbb.2020.172878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/28/2020] [Accepted: 02/20/2020] [Indexed: 01/08/2023]
Abstract
Olanzapine has been used for the treatment of schizophrenia and other mental disorders. However, it is associated with serious weight gain and other metabolic side-effects. The antagonistic affinity of olanzapine to muscarinic M3 receptors has been evidenced as one of the main contributors for its weight gain and other metabolic side-effects. Therefore, this study investigated whether the co-treatment of cevimeline (a M3 receptor agonist) could prevent the metabolic side-effects associated with olanzapine medication. Female Sprague Dawley rats were treated orally with olanzapine (2 mg/kg, t.i.d.) and/or cevimeline at 3 dosages (3, 6, 9 mg/kg, t.i.d.), or vehicle for two weeks. Weight gain and food/water intake were measured throughout the drug treatment period. Intraperitoneal glucose tolerance tests and open field tests were conducted. Olanzapine-treated rats demonstrated significantly elevated body weight gain, food intake, feeding efficiency, total white fat mass, liver mass, and plasma triglyceride levels, which could be partly reversed by the co-treatment with cevimeline in a dosage-dependent manner. In general, the body weight gain can only be reversed by the co-treatment of 9 mg/kg cevimeline. The cevimeline co-treatment decreased plasma triglyceride and glucose levels compared with olanzapine only treatment. The results suggested a dosage-dependent effect of cevimeline in ameliorating olanzapine-induced weight gain and metabolic side-effects, which supports further clinical trials using cevimeline to control weight gain and metabolic side-effects caused by antipsychotic medications.
Collapse
Affiliation(s)
- Jiamei Lian
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia; School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong 2522, NSW, Australia
| | - Chao Deng
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia; School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong 2522, NSW, Australia.
| |
Collapse
|
13
|
Sarsenbayeva A, Marques-Santos CM, Thombare K, Di Nunzio G, Almby KE, Lundqvist M, Eriksson JW, Pereira MJ. Effects of second-generation antipsychotics on human subcutaneous adipose tissue metabolism. Psychoneuroendocrinology 2019; 110:104445. [PMID: 31563732 DOI: 10.1016/j.psyneuen.2019.104445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Metabolic syndrome is prevalent in up to 50% of schizophrenia patients, which reduces their quality of life and their compliance with the treatment. It is unclear whether metabolic adverse effects of these agents are due to their direct effect on insulin-sensitive tissues or are secondary to increased adiposity. The study aimed to investigate the direct effects of the second-generation antipsychotics olanzapine and aripiprazole on human subcutaneous adipose tissue and isolated adipocyte metabolism. METHODS Abdominal subcutaneous adipose tissue needle biopsies were taken from 72 healthy subjects (49 F/23 M; age: 19-78 yr; BMI: 20.0-35.6 kg/m2). Isolated adipocytes or adipose tissue were respectively pre-incubated short- (30 min) and long-term (24 h, 72 h) with or without olanzapine (0.004 μM - 20 μM) and aripiprazole (0.002 μM - 100 μM). Pre-incubated adipose tissue was then snap-frozen for mRNA expression analysis of adipokines genes and genes involved in inflammation, adipogenesis, and mitochondrial function. Isolated adipocytes were used to measure basal and insulin-stimulated glucose uptake and lipolysis. RESULTS Acute treatment with a therapeutic concentration of olanzapine decreases basal lipolysis in isolated adipocytes; this effect was not observed after long-term incubation with the drug. Supra-therapeutic concentration of aripiprazole reduced basal and insulin-stimulated glucose uptake after short- and long-term pre-incubation. Both drugs at supra-therapeutic concentrations downregulated the expression of the pro-inflammatory cytokines IL6 and IL1B genes after 72 h incubation. Similarly, supra-therapeutic concentrations of both drugs and therapeutic concentration of olanzapine, reduced the expression of PPARGC1A, PDK4, and CPT1B genes involved in the regulation of mitochondrial functions. Neither of the antipsychotics affected the expression of the main adipokines LEP and ADIPOQ, genes involved in the regulation of lipid metabolism, LPL and FASN, nor the master adipogenesis regulator, PPARG. CONCLUSION Therapheutic concentrations of olanzapine and aripiprazole have a moderate direct effect on adipocyte lipid and glucose metabolism, respectively. At supra-therapeutic concentrations, both of the antipsychotics seem to act as anti-inflammatory agents and mildly suppressed genes involved in the regulation of mitochondrial functions, which could potentially contribute to metabolic adverse effects. Alternatively, second-generation antipsychotics could induce metabolic side effects via acting on other insulin-sensitive tissues and central nervous system.
Collapse
Affiliation(s)
- Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Cátia M Marques-Santos
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Ketan Thombare
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Giada Di Nunzio
- The Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Kristina E Almby
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Martin Lundqvist
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Abu Zeid O, Abdel-Aziz A, Rashed LA, Said ER. Role of the cutaneous extraneuronal cholinergic system in the pathogenesis of psoriasis: a case-control study. Clin Exp Dermatol 2019; 45:432-437. [PMID: 31614011 DOI: 10.1111/ced.14124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although during recent years there have been considerable advances in elucidating the mechanisms of psoriasis pathogenesis, its full understanding is still distant. A cholinergic dysfunction has been proposed in the pathophysiology of some inflammatory and autoimmune diseases, including psoriasis. AIM To determine tissue levels of acetylcholine (ACh) and its muscarinic and nicotinic receptors (mAChR and nAChR) in psoriasis vulgaris lesions in comparison with normal control skin. METHODS This case-control study included 30 patients with psoriasis vulgaris and 30 controls. A 4-mm punch skin biopsy was taken from the psoriatic plaques of patients and normal skin of controls. ACh level was measured in tissues using the colorimetric method, while mAChR and nAChR gene expression was determined by real-time PCR. RESULTS The level of ACh was significantly higher in patients (mean ± SD: 5.95 ± 2.69) than in controls (1.12 ± 0.34) (P < 0.001). mAChR and nAChR expressions were significantly higher in patients compared with the controls (P < 0.001). A significant positive correlation was detected between the expression of nAChR in patients and the duration of psoriasis (r = 0.46, P = 0.01), and the body mass index of the patients correlated positively with both nAChR (r = 0.40, P = 0.027) and mAChR expression (r = 0.448, P = 0.013). CONCLUSION Abnormalities in the cutaneous extraneuronal cholinergic system could be involved in the pathogenesis of psoriasis. The high expression of nAChRs in patients with longer disease durations might represent an attempt by the body to regulate the inflammatory cascade in psoriatic lesions. The high expression of mAChR in psoriatic lesions may provide a link between psoriasis and obesity.
Collapse
Affiliation(s)
- O Abu Zeid
- Department of Dermatology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - A Abdel-Aziz
- Department of Dermatology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - L A Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Giza, Egypt
| | - E R Said
- Department of Dermatology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
The beneficial effects of a muscarinic agonist on pancreatic β-cells. Sci Rep 2019; 9:16180. [PMID: 31700039 PMCID: PMC6838462 DOI: 10.1038/s41598-019-52691-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
The brain and nervous system play an important role in pancreatic β-cell function. This study investigated the role of muscarinic agonists or acetylcholine, which is the major neurotransmitter in the vagal nerve, in regulating pancreatic β-cell mass and glucose homeostasis. Administration of the muscarinic agonist bethanechol increased insulin secretion and improved glucose tolerance in insulin-receptor substrate 2 (IRS2)-knockout (IRS-2−/−) mice and diet-induced obesity mice. Oral administration of bethanechol increased β-cell mass and proliferation in wild-type mice, but not IRS-2−/− mice. The muscarinic agonist also increased the incorporation of 5-bromo-2′-deoxyuridine (BrdU) into islets isolated from wild-type mice and pancreatic β-cell line MIN6. The phosphorylation of protein kinase B (Akt) induced by oral administration of bethanechol was observed in wild-type mice, but not IRS-2−/− mice. The secretion of glucagon-like peptide-1 (GLP-1) was also stimulated by bethanechol in wild-type mice, and a GLP-1 antagonist partially inhibited the bethanechol-induced increase in β-cell mass. These results suggest that the muscarinic agonist exerted direct and indirect effects on β-cell proliferation that were dependent on the IRS-2/Akt pathway. The bethanechol-stimulated release of GLP-1 may be indirectly associated with β-cell proliferation.
Collapse
|
16
|
Wang L, Pydi SP, Cui Y, Zhu L, Meister J, Gavrilova O, Berdeaux R, Fortin JP, Bence KK, Vernochet C, Wess J. Selective activation of G s signaling in adipocytes causes striking metabolic improvements in mice. Mol Metab 2019; 27:83-91. [PMID: 31272886 PMCID: PMC6717953 DOI: 10.1016/j.molmet.2019.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/30/2019] [Accepted: 06/16/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Given the worldwide epidemics of obesity and type 2 diabetes, novel antidiabetic and appetite-suppressing drugs are urgently needed. Adipocytes play a central role in the regulation of whole-body glucose and energy homeostasis. The goal of this study was to examine the metabolic effects of acute and chronic activation of Gs signaling selectively in adipocytes (activated Gs stimulates cAMP production), both in lean and obese mice. METHODS To address this question, we generated a novel mutant mouse strain (adipo-GsD mice) that expressed a Gs-coupled designer G protein-coupled receptor (Gs DREADD or short GsD) selectively in adipocytes. Importantly, the GsD receptor can only be activated by administration of an exogenous agent (CNO) that is otherwise pharmacologically inert. The adipo-GsD mice were maintained on either regular chow or a high-fat diet and then subjected to a comprehensive series of metabolic tests. RESULTS Pharmacological (CNO) activation of the GsD receptor in adipocytes of adipo-GsD mice caused profound improvements in glucose homeostasis and protected mice against the metabolic deficits associated with the consumption of a calorie-rich diet. Moreover, chronic activation of Gs signaling in adipocytes led to a striking increase in energy expenditure and reduced food intake, resulting in a decrease in body weight and fat mass when mice consumed a calorie-rich diet. CONCLUSION Systematic studies with a newly developed mouse model enabled us to assess the metabolic consequences caused by acute or chronic activation of Gs signaling selectively in adipocytes. Most strikingly, chronic activation of this pathway led to reduced body fat mass and restored normal glucose homeostasis in obese mice. These findings are of considerable relevance for the development of novel antidiabetic and anti-obesity drugs.
Collapse
Affiliation(s)
- Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology and Center for Metabolic and Degenerative Diseases at the Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth) and Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Jean-Philippe Fortin
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA
| | - Kendra K Bence
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA
| | - Cecile Vernochet
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA.
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Falk S, Lund C, Clemmensen C. Muscarinic receptors in energy homeostasis: Physiology and pharmacology. Basic Clin Pharmacol Toxicol 2019; 126 Suppl 6:66-76. [PMID: 31464050 DOI: 10.1111/bcpt.13311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/12/2019] [Indexed: 11/27/2022]
Abstract
Despite increased awareness and intensified biomedical research efforts, the prevalence of obesity continues to rise worldwide. This is alarming, because obesity accelerates the progression of several chronic disorders, including type 2 diabetes, cancer and cardiovascular disease. Individuals who experience significant weight loss must combat powerful counter-regulatory energy homeostatic processes, and, typically, most individuals regain the lost weight. Therefore, decoding the neural mechanisms underlying the regulation of energy homeostasis is necessary for developing breakthroughs in obesity management. It has been known for decades that cholinergic neurotransmission both directly and indirectly modulates energy homeostasis and metabolic health. Despite this insight, the molecular details underlying the modulation remain ill-defined, and the potential for targeting cholinergic muscarinic receptors for treating metabolic disease is largely uncharted. In this MiniReview, we scrutinize the literature that has formed our knowledge of muscarinic acetylcholine receptors (mAChRs) in energy homeostasis. The role of mAChRs in canonical appetite-regulating circuits will be discussed as will the more indirect regulation of energy homoeostasis via neurocircuits linked to motivated behaviours and emotional states. Finally, we discuss the therapeutic prospects of targeting mAChRs for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Sarah Falk
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Lund
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Alves-Lopes R, Neves KB, Touyz RM. Muscarinic Receptor Type-3 in Hypertension and Cholinergic-Adrenergic Crosstalk: Genetic Insights and Potential for New Antihypertensive Targets. Can J Cardiol 2019; 35:555-557. [PMID: 30954389 PMCID: PMC6499128 DOI: 10.1016/j.cjca.2019.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland.
| |
Collapse
|
19
|
Tiwari P, Saxena A, Gupta N, Medicherla KM, Suravajhala P, Mathur SK. Systems Genomics of Thigh Adipose Tissue From Asian Indian Type-2 Diabetics Revealed Distinct Protein Interaction Hubs. Front Genet 2019; 9:679. [PMID: 30671081 PMCID: PMC6331691 DOI: 10.3389/fgene.2018.00679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
We performed a systematic analysis of genes implicated in thigh subcutaneous adipose tissue of Asian Indian Type 2 Diabetes Mellitus (AIT2DM) and created a phenome-interactome network. This analysis was performed on 60 subjects specific to limb thigh fat by integrating phenotypic traits and similarity scores associated with AIT2DM. Using a phenotypic attribute, a contextual neighbor was identified across all the traits, viz. body mass index (BMI) statistics, adipocyte size, lipid parameters, homeostatic model assessment- insulin resistance (HOMA-IR), HOMA-ß. In this work, we have attempted to characterize transcription signatures using the phenome-interactome maps where each of the traits under study including the intermediary phenotypes has a distinct set of genes forming the hubs. Furthermore, we have identified various clinical, biochemical, and radiological parameters which show significant correlation with distinct hubs. We observed a number of novel pathways and genes including those that are non-coding RNAs implicated in AIT2DM.We showed that they appear to be associated with pathways, viz. tyrosine kinase JAK2, NOTCH thereby recruiting signaling molecules such as STAT5 and Src family kinases on the cell surface regulated them and our analyses comprising significant hubs suggest that thigh subcutaneous adipose tissue plays a role in pathophysiology of AIT2DM.
Collapse
Affiliation(s)
- Pradeep Tiwari
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India.,Department of Endocrinology, Sawai Man Singh Medical College and Hospital, Jaipur, India.,Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur, India
| | - Aditya Saxena
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, India
| | - Nidhi Gupta
- Department of Biotechnology, The IIS University, Jaipur, India
| | - Krishna Mohan Medicherla
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India
| | - Prashanth Suravajhala
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India
| | - Sandeep Kumar Mathur
- Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur, India
| |
Collapse
|
20
|
Chowański S, Pacholska-Bogalska J, Rosiński G. Cholinergic Agonists and Antagonists Have an Effect on the Metabolism of the Beetle Tenebrio Molitor. Molecules 2018; 24:E17. [PMID: 30577556 PMCID: PMC6337165 DOI: 10.3390/molecules24010017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/09/2018] [Accepted: 12/18/2018] [Indexed: 11/16/2022] Open
Abstract
Synthetic insecticides are still widely used in plant protection. The main target for their action is the nervous system, in which the cholinergic system plays a vital role. Currently available insecticides have low selectivity and act on the cholinergic systems of invertebrates and vertebrates. Acetylcholine, a cholinergic system neurotransmitter, acts on cells by two types of receptors: nicotinic and muscarinic. In mammals, the role of muscarinic acetylcholine receptors (mAChRs) is quite well-known but in insects, is still not enough. Based on data indicating that the muscarinic cholinergic system strongly affects mammalian metabolism, we investigated if it similarly occurs in insects. We investigated the influence of agonists (acetylcholine, carbachol, and pilocarpine) and antagonists (tropane alkaloids: atropine and scopolamine) of mAChRs on the level of selected metabolites in Tenebrio molitor beetle trophic tissues. We analyzed the glycogen content in the fat body and midgut, the total free sugar concentration in the hemolymph and the lipid amount in the fat body. Moreover, we analyzed the levels of insulin-like peptides in the hemolymph. The tested compounds significantly influenced the mentioned parameters. They increased the glycogen content in the fat body and midgut but decreased the concentration of free sugars in the hemolymph. The observed effects were tissue-specific, and were also time- and dose-dependent. We used nonligated and neck-ligated larvae (to eliminate the influence of head factors on tissue metabolism) to determine whether the observed changes are the result of direct or indirect impacts on tissues. The obtained data suggest that the cholinergic system affects the fat body and midgut indirectly and directly and a pleiotropic role for mAChRs exists in the regulation of energy metabolism in insects. Moreover, tested compounds significantly affected the level of insulin-like peptides in hemolymph. Our studies for the first time showed that mAChRs are involved in regulation of insect metabolism of trophic tissues, and act on them directly and indirectly. Improved knowledge about insect cholinergic system may help in searching more selective and environment-friendly solutions in pest management.
Collapse
Affiliation(s)
- Szymon Chowański
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, 61-712 Poznań, Poland.
| | - Joanna Pacholska-Bogalska
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, 61-712 Poznań, Poland.
| | - Grzegorz Rosiński
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, 61-712 Poznań, Poland.
| |
Collapse
|
21
|
Couselo-Seijas M, López-Canoa JN, Agra-Bermejo RM, Díaz-Rodriguez E, Fernandez AL, Martinez-Cereijo JM, Durán-Muñoz D, Bravo SB, Velo A, González-Melchor L, Fernández-López XA, Martínez-Sande JL, García-Seara J, González-Juanatey JR, Rodriguez-Mañero M, Eiras S. Cholinergic activity regulates the secretome of epicardial adipose tissue: Association with atrial fibrillation. J Cell Physiol 2018; 234:10512-10522. [PMID: 30480808 DOI: 10.1002/jcp.27723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/16/2018] [Indexed: 12/18/2022]
Abstract
Botulinum toxin injection on epicardial fat, which inhibits acetylcholine (ACh) release, reduced the presence of atrial fibrillation (AF) in patients after heart surgery. Thus, we wanted to study the profile of the released proteins of epicardial adipose tissue (EAT) under cholinergic activity (ACh treatment) and their value as AF predictors. Biopsies, explants, or primary cultures were obtained from the EAT of 85 patients that underwent open heart surgery. The quantification of muscarinic receptors (mAChR) by real-time polymerase chain reaction or western blot showed their expression in EAT. Moreover, mAChR Type 3 was upregulated after adipogenesis induction (p < 0.05). Cholinergic fibers in EAT were detected by vesicular ACh transporter levels and/or acetylcholinesterase activity. ACh treatment modified the released proteins by EAT, which were identified by nano-high-performance liquid chromatography and TripleTOF analysis. These differentially released proteins were involved in cell structure, inflammation, or detoxification. After testing the plasma levels of alpha-defensin 3 (inflammation-involved protein) of patients who underwent open heart surgery ( n = 24), we observed differential levels between the patients who developed or did not develop postsurgery AF (1.58 ± 1.61 ng/ml vs. 6.2 ± 5.6 ng/ml; p < 0.005). The cholinergic activity on EAT might suggest a new mechanism for studying the interplay among EAT, autonomic nervous system dysfunction, and AF.
Collapse
Affiliation(s)
- Marinela Couselo-Seijas
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose N López-Canoa
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Rosa M Agra-Bermejo
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| | - Esther Díaz-Rodriguez
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Angel L Fernandez
- CIBERCV, Madrid, Spain.,Heart Surgery Department, University Clinical Hospital of Santiago dtye Compostela, Santiago de Compostela, Spain
| | - Jose M Martinez-Cereijo
- Heart Surgery Department, University Clinical Hospital of Santiago dtye Compostela, Santiago de Compostela, Spain
| | - Dario Durán-Muñoz
- CIBERCV, Madrid, Spain.,Heart Surgery Department, University Clinical Hospital of Santiago dtye Compostela, Santiago de Compostela, Spain
| | - Susana B Bravo
- Proteomic Unit of Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alba Velo
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Laila González-Melchor
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Xesus A Fernández-López
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose L Martínez-Sande
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| | - Javier García-Seara
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| | - Jose R González-Juanatey
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| | - Moises Rodriguez-Mañero
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| | - Sonia Eiras
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
22
|
Abstract
G protein-coupled receptors (GPCRs) constitute a large family of receptors that activate intracellular signaling pathways upon detecting specific extracellular ligands. While many aspects of GPCR signaling have been uncovered through decades of studies, some fundamental properties, like its channel capacity—a measure of how much information a given transmission system can reliably transduce—are still debated. Previous studies concluded that GPCRs in individual cells could transmit around one bit of information about the concentration of the ligands, allowing only for a reliable on or off response. Using muscarinic receptor-induced calcium response measured in individual cells upon repeated stimulation, we show that GPCR signaling systems possess a significantly higher capacity. We estimate the channel capacity of this system to be above two, implying that at least four concentration levels of the agonist can be distinguished reliably. These findings shed light on the basic principles of GPCR signaling. G protein-coupled receptors (GPCRs) activate intracellular signalling pathways upon extracellular stimulation. Here authors record single cell responses of GPCR signalling which allows the direct estimation of its channel capacity for each cell along with the reproducibility of its response.
Collapse
|
23
|
Cholinergic signaling mediates the effects of xenin-25 on secretion of pancreatic polypeptide but not insulin or glucagon in humans with impaired glucose tolerance. PLoS One 2018; 13:e0192441. [PMID: 29466430 PMCID: PMC5821323 DOI: 10.1371/journal.pone.0192441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 01/10/2018] [Indexed: 01/14/2023] Open
Abstract
We previously demonstrated that infusion of an intestinal peptide called xenin-25 (Xen) amplifies the effects of glucose-dependent insulinotropic polypeptide (GIP) on insulin secretion rates (ISRs) and plasma glucagon levels in humans. However, these effects of Xen, but not GIP, were blunted in humans with type 2 diabetes. Thus, Xen rather than GIP signaling to islets fails early during development of type 2 diabetes. The current crossover study determines if cholinergic signaling relays the effects of Xen on insulin and glucagon release in humans as in mice. Fasted subjects with impaired glucose tolerance were studied. On eight separate occasions, each person underwent a single graded glucose infusion- two each with infusion of albumin, Xen, GIP, and GIP plus Xen. Each infusate was administered ± atropine. Heart rate and plasma glucose, insulin, C-peptide, glucagon, and pancreatic polypeptide (PP) levels were measured. ISRs were calculated from C-peptide levels. All peptides profoundly increased PP responses. From 0 to 40 min, peptide(s) infusions had little effect on plasma glucose concentrations. However, GIP, but not Xen, rapidly and transiently increased ISRs and glucagon levels. Both responses were further amplified when Xen was co-administered with GIP. From 40 to 240 min, glucose levels and ISRs continually increased while glucagon concentrations declined, regardless of infusate. Atropine increased resting heart rate and blocked all PP responses but did not affect ISRs or plasma glucagon levels during any of the peptide infusions. Thus, cholinergic signaling mediates the effects of Xen on insulin and glucagon release in mice but not humans.
Collapse
|
24
|
Farhangi MA, Mesgari-Abbasi M, Nameni G, Hajiluian G, Shahabi P. The effects of vitamin D administration on brain inflammatory markers in high fat diet induced obese rats. BMC Neurosci 2017; 18:81. [PMID: 29281967 PMCID: PMC5745807 DOI: 10.1186/s12868-017-0400-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/19/2017] [Indexed: 11/10/2022] Open
Abstract
Background Obesity induced brain inflammation is associated with cognitive disorders. We aimed to investigate the influence of vitamin D on hypothalamus and hippocampus inflammatory response in high-fat diet induced obese rats. Methods In the beginning of the study, 40 rats were divided into two groups: control diet and high fat diet (HFD) for 16 weeks; then each group subdivided into two groups including: N, ND + vitamin D, HFD and HFD + vitamin D. Vitamin D supplementation was done for 5 weeks at 500 IU/kg dosage. IL-6, IL-1β, NF-Kβ and acetylcholine (ACH) and brain derived neurotropic factor (BDNF) concentrations in hippocampus and hypothalamus homogenate samples were measured by commercial ELISA kits. Results Vitamin D administration, reduced food intake and weight gain in studied groups (P < 0.001). Vitamin D reduced hippocampus acetylcholine concentrations in ND + vitamin D group (P < 0.001). High fat diet increased hippocampus IL-6 concentrations significantly (P < 0.05) compared with normal diet receiving groups. Vitamin D could not have significant effects on IL-6 concentrations. Vitamin D administrations reduced IL-1β, NF-Kβ and acetylcholine concentration and BDNF concentrations in ND + vitamin D compared with ND group. These reductions were not significant in HFD + vitamin D versus HFD group. Conclusion According to our results, vitamin D reduced food intake and weight gain and modulated the HFD induced inflammatory response in hippocampus and hypothalamus of high fat diet induced obesity. Therefore, this neurosteroid, can be suggested as a supplemental therapeutic tool in prevention of obesity related cognitive and neurodegenerative problems.
Collapse
Affiliation(s)
- Mahdieh Abbasalizad Farhangi
- Drug Applied Research Center, Nutrition Research Center, Department of Community Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri Street, Tabriz, Iran.
| | - Mehran Mesgari-Abbasi
- Drug Applied Research Center, Nutrition Research Center, Department of Community Nutrition, Tabriz University of Medical Sciences, Attar Neyshabouri Street, Tabriz, Iran
| | - Ghazaleh Nameni
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghazaleh Hajiluian
- Nutrition Research Center, Department of Community Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Wang Q, Pronin AN, Levay K, Almaca J, Fornoni A, Caicedo A, Slepak VZ. Regulator of G-protein signaling Gβ5-R7 is a crucial activator of muscarinic M3 receptor-stimulated insulin secretion. FASEB J 2017; 31:4734-4744. [PMID: 28687610 DOI: 10.1096/fj.201700197rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 12/20/2022]
Abstract
In pancreatic β cells, muscarinic cholinergic receptor M3 (M3R) stimulates glucose-induced secretion of insulin. Regulator of G-protein signaling (RGS) proteins are critical modulators of GPCR activity, yet their role in β cells remains largely unknown. R7 subfamily RGS proteins are stabilized by the G-protein subunit Gβ5, such that the knockout of the Gnb5 gene results in degradation of all R7 subunits. We found that Gnb5 knockout in mice or in the insulin-secreting MIN6 cell line almost completely eliminates insulinotropic activity of M3R. Moreover, overexpression of Gβ5-RGS7 strongly promotes M3R-stimulated insulin secretion. Examination of this noncanonical mechanism in Gnb5-/- MIN6 cells showed that cAMP, diacylglycerol, or Ca2+ levels were not significantly affected. There was no reduction in the amplitude of free Ca2+ responses in islets from the Gnb5-/- mice, but the frequency of Ca2+ oscillations induced by cholinergic agonist was lowered by more than 30%. Ablation of Gnb5 impaired M3R-stimulated phosphorylation of ERK1/2. Stimulation of the ERK pathway in Gnb5-/- cells by epidermal growth factor restored M3R-stimulated insulin release to near normal levels. Identification of the novel role of Gβ5-R7 in insulin secretion may lead to a new therapeutic approach for improving pancreatic β-cell function.-Wang, Q., Pronin, A. N., Levay, K., Almaca, J., Fornoni, A., Caicedo, A., Slepak, V. Z. Regulator of G-protein signaling Gβ5-R7 is a crucial activator of muscarinic M3 receptor-stimulated insulin secretion.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; and
| | - Alexey N Pronin
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; and
| | - Konstantin Levay
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; and
| | - Joana Almaca
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Alessia Fornoni
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Alejandro Caicedo
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; and
| |
Collapse
|
26
|
Olsen MK, Johannessen H, Cassie N, Barrett P, Takeuchi K, Kulseng B, Chen D, Zhao CM. Steady-state energy balance in animal models of obesity and weight loss. Scand J Gastroenterol 2017; 52:442-449. [PMID: 27996337 DOI: 10.1080/00365521.2016.1267791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE We wanted to exam the steady-state energy balance by using high-fat diet-induced obese (DIO) rats and mice as models for positive energy balance, and gastric bypassed (GB) rats and gene knockout of muscarinic acetylcholine M3 receptor (M3KO) mice as models for negative energy balance. METHODS One hundred and thirty-two rats and mice were used. Energy balance was measured by a comprehensive laboratory animal monitoring system. Gene expression was analysed by in situ hybridisation in M3KO mice. RESULTS DIO rats reached the plateau of body weight 28 weeks after starting high-fat diet (25% heavier than controls), whereas DIO mice reached the plateau after 6 weeks (23% heavier than controls). At the plateau, DIO rats had higher calorie intake during the light phase but not during the dark phase, while mice had the same calorie intake per day as controls. DIO rats and mice had lower energy expenditure (EE) and respiratory exchange ratio (RER) than controls. GB-rats reached the plateau (15% weight loss) 2 weeks after surgery and had the same calorie intake as sham-operated controls. EE, but not RER, was higher in GB rats than controls during the dark phase. The lean M3KO mice (25% lighter than wild-type (WT) mice at the plateau between 6 and 15 months of age) had the same calorie intake but higher EE, RER and hypothalamic mRNA expression of NPY, AgRP and leptin receptor than WT mice. CONCLUSION When body weight gain or loss reached a plateau, the steady-state energy balance was mainly maintained by EE and/or RER rather than calorie intake.
Collapse
Affiliation(s)
- Magnus Kringstad Olsen
- a Department of Cancer Research and Molecular Medicine , Norwegian University of Science and Technology , Trondheim , Norway
| | - Helene Johannessen
- a Department of Cancer Research and Molecular Medicine , Norwegian University of Science and Technology , Trondheim , Norway
| | - Nikki Cassie
- b The Rowett Institute, University of Aberdeen , Aberdeen , Scotland
| | - Perry Barrett
- b The Rowett Institute, University of Aberdeen , Aberdeen , Scotland
| | - Koji Takeuchi
- c Department of Pharmacology and Experimental Therapeutics , Kyoto Pharmaceutical University , Kyoto , Japan
| | - Bård Kulseng
- a Department of Cancer Research and Molecular Medicine , Norwegian University of Science and Technology , Trondheim , Norway.,d Department of Surgery , Centre for Obesity Research and Innovation, St. Olav's University Hospital , Trondheim , Norway
| | - Duan Chen
- a Department of Cancer Research and Molecular Medicine , Norwegian University of Science and Technology , Trondheim , Norway.,d Department of Surgery , Centre for Obesity Research and Innovation, St. Olav's University Hospital , Trondheim , Norway
| | - Chun-Mei Zhao
- a Department of Cancer Research and Molecular Medicine , Norwegian University of Science and Technology , Trondheim , Norway
| |
Collapse
|
27
|
Jeong JH, Lee DK, Jo YH. Cholinergic neurons in the dorsomedial hypothalamus regulate food intake. Mol Metab 2017; 6:306-312. [PMID: 28271037 PMCID: PMC5323886 DOI: 10.1016/j.molmet.2017.01.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 01/13/2023] Open
Abstract
Objective Central cholinergic neural circuits play a role in the regulation of feeding behavior. The dorsomedial hypothalamus (DMH) is considered the appetite-stimulating center and contains cholinergic neurons. Here, we study the role of DMH cholinergic neurons in the control of food intake. Methods To selectively stimulate DMH cholinergic neurons, we expressed stimulatory designer receptors exclusively activated by designer drugs (DREADDs) and channelrhodopsins in DMH cholinergic neurons by injection of adeno-associated virus (AAV) vectors into the DMH of choline acetyltransferase (ChAT)-IRES-Cre mice. We also generated transgenic mice expressing channelrhodopsins in cholinergic neurons with the Cre-LoxP technique. To delete the Chat gene exclusively in the DMH, we injected an AAV carrying a Cre recombinase transgene into the DMH of floxed ChAT mice. Food intake was measured with and without selective stimulation of DMH cholinergic neurons. Results Mice lacking the Chat gene in the DMH show reduced body weight as compared to control. Chemogenetic activation of DMH cholinergic neurons promotes food intake. This orexigenic effect is further supported by experiments of optogenetic stimulation of DMH cholinergic neurons. DMH cholinergic neurons innervate pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus (ARC). Treatment with acetylcholine (ACh) enhances GABAergic inhibitory transmission to ARC POMC neurons that is blocked by the muscarinic receptor antagonist. Direct activation of cholinergic fibers in the ARC readily stimulates food intake that is also abolished by the muscarinic receptor antagonist. Conclusion ACh released from DMH cholinergic neurons regulates food intake and body weight. This effect is mediated in part through regulation of ARC POMC neurons. Activation of muscarinic receptors on GABAergic axon terminals enhances inhibitory tone to ARC POMC neurons. Hence, this novel DMHACh → ARCPOMC pathway plays an important role in the control of food intake and body weight. DMH cholinergic neurons innervate ARC POMC neurons. Activation of muscarinic receptors enhances inhibitory tone to ARC POMC neurons. Stimulation of DMH cholinergic neurons promotes food intake. This orexigenic effect is abolished by the muscarinic receptor antagonist. Mice lacking the Chat gene in the DMH show reduced body weight.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, NY, USA
| | - Dong Kun Lee
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, NY, USA; Department of Physiology, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Young-Hwan Jo
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, NY, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
28
|
Bradley SJ, Tobin AB. Design of Next-Generation G Protein-Coupled Receptor Drugs: Linking Novel Pharmacology and In Vivo Animal Models. Annu Rev Pharmacol Toxicol 2016; 56:535-59. [PMID: 26738479 DOI: 10.1146/annurev-pharmtox-011613-140012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the fact that G protein-coupled receptors (GPCRs) are the most successful drug targets in history, this supergene family of cell surface receptors has yet to be fully exploited as targets in the treatment of human disease. Here, we present optimism that this may change in the future by reviewing the substantial progress made in the understanding of GPCR molecular pharmacology that has generated an extensive toolbox of ligand types that include orthosteric, allosteric, and bitopic ligands, many of which show signaling bias. We discuss how combining these advances with recently described transgenic, chemical genetic, and optogenetic animal models will provide the framework to allow for the rational design of next-generation GPCR drugs that possess increased therapeutic efficacy and decreased adverse/toxic responses.
Collapse
Affiliation(s)
- Sophie J Bradley
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| | - Andrew B Tobin
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| |
Collapse
|
29
|
Jeong JH, Woo YJ, Chua S, Jo YH. Single-Cell Gene Expression Analysis of Cholinergic Neurons in the Arcuate Nucleus of the Hypothalamus. PLoS One 2016; 11:e0162839. [PMID: 27611685 PMCID: PMC5017726 DOI: 10.1371/journal.pone.0162839] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/29/2016] [Indexed: 01/18/2023] Open
Abstract
The cholinoceptive system in the hypothalamus, in particular in the arcuate nucleus (ARC), plays a role in regulating food intake. Neurons in the ARC contain multiple neuropeptides, amines, and neurotransmitters. To study molecular and neurochemical heterogeneity of ARC neurons, we combine single-cell qRT-PCR and single-cell whole transcriptome amplification methods to analyze expression patterns of our hand-picked 60 genes in individual neurons in the ARC. Immunohistochemical and single-cell qRT-PCR analyses show choline acetyltransferase (ChAT)-expressing neurons in the ARC. Gene expression patterns are remarkably distinct in each individual cholinergic neuron. Two-thirds of cholinergic neurons express tyrosine hydroxylase (Th) mRNA. A large subset of these Th-positive cholinergic neurons is GABAergic as they express the GABA synthesizing enzyme glutamate decarboxylase and vesicular GABA transporter transcripts. Some cholinergic neurons also express the vesicular glutamate transporter transcript gene. POMC and POMC-processing enzyme transcripts are found in a subpopulation of cholinergic neurons. Despite this heterogeneity, gene expression patterns in individual cholinergic cells appear to be highly regulated in a cell-specific manner. In fact, membrane receptor transcripts are clustered with their respective intracellular signaling and downstream targets. This novel population of cholinergic neurons may be part of the neural circuitries that detect homeostatic need for food and control the drive to eat.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young Jae Woo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
| | - Streamson Chua
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young-Hwan Jo
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, United States of America
- * E-mail:
| |
Collapse
|
30
|
Chowdhury S, Wang S, Dunai J, Kilpatrick R, Oestricker LZ, Wallendorf MJ, Patterson BW, Reeds DN, Wice BM. Hormonal Responses to Cholinergic Input Are Different in Humans with and without Type 2 Diabetes Mellitus. PLoS One 2016; 11:e0156852. [PMID: 27304975 PMCID: PMC4909255 DOI: 10.1371/journal.pone.0156852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
Abstract
Peripheral muscarinic acetylcholine receptors regulate insulin and glucagon release in rodents but their importance for similar roles in humans is unclear. Bethanechol, an acetylcholine analogue that does not cross the blood-brain barrier, was used to examine the role of peripheral muscarinic signaling on glucose homeostasis in humans with normal glucose tolerance (NGT; n = 10), impaired glucose tolerance (IGT; n = 11), and type 2 diabetes mellitus (T2DM; n = 9). Subjects received four liquid meal tolerance tests, each with a different dose of oral bethanechol (0, 50, 100, or 150 mg) given 60 min before a meal containing acetaminophen. Plasma pancreatic polypeptide (PP), glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), glucose, glucagon, C-peptide, and acetaminophen concentrations were measured. Insulin secretion rates (ISRs) were calculated from C-peptide levels. Acetaminophen and PP concentrations were surrogate markers for gastric emptying and cholinergic input to islets. The 150 mg dose of bethanechol increased the PP response 2-fold only in the IGT group, amplified GLP-1 release in the IGT and T2DM groups, and augmented the GIP response only in the NGT group. However, bethanechol did not alter ISRs or plasma glucose, glucagon, or acetaminophen concentrations in any group. Prior studies showed infusion of xenin-25, an intestinal peptide, delays gastric emptying and reduces GLP-1 release but not ISRs when normalized to plasma glucose levels. Analysis of archived plasma samples from this study showed xenin-25 amplified postprandial PP responses ~4-fold in subjects with NGT, IGT, and T2DM. Thus, increasing postprandial cholinergic input to islets augments insulin secretion in mice but not humans. Trial Registration: ClinicalTrials.gov NCT01434901
Collapse
Affiliation(s)
- Sara Chowdhury
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Songyan Wang
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Judit Dunai
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Rachel Kilpatrick
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Lauren Z. Oestricker
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Michael J. Wallendorf
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Bruce W. Patterson
- Department of Internal Medicine, Division of Nutritional Science, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Dominic N. Reeds
- Department of Internal Medicine, Division of Nutritional Science, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Burton M. Wice
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
31
|
Regulation of arcuate genes by developmental exposures to endocrine-disrupting compounds in female rats. Reprod Toxicol 2016; 62:18-26. [PMID: 27103539 DOI: 10.1016/j.reprotox.2016.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/11/2016] [Accepted: 04/15/2016] [Indexed: 01/09/2023]
Abstract
Developmental exposure to endocrine-disrupting compounds (EDCs) alters reproduction and energy homeostasis, both of which are regulated by the arcuate nucleus (ARC). Little is known about the effects of EDC on ARC gene expression. In Experiment #1, pregnant dams were treated with either two doses of bisphenol A (BPA) or oil from embryonic day (E)18-21. Neonates were injected from postnatal day (PND)0-7. Vaginal opening, body weights, and ARC gene expression were measured. Chrm3 (muscarinic receptor 3) and Adipor1 (adiponectin receptor 1) were decreased by BPA. Bdnf (brain-derived neurotropic factor), Igf1 (insulin-like growth factor 1), Htr2c (5-hydroxytryptamine receptor), and Cck2r (cholescystokinin 2 receptor) were impacted. In Experiment #2, females were exposed to BPA, diethylstilbestrol (DES), di(2-ethylhexyl)phthalate, or methoxychlor (MXC) during E11-PND7. MXC and DES advanced the age of vaginal opening and ARC gene expression was impacted. These data indicate that EDCs alter ARC genes involved in reproduction and energy homeostasis in females.
Collapse
|
32
|
Beckmann J, Dittmann N, Schütz I, Klein J, Lips KS. Effect of M3 muscarinic acetylcholine receptor deficiency on collagen antibody-induced arthritis. Arthritis Res Ther 2016; 18:17. [PMID: 26785775 PMCID: PMC4719200 DOI: 10.1186/s13075-016-0926-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 01/07/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND There is increasing evidence that the non-neuronal cholinergic system might be of importance for the pathology of rheumatoid arthritis. The role of M3 muscarinic acetylcholine receptor (M3R) in this regard has, however, not been investigated to date. Thus, in the present study we analyzed if M3R deficiency might have a protective effect on experimentally induced arthritis. METHODS Collagen antibody-induced arthritis (CAIA) was evoked in M3R-deficient (M3R(-/-)) mice and wild-type (WT) littermates. Severity of arthritis was assessed by scoring of paw swelling. The joints of arthritic and nonarthritic animals were analyzed for histopathological changes regarding synovial tissue, cartilage degradation and bone destruction. Further, gene expression analysis of respective markers was performed. Systemic and local inflammatory response was determined by flow cytometry and immunohistochemistry for leukocytes as well as mRNA and protein measurements for pro-inflammatory cytokines and chemokines. RESULTS In arthritic M3R(-/-) mice the number of leukocytes, specifically neutrophils, was enhanced even though clinical arthritis score was not significantly different between WT and M3R(-/-) mice with CAIA. In M3R(-/-) mice, levels of neutrophil chemoattractant chemokine C-X-C-motif ligand 2 (CXCL2) as well as the pro-inflammatory cytokine interleukin-6 were already strongly increased in mice with low arthritis score, whereas WT mice only showed prominent expression of these markers when reaching high arthritis scores. Furthermore, arthritic M3R(-/-) mice displayed a stronger degradation of collagen II in the articular cartilage and, most strikingly, histopathological evaluation revealed more severe bone destruction in arthritic mice with M3R deficiency compared to WT littermates. Moreover, in M3R(-/-) mice, gene expression of markers for bone degradation (matrix metalloproteinase 13, cathepsin K and receptor activator of nuclear factor-κB ligand) was already increased in mice with low arthritis score. CONCLUSIONS Taken together, the present study shows that while M3R(-/-) mice were not protected from CAIA, they had a tendency toward a higher inflammatory response after arthritis induction than WT mice. Further, arthritis-induced joint destruction was significantly stronger in mice with M3R deficiency, indicating that stimulation of M3R might have protective effects on arthritis.
Collapse
Affiliation(s)
- Janet Beckmann
- Laboratory of Experimental Trauma Surgery, Justus-Liebig University Giessen, Kerkraderstrasse 9, 35394, Giessen, Germany.
| | - Nicole Dittmann
- Laboratory of Experimental Trauma Surgery, Justus-Liebig University Giessen, Kerkraderstrasse 9, 35394, Giessen, Germany.
| | - Iris Schütz
- Laboratory of Experimental Trauma Surgery, Justus-Liebig University Giessen, Kerkraderstrasse 9, 35394, Giessen, Germany.
| | - Jochen Klein
- Department of Pharmacology, School of Pharmacy, Goethe-University Frankfurt, Max-von-Laue Strasse 9, 60438, Frankfurt am Main, Germany.
| | - Katrin Susanne Lips
- Laboratory of Experimental Trauma Surgery, Justus-Liebig University Giessen, Kerkraderstrasse 9, 35394, Giessen, Germany.
| |
Collapse
|
33
|
Gsα deficiency in adipose tissue improves glucose metabolism and insulin sensitivity without an effect on body weight. Proc Natl Acad Sci U S A 2015; 113:446-51. [PMID: 26712027 DOI: 10.1073/pnas.1517142113] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Gsα, the G protein that transduces receptor-stimulated cAMP generation, mediates sympathetic nervous system stimulation of brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT), which are both potential targets for treating obesity, as well as lipolysis. We generated a mouse line with Gsα deficiency in mature BAT and WAT adipocytes (Ad-GsKO). Ad-GsKO mice had impaired BAT function, absent browning of WAT, and reduced lipolysis, and were therefore cold-intolerant. Despite the presence of these abnormalities, Ad-GsKO mice maintained normal energy balance on both standard and high-fat diets, associated with decreases in both lipolysis and lipid synthesis. In addition, Ad-GsKO mice maintained at thermoneutrality on a standard diet also had normal energy balance. Ad-GsKO mice had improved insulin sensitivity and glucose metabolism, possibly secondary to the effects of reduced lipolysis and lower circulating fatty acid binding protein 4 levels. Gsα signaling in adipose tissues may therefore affect whole-body glucose metabolism in the absence of an effect on body weight.
Collapse
|
34
|
Jeong JH, Lee DK, Blouet C, Ruiz HH, Buettner C, Chua S, Schwartz GJ, Jo YH. Cholinergic neurons in the dorsomedial hypothalamus regulate mouse brown adipose tissue metabolism. Mol Metab 2015; 4:483-92. [PMID: 26042202 PMCID: PMC4443291 DOI: 10.1016/j.molmet.2015.03.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 01/01/2023] Open
Abstract
Objective Brown adipose tissue (BAT) thermogenesis is critical in maintaining body temperature. The dorsomedial hypothalamus (DMH) integrates cutaneous thermosensory signals and regulates adaptive thermogenesis. Here, we study the function and synaptic connectivity of input from DMH cholinergic neurons to sympathetic premotor neurons in the raphe pallidus (Rpa). Methods In order to selectively manipulate DMH cholinergic neuron activity, we generated transgenic mice expressing channelrhodopsin fused to yellow fluorescent protein (YFP) in cholinergic neurons (choline acetyltransferase (ChAT)-Cre::ChR2-YFP) with the Cre-LoxP technique. In addition, we used an adeno-associated virus carrying the Cre recombinase gene to delete the floxed Chat gene in the DMH. Physiological studies in response to optogenetic stimulation of DMH cholinergic neurons were combined with gene expression and immunocytochemical analyses. Results A subset of DMH neurons are ChAT-immunopositive neurons. The activity of these neurons is elevated by warm ambient temperature. A phenotype-specific neuronal tracing shows that DMH cholinergic neurons directly project to serotonergic neurons in the Rpa. Optical stimulation of DMH cholinergic neurons decreases BAT activity, which is associated with reduced body core temperature. Furthermore, elevated DMH cholinergic neuron activity decreases the expression of BAT uncoupling protein 1 (Ucp1) and peroxisome proliferator-activated receptor γ coactivator 1 α (Pgc1α) mRNAs, markers of BAT activity. Injection of M2-selective muscarinic receptor antagonists into the 4th ventricle abolishes the effect of optical stimulation. Single cell qRT-PCR analysis of retrogradely identified BAT-projecting neurons in the Rpa shows that all M2 receptor-expressing neurons contain tryptophan hydroxylase 2. In animals lacking the Chat gene in the DMH, exposure to warm temperature reduces neither BAT Ucp1 nor Pgc1α mRNA expression. Conclusion DMH cholinergic neurons directly send efferent signals to sympathetic premotor neurons in the Rpa. Elevated cholinergic input to this area reduces BAT activity through activation of M2 mAChRs on serotonergic neurons. Therefore, the direct DMHACh–Rpa5-HT pathway may mediate physiological heat-defense responses to elevated environmental temperature.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Dong Kun Lee
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Clemence Blouet
- Medical Research Council Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Henry H Ruiz
- Diabetes, Obesity & Metabolism Institute and Department of Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA ; Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Christoph Buettner
- Diabetes, Obesity & Metabolism Institute and Department of Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA ; Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Streamson Chua
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Gary J Schwartz
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Young-Hwan Jo
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA ; Department of Molecular Pharmacology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
35
|
Olianas MC, Dedoni S, Onali P. Involvement of store-operated Ca(2+) entry in activation of AMP-activated protein kinase and stimulation of glucose uptake by M3 muscarinic acetylcholine receptors in human neuroblastoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:3004-17. [PMID: 25242372 DOI: 10.1016/j.bbamcr.2014.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 01/08/2023]
Abstract
Gq/11-coupled muscarinic acetylcholine receptors (mAChRs) belonging to M1, M3 and M5 subtypes have been shown to activate the metabolic sensor AMP-activated protein kinase (AMPK) through Ca(2+)/calmodulin-dependent protein kinase kinase-β (CaMKKβ)-mediated phosphorylation at Thr172. However, the source of Ca(2+) required for this response has not been yet elucidated. Here, we investigated the involvement of store-operated Ca(2+) entry (SOCE) in AMPK activation by pharmacologically defined M3 mAChRs in human SH-SY5Y neuroblastoma cells. In Ca(2+)-free medium the cholinergic agonist carbachol (CCh) caused a transient increase of phospho-Thr172 AMPK that rapidly ceased within 2min. Conversely, in the presence of extracellular Ca(2+) CCh-induced AMPK phosphorylation lasted for at least 180min. The SOCE modulator 2-aminoethoxydiphephenyl borate (2-APB), at a concentration (50μM) that suppressed CCh-induced intracellular Ca(2+) ([Ca(2+)]i) plateau, inhibited CCh-induced AMPK phosphorylation. CCh triggered the activation of the endoplasmic reticulum Ca(2+) sensor stromal interaction molecule (STIM) 1, as indicated by redistribution of STIM1 immunofluorescence into puncta, and promoted the association of STIM1 with the SOCE channel component Orai1. Cell depletion of STIM1 by siRNA treatment reduced both CCh-induced [Ca(2+)]i plateau and AMPK activation. M3 mAChRs increased glucose uptake and this response required extracellular Ca(2+) and was inhibited by 2-APB, STIM1 knockdown, CaMKKβ and AMPK inhibitors, and adenovirus infection with dominant negative AMPK. Thus, the study provides evidence that SOCE is required for sustained activation of AMPK and stimulation of downstream glucose uptake by M3 mAChRs and suggests that SOCE is a critical process connecting M3 mAChRs to the control of neuronal energy metabolism.
Collapse
Affiliation(s)
- Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
36
|
Tao YX, Liang XF. G Protein-Coupled Receptors as Regulators of Glucose Homeostasis and Therapeutic Targets for Diabetes Mellitus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:1-21. [DOI: 10.1016/b978-0-12-800101-1.00001-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Abstract
Second generation antipsychotics (SGAs) are widely prescribed to treat various disorders, most notably schizophrenia and bipolar disorder; however, SGAs can cause abnormal glucose metabolism that can lead to insulin-resistance and type 2 diabetes mellitus side-effects by largely unknown mechanisms. This review explores the potential candidature of the acetylcholine (ACh) muscarinic M3 receptor (M3R) as a prime mechanistic and possible therapeutic target of interest in SGA-induced insulin dysregulation. Studies have identified that SGA binding affinity to the M3R is a predictor of diabetes risk; indeed, olanzapine and clozapine, SGAs with the highest clinical incidence of diabetes side-effects, are potent M3R antagonists. Pancreatic M3Rs regulate the glucose-stimulated cholinergic pathway of insulin secretion; their activation on β-cells stimulates insulin secretion, while M3R blockade decreases insulin secretion. Genetic modification of M3Rs causes robust alterations in insulin levels and glucose tolerance in mice. Olanzapine alters M3R density in discrete nuclei of the hypothalamus and caudal brainstem, regions that regulate glucose homeostasis and insulin secretion through vagal innervation of the pancreas. Furthermore, studies have demonstrated a dynamic sensitivity of hypothalamic and brainstem M3Rs to altered glucometabolic status of the body. Therefore, the M3R is in a prime position to influence glucose homeostasis through direct effects on pancreatic β-cells and by potentially altering signalling in the hypothalamus and brainstem. SGA-induced insulin dysregulation may be partly due to blockade of central and peripheral M3Rs, causing an initial disruption to insulin secretion and glucose homeostasis that can progressively lead to insulin resistance and diabetes during chronic treatment.
Collapse
|
38
|
Neuropeptide y gates a stress-induced, long-lasting plasticity in the sympathetic nervous system. J Neurosci 2013; 33:12705-17. [PMID: 23904607 DOI: 10.1523/jneurosci.3132-12.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acute stress evokes the fight-or-flight reflex, which via release of the catecholamine hormones affects the function of every major organ. Although the reflex is transient, it has lasting consequences that produce an exaggerated response when stress is reexperienced. How this change is encoded is not known. We investigated whether the reflex affects the adrenal component of the sympathetic nervous system, a major branch of the stress response. Mice were briefly exposed to the cold-water forced swim test (FST) which evoked an increase in circulating catecholamines. Although this hormonal response was transient, the FST led to a long-lasting increase in the catecholamine secretory capacity measured amperometrically from chromaffin cells and in the expression of tyrosine hydroxylase. A variety of approaches indicate that these changes are regulated postsynaptically by neuropeptide Y (NPY), an adrenal cotransmitter. Using immunohistochemistry, RT-PCR, and NPY(GFP) BAC mice, we find that NPY is synthesized by all chromaffin cells. Stress failed to increase secretory capacity in NPY knock-out mice. Genetic or pharmacological interference with NPY and Y1 (but not Y2 or Y5) receptor signaling attenuated the stress-induced change in tyrosine hydroxylase expression. These results indicate that, under basal conditions, adrenal signaling is tonically inhibited by NPY, but stress overrides this autocrine negative feedback loop. Because acute stress leads to a lasting increase in secretory capacity in vivo but does not alter sympathetic tone, these postsynaptic changes appear to be an adaptive response. We conclude that the sympathetic limb of the stress response exhibits an activity-dependent form of long-lasting plasticity.
Collapse
|
39
|
Kruse AC, Li J, Hu J, Kobilka BK, Wess J. Novel insights into M3 muscarinic acetylcholine receptor physiology and structure. J Mol Neurosci 2013; 53:316-23. [PMID: 24068573 DOI: 10.1007/s12031-013-0127-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/16/2013] [Indexed: 01/01/2023]
Abstract
Recent studies with M3 muscarinic acetylcholine receptor (M3R) mutant mice suggest that drugs selectively targeting this receptor subtype may prove useful for the treatment of various pathophysiological conditions. Moreover, the use of M3R-based designer G protein-coupled receptors (GPCRs) has provided novel insights into how Gq-coupled GPCRs can modulate whole-body glucose homeostasis by acting on specific peripheral cell types. More recently, we succeeded in using X-ray crystallography to determine the structure of the M3R bound to the bronchodilating drug tiotropium, a muscarinic antagonist (inverse agonist). This new structural information should facilitate the development of orthosteric or allosteric M3R-selective drugs that are predicted to have considerable therapeutic potential.
Collapse
Affiliation(s)
- Andrew C Kruse
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | |
Collapse
|
40
|
Wu JJ, Liu J, Chen EB, Wang JJ, Cao L, Narayan N, Fergusson MM, Rovira II, Allen M, Springer DA, Lago CU, Zhang S, DuBois W, Ward T, deCabo R, Gavrilova O, Mock B, Finkel T. Increased mammalian lifespan and a segmental and tissue-specific slowing of aging after genetic reduction of mTOR expression. Cell Rep 2013; 4:913-20. [PMID: 23994476 PMCID: PMC3784301 DOI: 10.1016/j.celrep.2013.07.030] [Citation(s) in RCA: 234] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 07/11/2013] [Accepted: 07/24/2013] [Indexed: 12/31/2022] Open
Abstract
We analyzed aging parameters using a mechanistic target of rapamycin (mTOR) hypomorphic mouse model. Mice with two hypomorphic (mTOR(Δ/Δ)) alleles are viable but express mTOR at approximately 25% of wild-type levels. These animals demonstrate reduced mTORC1 and mTORC2 activity and exhibit an approximately 20% increase in median survival. While mTOR(Δ/Δ) mice are smaller than wild-type mice, these animals do not demonstrate any alterations in normalized food intake, glucose homeostasis, or metabolic rate. Consistent with their increased lifespan, mTOR(Δ/Δ) mice exhibited a reduction in a number of aging tissue biomarkers. Functional assessment suggested that, as mTOR(Δ/Δ) mice age, they exhibit a marked functional preservation in many, but not all, organ systems. Thus, in a mammalian model, while reducing mTOR expression markedly increases overall lifespan, it affects the age-dependent decline in tissue and organ function in a segmental fashion.
Collapse
Affiliation(s)
- J. Julie Wu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Jie Liu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Edmund B. Chen
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Jennifer J. Wang
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, China Medical University, Shenyang 110001 China
| | - Nisha Narayan
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Marie M. Fergusson
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Ilsa I. Rovira
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Michele Allen
- Murine Phenotyping Core, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Cory U. Lago
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH Bethesda, MD 20892
| | - Wendy DuBois
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH Bethesda, MD 20892
| | - Theresa Ward
- Translational Gerontology Branch, National Institute of Aging, NIH Baltimore MD 21224
| | - Rafael deCabo
- Translational Gerontology Branch, National Institute of Aging, NIH Baltimore MD 21224
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes, Digestive and Kidney Disease, NIH Bethesda, MD 20892
| | - Beverly Mock
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH Bethesda, MD 20892
| | - Toren Finkel
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH Bethesda, MD 20892
| |
Collapse
|
41
|
Impaired β-cell function in the adult offspring of rats fed a protein-restricted diet during lactation is associated with changes in muscarinic acetylcholine receptor subtypes. Br J Nutr 2013; 111:227-35. [DOI: 10.1017/s0007114513002213] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Impaired pancreatic β-cell function, as observed in the cases of early nutrition disturbance, is a major hallmark of metabolic diseases arising in adulthood. In the present study, we aimed to investigate the function/composition of the muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3, in the pancreatic islets of adult offspring of rats that were protein malnourished during lactation. Neonates were nursed by mothers that were fed either a low-protein (4 %, LP) or a normal-protein (23 %, NP) diet. Adult rats were pre-treated with anti-muscarinic drugs and subjected to the glucose tolerance test; the function and protein expression levels of M2mAChR and M3mAChR were determined. The LP rats were lean and hypoinsulinaemic. The selective M2mAChR antagonist methoctramine increased insulinaemia by 31 % in the NP rats and 155 % in the LP rats, and insulin secretion was increased by 32 % in the islets of the NP rats and 88 % in those of the LP rats. The selective M3mAChR antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide decreased insulinaemia by 63 % in the NP rats and 40 % in the LP rats and reduced insulin release by 41 % in the islets of the NP rats and 28 % in those of the LP rats. The protein expression levels of M2mAChR and M3mAChR were 57 % higher and 53 % lower, respectively, in the islets of the LP rats than in those of the NP rats. The expression and functional compositions of M2mAChR and M3mAChR were altered in the islets of the LP rats, as a result of metabolic programming caused by the protein-restricted diet, which might be another possible effect involved in the weak insulin secretion ability of the islets of the programmed adult rats.
Collapse
|
42
|
Abstract
Acetylcholine, the first chemical to be identified as a neurotransmitter, is packed in synaptic vesicles by the activity of VAChT (vesicular acetylcholine transporter). A decrease in VAChT expression has been reported in a number of diseases, and this has consequences for the amount of acetylcholine loaded in synaptic vesicles as well as for neurotransmitter release. Several genetically modified mice targeting the VAChT gene have been generated, providing novel models to understand how changes in VAChT affect transmitter release. A surprising finding is that most cholinergic neurons in the brain also can express a second type of vesicular neurotransmitter transporter that allows these neurons to secrete two distinct neurotransmitters. Thus a given neuron can use two neurotransmitters to regulate different physiological functions. In addition, recent data indicate that non-neuronal cells can also express the machinery used to synthesize and release acetylcholine. Some of these cells rely on VAChT to secrete acetylcholine with potential physiological consequences in the periphery. Hence novel functions for the oldest neurotransmitter known are emerging with the potential to provide new targets for the treatment of several pathological conditions.
Collapse
|
43
|
Lomax MA, Karamanlidis G, Laws J, Cremers SG, Weinberg PD, Clarke L. Pigs fed saturated fat/cholesterol have a blunted hypothalamic-pituitary-adrenal function, are insulin resistant and have decreased expression of IRS-1, PGC1α and PPARα. J Nutr Biochem 2013; 24:656-63. [DOI: 10.1016/j.jnutbio.2012.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/22/2012] [Accepted: 03/15/2012] [Indexed: 10/28/2022]
|
44
|
Zhang S, Hyrc K, Wang S, Wice BM. Xenin-25 increases cytosolic free calcium levels and acetylcholine release from a subset of myenteric neurons. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1347-55. [PMID: 23086920 PMCID: PMC3532549 DOI: 10.1152/ajpgi.00116.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Xenin-25 (Xen) is a 25 amino acid neurotensin-related peptide reportedly produced with glucose-dependent insulinotropic polypeptide (GIP) by a subset of K cells in the proximal gut. We previously showed exogenously administered Xen, with GIP but not alone, increases insulin secretion in humans and mice. In mice, this effect is indirectly mediated via a central nervous system-independent cholinergic relay in the periphery. Xen also delays gastric emptying, reduces food intake, induces gall bladder contractions, and increases gut motility and secretion from the exocrine pancreas, suggesting that some effects of Xen could be mediated by myenteric neurons (MENs). To determine whether Xen activates these neurons, MENs were isolated from guinea pig proximal small intestines. Cells expressed neuronal markers and exhibited typical neuron-like morphology with extensive outgrowths emanating from cell bodies. Cytosolic free Ca(2+) levels ([Ca(2+)](i)) were measured using Fura-2. ATP/UTP, KCl, and forskolin increased [Ca(2+)](i) in 99.6%, 92%, and 23% of the MENs imaged, respectively, indicating that they are functional and activated by nucleotide receptor signaling, direct depolarization, and cAMP. [Ca(2+)](i) increased in only 12.7% of MENs treated with Xen. This rise was blocked by pretreatment with EGTA, diazoxide, SR48692, and neurotensin. Thus the Xen-mediated increase in [Ca(2+)](i) involves influx of extracellular Ca(2+) and activation of neurotensin receptor-1 (NTSR1). Xen also increased acetylcholine release from MENs. Amylin, produced by β-and enteroendocrine cells, delays gastric emptying and increased [Ca(2+)](i) almost exclusively in Xen-responsive MENs. Immunohistochemistry demonstrated NTSR1 expression in human duodenal MENs. Thus myenteric rather than central neurons could mediate some effects of Xen and amylin.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
45
|
Guo T, Bond ND, Jou W, Gavrilova O, Portas J, McPherron AC. Myostatin inhibition prevents diabetes and hyperphagia in a mouse model of lipodystrophy. Diabetes 2012; 61:2414-23. [PMID: 22596054 PMCID: PMC3447905 DOI: 10.2337/db11-0915] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lipodystrophies are characterized by a loss of white adipose tissue, which causes ectopic lipid deposition, peripheral insulin resistance, reduced adipokine levels, and increased food intake (hyperphagia). The growth factor myostatin (MSTN) negatively regulates skeletal muscle growth, and mice with MSTN inhibition have reduced adiposity and improved insulin sensitivity. MSTN inhibition may therefore be efficacious in ameliorating diabetes. To test this hypothesis, we inhibited MSTN signaling in a diabetic model of generalized lipodystrophy to analyze its effects on glucose metabolism separate from effects on adipose mass. A-ZIP/F1 lipodystrophic mice were crossed to mice expressing a dominant-negative MSTN receptor (activin receptor type IIB) in muscle. MSTN inhibition in A-ZIP/F1 mice reduced blood glucose, serum insulin, triglyceride levels, and the rate of triglyceride synthesis, and improved insulin sensitivity. Unexpectedly, hyperphagia was normalized by MSTN inhibition in muscle. Blood glucose and hyperphagia were reduced in double mutants independent of the adipokine leptin. These results show that the effect of MSTN inhibition on insulin sensitivity is not secondary to an effect on adipose mass and that MSTN inhibition may be an effective treatment for diabetes. These results further suggest that muscle may play a heretofore unappreciated role in regulating food intake.
Collapse
Affiliation(s)
- Tingqing Guo
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Nichole D. Bond
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - William Jou
- Mouse Metabolic Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Oksana Gavrilova
- Mouse Metabolic Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jennifer Portas
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Alexandra C. McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
- Corresponding author: Alexandra C. McPherron,
| |
Collapse
|
46
|
Caicedo A. Paracrine and autocrine interactions in the human islet: more than meets the eye. Semin Cell Dev Biol 2012; 24:11-21. [PMID: 23022232 DOI: 10.1016/j.semcdb.2012.09.007] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 09/19/2012] [Indexed: 12/20/2022]
Abstract
The pancreatic islet secretes the hormones insulin and glucagon to regulate glucose metabolism. To generate an adequate secretory response, islet endocrine cells must receive multiple regulatory signals relaying information about changes in the internal and external environments. Islet cells also need to be made aware about the functional status of neighboring cells through paracrine interactions. All this information is used to orchestrate a hormonal response that contributes to glucose homeostasis. Several neurotransmitters have been proposed to work as paracrine signals in the islet. Most of these, however, have yet to meet the criteria to be considered bona fide paracrine signals, in particular in human islets. Here, we review recent findings describing autocrine and paracrine signaling mechanisms in human islets. These recent results are showing an increasingly complex picture of paracrine interactions in the human islet and emphasize that results from other species cannot be readily extrapolated to the human context. Investigators are unveiling new signaling mechanisms or finding new roles for known paracrine signals in human islets. While it is too early to provide a synthesis, the field of islet research is defining the paracrine and autocrine components that will be used to generate models about how islet function is regulated. Meanwhile, the identified signaling pathways can be proposed as therapeutic targets for treating diabetes, a devastating disease affecting millions worldwide.
Collapse
Affiliation(s)
- Alejandro Caicedo
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
47
|
Weston-Green K, Huang XF, Lian J, Deng C. Effects of olanzapine on muscarinic M3 receptor binding density in the brain relates to weight gain, plasma insulin and metabolic hormone levels. Eur Neuropsychopharmacol 2012; 22:364-73. [PMID: 21982116 DOI: 10.1016/j.euroneuro.2011.09.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 08/01/2011] [Accepted: 09/01/2011] [Indexed: 12/19/2022]
Abstract
The second generation antipsychotic drug (SGA) olanzapine has an efficacy to treat schizophrenia, but can cause obesity and type II diabetes mellitus. Cholinergic muscarinic M3 receptors (M3R) are expressed on pancreatic β-cells and in the brain where they influence insulin secretion and may regulate other metabolic hormones via vagal innervation of the gastrointestinal tract. Olanzapine's M3R antagonism is an important risk factor for its diabetogenic liability. However, the effects of olanzapine on central M3Rs are unknown. Rats were treated with 0.25, 0.5, 1.0 or 2.0 mg olanzapine/kg or vehicle (3×/day, 14-days). M3R binding densities in the hypothalamic arcuate (Arc) and ventromedial nuclei (VMH), and dorsal vagal complex (DVC) of the brainstem were investigated using [3H]4-DAMP plus pirenzepine and AF-DX116. M3R binding correlations to body weight, food intake, insulin, ghrelin and cholecystokinin (CCK) were analyzed. Olanzapine increased M3R binding density in the Arc, VMH and DVC, body weight, food intake, circulating plasma ghrelin and CCK levels, and decreased plasma insulin and glucose. M3R negatively correlated to insulin, and positively correlated to ghrelin, CCK, food intake and body weight. Increased M3R density is a compensatory up-regulation in response to olanzapine's M3R antagonism. Olanzapine acts on M3R in regions of the brain that control food intake and insulin secretion. Olanzapine's M3R blockade in the brain may inhibit the acetylcholine pathway for insulin secretion. These findings support a role for M3Rs in the modulation of insulin, ghrelin and CCK via the vagus nerve and provide a mechanism for olanzapine's diabetogenic and weight gain liability.
Collapse
Affiliation(s)
- Katrina Weston-Green
- Centre for Translational Neuroscience, School of Health Sciences, University of Wollongong, Wollongong, 2522, NSW, Australia
| | | | | | | |
Collapse
|
48
|
Abstract
Muscarinic acetylcholine (ACh) receptors (mAChRs; M₁-M₅) regulate the activity of an extraordinarily large number of important physiological processes. During the past 10-15 years, studies with whole-body M₁-M₅ mAChR knockout mice have provided many new insights into the physiological and pathophysiological roles of the individual mAChR subtypes. This review will focus on the characterization of a novel generation of mAChR mutant mice, including mice in which distinct mAChR genes have been excised in a tissue- or cell type-specific fashion, various transgenic mouse lines that overexpress wild-type or different mutant M₃ mAChRs in certain tissues or cells only, as well as a novel M₃ mAChR knockin mouse strain deficient in agonist-induced M₃ mAChR phosphorylation. Phenotypic analysis of these new animal models has greatly advanced our understanding of the physiological roles of the various mAChR subtypes and has identified potential targets for the treatment of type 2 diabetes, schizophrenia, Parkinson's disease, drug addiction, cognitive disorders, and several other pathophysiological conditions.
Collapse
|
49
|
Wu G, Zhang L, Li T, Lopaschuk G, Vance DE, Jacobs RL. Choline Deficiency Attenuates Body Weight Gain and Improves Glucose Tolerance in ob/ob Mice. J Obes 2012; 2012:319172. [PMID: 22778916 PMCID: PMC3385711 DOI: 10.1155/2012/319172] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 02/02/2012] [Accepted: 02/16/2012] [Indexed: 02/07/2023] Open
Abstract
Previous studies demonstrated that choline supply is directly linked to high-fat-diet-induced obesity and insulin resistance in mice. The aim of this study was to evaluate if choline supply could also modulate obesity and insulin resistance caused by a genetic defect. Eight-week-old male ob/ob mice were fed for two months with either choline-deficient or choline-supplemented diet. Tissue weight including fat mass and lean mass was assessed. Intracellular signaling, plasma glucagon and insulin, and glucose and insulin tolerance tests were also investigated. The choline-deficient diet slowed body weight gain and decreased fat mass. Choline deficiency also decreased plasma glucose level and improved glucose and insulin tolerance although fatty liver was exacerbated. Increased adipose lipolytic activity, decreased plasma glucagon and reduced expression of hepatic glucagon receptor were also observed with the choline-deficient diet. Our results demonstrate that a choline-deficient diet can decrease fat mass and improve glucose tolerance in obese and diabetic mice caused by a genetic defect.
Collapse
Affiliation(s)
- Gengshu Wu
- Group on the Molecular and Cell Biology of Lipids and Department of Biochemistry, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Liyan Zhang
- Cardiovascular Research Group, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Tete Li
- Group on the Molecular and Cell Biology of Lipids and Department of Biochemistry, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Gary Lopaschuk
- Cardiovascular Research Group, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Dennis E. Vance
- Group on the Molecular and Cell Biology of Lipids and Department of Biochemistry, University of Alberta, Edmonton, AB, Canada T6G 2S2
- *Dennis E. Vance:
| | - René L. Jacobs
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada T6G 2S2
| |
Collapse
|
50
|
Satyanarayana A, Klarmann KD, Gavrilova O, Keller JR. Ablation of the transcriptional regulator Id1 enhances energy expenditure, increases insulin sensitivity, and protects against age and diet induced insulin resistance, and hepatosteatosis. FASEB J 2011; 26:309-23. [PMID: 21990377 DOI: 10.1096/fj.11-190892] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Obesity is a major health concern that contributes to the development of diabetes, hyperlipidemia, coronary artery disease, and cancer. Id proteins are helix-loop-helix transcription factors that regulate the proliferation and differentiation of cells from multiple tissues, including adipocytes. We screened mouse tissues for the expression of Id1 and found that Id1 protein is highly expressed in brown adipose tissue (BAT) and white adipose tissue (WAT), suggesting a role for Id1 in adipogenesis and cell metabolism. Id1(-/-) mice are viable but show a significant reduction in fat mass (P<0.005) over the life of the animal that was not due to decreased number of adipocytes. Analysis of Id1(-/-) mice revealed higher energy expenditure, increased lipolysis, and fatty acid oxidation, resulting in reduced triglyceride accumulation in WAT compared to Id1(+/+) mice. Serum levels of triglycerides (193.9±32.2 vs. 86.5±33.8, P<0.0005), cholesterol (189.4±33.8 vs. 110.6±8.23, P<0.0005) and leptin (1263±835 vs. 222±260, P<0.005) were significantly lower in aged Id1(-/-) mice compared to Id1(+/+) mice. Id1-deficient mice have higher resting (P<0.005) and total (P<0.05) O(2) consumption and lower respiratory exchange ratio (P<0.005), confirming that Id1(-/-) mice use a higher proportion of lipid as an energy source for the increased energy expenditure. The expression of PGC1α and UCP1 were 2- to 3-fold up-regulated in Id1(-/-) BAT, suggesting that loss of Id1 increases thermogenesis. As a consequence of higher energy expenditure and reduced fat mass, Id1(-/-) mice displayed enhanced insulin sensitivity. Id1 deficiency protected mice against age- and high-fat-diet-induced adiposity, insulin resistance, and hepatosteatosis. Our findings suggest that Id1 plays a critical role in the regulation of energy homeostasis and could be a potential target in the treatment of insulin resistance and fatty liver disease.
Collapse
Affiliation(s)
- Ande Satyanarayana
- Center for Cancer Research, National Cancer, Institute-Frederick, Frederick, MD 21702-1201, USA
| | | | | | | |
Collapse
|