1
|
Santiago-Marrero I, Liu F, Wang H, Arzola EP, Xiong WC, Mei L. Energy Expenditure Homeostasis Requires ErbB4, an Obesity Risk Gene, in the Paraventricular Nucleus. eNeuro 2023; 10:ENEURO.0139-23.2023. [PMID: 37669858 PMCID: PMC10521346 DOI: 10.1523/eneuro.0139-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Obesity affects more than a third adult population in the United States; the prevalence is even higher in patients with major depression disorders. GWAS studies identify the receptor tyrosine kinase ErbB4 as a risk gene for obesity and for major depression disorders. We found that ErbB4 was enriched in the paraventricular nucleus of the hypothalamus (PVH). To investigate its role in metabolism, we deleted ErbB4 by injecting a Cre-expressing virus into the PVH of ErbB4-floxed male mice and found that PVH ErbB4 deletion increased weight gain without altering food intake. ErbB4 PVH deletion also reduced nighttime activity and decreased intrascapular brown adipose tissue (iBAT) thermogenesis. Analysis of covariance (ANCOVA) revealed that ErbB4 PVH deletion reduced O2 consumption, CO2 production and heat generation in a manner independent of body weight. Immunostaining experiments show that ErbB4+ neurons in the PVH were positive for oxytocin (OXT); ErbB4 PVH deletion reduces serum levels of OXT. We characterized mice where ErbB4 was specifically mutated in OXT+ neurons and found reduction in energy expenditure, phenotypes similar to PVH ErbB4 deletion. Taken together, our data indicate that ErbB4 in the PVH regulates metabolism likely through regulation of OXT expressing neurons, reveal a novel function of ErbB4 and provide insight into pathophysiological mechanisms of depression-associated obesity.
Collapse
Affiliation(s)
- Ivan Santiago-Marrero
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Fang Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Emily P Arzola
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
- Chinese Institutes for Medical Research, Beijing 100005, China
- Capital Medical University, Beijing 100054, China
| |
Collapse
|
2
|
Gene expression associations with body mass index in the Multi-Ethnic Study of Atherosclerosis. Int J Obes (Lond) 2023; 47:109-116. [PMID: 36463326 PMCID: PMC9990473 DOI: 10.1038/s41366-022-01240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND/OBJECTIVES Obesity, defined as excessive fat accumulation that represents a health risk, is increasing in adults and children, reaching global epidemic proportions. Body mass index (BMI) correlates with body fat and future health risk, yet differs in prediction by fat distribution, across populations and by age. Nonetheless, few genetic studies of BMI have been conducted in ancestrally diverse populations. Gene expression association with BMI was assessed in the Multi-Ethnic Study of Atherosclerosis (MESA) in four self-identified race and ethnicity (SIRE) groups to identify genes associated with obesity. SUBJECTS/METHODS RNA-sequencing was performed on 1096 MESA participants (37.8% white, 24.3% Hispanic, 28.4% African American, and 9.5% Chinese American) and linear models were used to assess the association of expression from each gene for its effect on BMI, adjusting for age, sex, sequencing center, study site, five expression and four genetic principal components in each self-identified race group. Sample-size-weighted meta-analysis was performed to identify genes with BMI-associated expression across ancestry groups. RESULTS Within individual SIRE groups, there were zero to three genes whose expression is significantly (p < 1.97 × 10-6) associated with BMI. Across all groups, 45 genes were identified by meta-analysis whose expression was significantly associated with BMI, explaining 29.7% of BMI variation. The 45 genes are expressed in a variety of tissues and cell types and are enriched for obesity-related processes including erythrocyte function, oxygen binding and transport, and JAK-STAT signaling. CONCLUSIONS We have identified genes whose expression is significantly associated with obesity in a multi-ethnic cohort. We have identified novel genes associated with BMI as well as confirmed previously identified genes from earlier genetic analyses. These novel genes and their biological pathways represent new targets for understanding the biology of obesity as well as new therapeutic intervention to reduce obesity and improve global public health.
Collapse
|
3
|
Micó-Carnero M, Casillas-Ramírez A, Sánchez-González A, Rojano-Alfonso C, Peralta C. The Role of Neuregulin-1 in Steatotic and Non-Steatotic Liver Transplantation from Brain-Dead Donors. Biomedicines 2022; 10:biomedicines10050978. [PMID: 35625715 PMCID: PMC9138382 DOI: 10.3390/biomedicines10050978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/29/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background. Brain death (BD) and steatosis are key risk factors to predict adverse post-transplant outcomes. We investigated the role of Neuregulin-1 (NRG1) in rat steatotic and non-steatotic liver transplantation (LT) from brain death donors (DBD). Methods: NRG1 pathways were characterized after surgery. Results: NRG1 and p21-activated kinase 1 (PAK1) levels increased in steatotic and non-steatotic grafts from DBDs. The abolishment of NRG1 effects reduced PAK1. When the effect of either NRG1 nor PAK1 was inhibited, injury and regenerative failure were exacerbated. The benefits of the NRG-1-PAK1 axis in liver grafts from DBDs were associated with increased vascular endothelial growth factor-A (VEGFA) and insulin growth factor-1 (IGF1) levels, respectively. Indeed, VEGFA administration in non-steatotic livers and IGF1 treatment in steatotic grafts prevented damage and regenerative failure resulting from the inhibition of either NRG1 or PAK-1 activity in each type of liver. Exogenous NRG1 induced greater injury than BD induction. Conclusions: This study indicates the benefits of endogenous NRG1 in liver grafts from DBDs and underscores the specificity of the NRG1 signaling pathway depending on the type of liver: NRG1-PAK1-VEGFA in non-steatotic livers and NRG1-PAK1-IGF1 in steatotic livers. Exogenous NRG1 is not an appropriate strategy to apply to liver grafts from DBD.
Collapse
Affiliation(s)
- Marc Micó-Carnero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico; (A.C.-R.); (A.S.-G.)
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros 87300, Mexico
| | - Alfredo Sánchez-González
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico; (A.C.-R.); (A.S.-G.)
| | - Carlos Rojano-Alfonso
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
- Correspondence: ; Tel.: +34-932-275-400
| |
Collapse
|
4
|
Vega-Torres JD, Ontiveros-Angel P, Terrones E, Stuffle EC, Solak S, Tyner E, Oropeza M, dela Peña I, Obenaus A, Ford BD, Figueroa JD. Short-term exposure to an obesogenic diet during adolescence elicits anxiety-related behavior and neuroinflammation: modulatory effects of exogenous neuregulin-1. Transl Psychiatry 2022; 12:83. [PMID: 35220393 PMCID: PMC8882169 DOI: 10.1038/s41398-022-01788-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 11/21/2022] Open
Abstract
Childhood obesity leads to hippocampal atrophy and altered cognition. However, the molecular mechanisms underlying these impairments are poorly understood. The neurotrophic factor neuregulin-1 (NRG1) and its cognate ErbB4 receptor play critical roles in hippocampal maturation and function. This study aimed to determine whether exogenous NRG1 administration reduces hippocampal abnormalities and neuroinflammation in rats exposed to an obesogenic Western-like diet (WD). Lewis rats were randomly divided into four groups (12 rats/group): (1) control diet+vehicle (CDV); (2) CD + NRG1 (CDN) (daily intraperitoneal injections: 5 μg/kg/day; between postnatal day, PND 21-PND 41); (3) WD + VEH (WDV); (4) WD + NRG1 (WDN). Neurobehavioral assessments were performed at PND 43-49. Brains were harvested for MRI and molecular analyses at PND 49. We found that NRG1 administration reduced hippocampal volume (7%) and attenuated hippocampal-dependent cued fear conditioning in CD rats (56%). NRG1 administration reduced PSD-95 protein expression (30%) and selectively reduced hippocampal cytokine levels (IL-33, GM-CSF, CCL-2, IFN-γ) while significantly impacting microglia morphology (increased span ratio and reduced circularity). WD rats exhibited reduced right hippocampal volume (7%), altered microglia morphology (reduced density and increased lacunarity), and increased levels of cytokines implicated in neuroinflammation (IL-1α, TNF-α, IL-6). Notably, NRG1 synergized with the WD to increase hippocampal ErbB4 phosphorylation and the tumor necrosis alpha converting enzyme (TACE/ADAM17) protein levels. Although the results did not provide sufficient evidence to conclude that exogenous NRG1 administration is beneficial to alleviate obesity-related outcomes in adolescent rats, we identified a potential novel interaction between obesogenic diet exposure and TACE/ADAM17-NRG1-ErbB4 signaling during hippocampal maturation. Our results indicate that supraoptimal ErbB4 activities may contribute to the abnormal hippocampal structure and cognitive vulnerabilities observed in obese individuals.
Collapse
Affiliation(s)
- Julio David Vega-Torres
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Perla Ontiveros-Angel
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Esmeralda Terrones
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Erwin C. Stuffle
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Sara Solak
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Emma Tyner
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Marie Oropeza
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Ike dela Peña
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Andre Obenaus
- grid.266093.80000 0001 0668 7243Department of Pediatrics, University of California-Irvine, Irvine, CA USA
| | - Byron D. Ford
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA USA
| | - Johnny D. Figueroa
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| |
Collapse
|
5
|
Götze T, Soto-Bernardini MC, Zhang M, Mießner H, Linhoff L, Brzózka MM, Velanac V, Dullin C, Ramos-Gomes F, Peng M, Husseini H, Schifferdecker E, Fledrich R, Sereda MW, Willig K, Alves F, Rossner MJ, Nave KA, Zhang W, Schwab MH. Hyperactivity is a Core Endophenotype of Elevated Neuregulin-1 Signaling in Embryonic Glutamatergic Networks. Schizophr Bull 2021; 47:1409-1420. [PMID: 33871014 PMCID: PMC8379540 DOI: 10.1093/schbul/sbab027] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The neuregulin 1 (NRG1) ErbB4 module is at the core of an "at risk" signaling pathway in schizophrenia. Several human studies suggest hyperstimulation of NRG1-ErbB4 signaling as a plausible pathomechanism; however, little is known about the significance of stage-, brain area-, or neural cell type-specific NRG1-ErbB4 hyperactivity for disease-relevant brain endophenotypes. To address these spatiotemporal aspects, we generated transgenic mice for Cre recombinase-mediated overexpression of cystein-rich domain (CRD) NRG1, the most prominent NRG1 isoform in the brain. A comparison of "brain-wide" vs cell type-specific CRD-NRG1 overexpressing mice revealed that pathogenic CRD-NRG1 signals for ventricular enlargement and neuroinflammation originate outside glutamatergic neurons and suggests a subcortical function of CRD-NRG1 in the control of body weight. Embryonic onset of CRD-NRG1 in glutamatergic cortical networks resulted in reduced inhibitory neurotransmission and locomotor hyperactivity. Our findings identify ventricular enlargement and locomotor hyperactivity, 2 main endophenotypes of schizophrenia, as specific consequences of spatiotemporally distinct expression profiles of hyperactivated CRD-NRG1 signaling.
Collapse
Affiliation(s)
- Tilmann Götze
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Maria Clara Soto-Bernardini
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Present address: Center for Research in Biotechnology (CIB)/Costa Rica Institute of Technology (TEC), Cartago, Costa Rica
| | - Mingyue Zhang
- Laboratory of Molecular Psychiatry, Department of Mental Health, Westfälische Wilhelm-University of Münster, Münster, Germany
| | - Hendrik Mießner
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany,Present address: Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Lisa Linhoff
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Department of Neurology, University Medicine Göttingen (UMG), Göttingen, Germany
| | - Magdalena M Brzózka
- Department of Psychiatry, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Viktorija Velanac
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Christian Dullin
- Institute for Diagnostic and Interventional Radiology, University Medical Center, Goettingen, Germany,Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Italian Synchrotron “Elettra,"Trieste, Italy
| | - Fernanda Ramos-Gomes
- Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Maja Peng
- Laboratory of Molecular Psychiatry, Department of Mental Health, Westfälische Wilhelm-University of Münster, Münster, Germany
| | - Hümeyra Husseini
- Laboratory of Molecular Psychiatry, Department of Mental Health, Westfälische Wilhelm-University of Münster, Münster, Germany
| | - Eva Schifferdecker
- Laboratory of Molecular Psychiatry, Department of Mental Health, Westfälische Wilhelm-University of Münster, Münster, Germany
| | - Robert Fledrich
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Michael W Sereda
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Department of Neurology, University Medicine Göttingen (UMG), Göttingen, Germany
| | - Katrin Willig
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany,Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Frauke Alves
- Institute for Diagnostic and Interventional Radiology, University Medical Center, Goettingen, Germany,Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Moritz J Rossner
- Department of Psychiatry, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Mental Health, Westfälische Wilhelm-University of Münster, Münster, Germany
| | - Markus H Schwab
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany,Cellular Neurophysiology, Hannover Medical School, Hannover, Germany,Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany,To whom correspondence should be addressed; tel: +49-341-97-25677; fax: +49-341-97-15049, e-mail:
| |
Collapse
|
6
|
Meng D, Pan H, Chen Y, Ding J, Dai Y. Roles and mechanisms of NRG1 in modulating the pathogenesis of NAFLD through ErbB3 signaling in hepatocytes (NRG1 modulates NAFLD through ErbB3 signaling). Obes Res Clin Pract 2021; 15:145-151. [PMID: 33541789 DOI: 10.1016/j.orcp.2021.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is an emerging chronic liver disease. However, the underlying mechanisms remained poorly understood. Neuregulin (NRG) family participate in energy metabolism, and might be related to NAFLD. METHODS L02 cells were exposed to oleic acid to establish a cellular model of NAFLD. We analyzed the NAFLD cells with NRG1 and subsequent ErbB3 siRNA treatment. Cellular total lipid was stained by Oil Red O, while triglyceride content and inflammation markers were measured by enzymatic kits. The expressions of down-stream molecules were evaluated by western blot. RESULTS In vitro, NRG1 could alleviate the steatosis of NAFLD, and inhibit the expression of IL-6 and TNF-α. The downregulation of ErbB3 aggravated steatosis, improved the levels of triglyceride, IL-6 and TNF-α in NRG1-treated NAFLD. Moreover, NRG1 treatment up-regulated ErbB3 phosphorylation, and increased the expression of PI3K and phosphorylation-AKT. When NRG1-treated NAFLD cells were transfected with ErbB3 siRNA, the expressions of ErbB3, p-ErbB3, p-AKT and PI3K were all reduced. CONCLUSION NRG1 might play a protective role in the pathogenesis of NAFLD through ErbB3 phosphorylation to modulate the activation of PI3K-AKT pathway. The findings will expand the understanding of the mechanisms of NAFLD, and provide potential therapeutic targets.
Collapse
Affiliation(s)
- Di Meng
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| | - Hongying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Youwei Chen
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Jiexia Ding
- Department of Infectious Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yining Dai
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
7
|
Zhang L, Lu B, Wang W, Miao S, Zhou S, Cheng X, Zhu J, Liu C. Alteration of serum neuregulin 4 and neuregulin 1 in gestational diabetes mellitus. Ther Adv Endocrinol Metab 2021; 12:20420188211049614. [PMID: 34646438 PMCID: PMC8504227 DOI: 10.1177/20420188211049614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/12/2021] [Indexed: 12/14/2022] Open
Abstract
CONTEXT Neuregulin 4 (Nrg4) and neuregulin 1 (Nrg1) have been shown to play vital roles in several disorders of glucose metabolism. The pathophysiological role of Nrg4 and Nrg1 in gestational diabetes mellitus (GDM), however, remains poorly understood. We assessed the clinical relevance of the two cytokines in patients with GDM. METHODS The study recruited 36 GDM patients and 38 age-matched, gestational age (24-28 weeks of gestation)-matched, and BMI (during pregnancy)-matched controls in this study. Serum Nrg4 and Nrg1 were measured using ELISA. Inflammatory factors such as IL-6, IL-1β, leptin, TNF-α, and monocyte chemotactic protein 1 (MCP-1) were determined via Luminex technique. RESULTS Serum Nrg4 in GDM patients was significantly lower than that in the controls, while Nrg1 was significantly higher in the GDM group (p < 0.01). Inflammatory factors such as IL-6, leptin, and TNF-α were significantly increased in GDM patients, while MCP-1 and IL-1β were not significantly different between the two groups. In addition, serum Nrg4 was negatively correlated with fasting glucose (r = -0.438, p = 0.008), HOMA-IR (r = -0.364, p = 0.029), IL-6 (r = -0.384, p = 0.021), leptin (r = -0.393, p = 0.018), TNF-α (r = -0.346, p = 0.039), and MCP-1 (r = -0.342, p = 0.041), and positively correlated with high-density lipoprotein cholesterol (HDL-C) (r = -0.357, p = 0.033) in GDM group. Serum Nrg1 was positively correlated with BMI (r = 0.452, p = 0.006), fasting glucose (r = 0.424, p = 0.010), HOMA-IR (r = 0.369, p = 0.027), and triglyceride (r = 0.439, p = 0.007). The decrease of Nrg4 and the increase of Nrg1 were significantly related to the increased prevalence of GDM. Finally, ROC curve results indicated that Nrg1 combined with IL-6 and TNF-α might be an effective means for GDM screening. CONCLUSIONS Lower circulating Nrg4 and higher circulating Nrg1 serve risk factors of GDM. Nrg1 combined with IL-6 and TNF-α might be a potential tool for GDM screening.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, Binzhou, China
| | - Bi Lu
- Department of Rheumatology and Endocrinology, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Wenhua Wang
- Department of Neurology, Wuhan Fourth Hospital, Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shifeng Miao
- Department of Cardiology, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Shuru Zhou
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Xingbo Cheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Zhu
- Department of Cardiology, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | | |
Collapse
|
8
|
Chitre AS, Polesskaya O, Holl K, Gao J, Cheng R, Bimschleger H, Garcia Martinez A, George T, Gileta AF, Han W, Horvath A, Hughson A, Ishiwari K, King CP, Lamparelli A, Versaggi CL, Martin C, St Pierre CL, Tripi JA, Wang T, Chen H, Flagel SB, Meyer P, Richards J, Robinson TE, Palmer AA, Solberg Woods LC. Genome-Wide Association Study in 3,173 Outbred Rats Identifies Multiple Loci for Body Weight, Adiposity, and Fasting Glucose. Obesity (Silver Spring) 2020; 28:1964-1973. [PMID: 32860487 PMCID: PMC7511439 DOI: 10.1002/oby.22927] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Obesity is influenced by genetic and environmental factors. Despite the success of human genome-wide association studies, the specific genes that confer obesity remain largely unknown. The objective of this study was to use outbred rats to identify the genetic loci underlying obesity and related morphometric and metabolic traits. METHODS This study measured obesity-relevant traits, including body weight, body length, BMI, fasting glucose, and retroperitoneal, epididymal, and parametrial fat pad weight in 3,173 male and female adult N/NIH heterogeneous stock (HS) rats across three institutions, providing data for the largest rat genome-wide association study to date. Genetic loci were identified using a linear mixed model to account for the complex family relationships of the HS and using covariates to account for differences among the three phenotyping centers. RESULTS This study identified 32 independent loci, several of which contained only a single gene (e.g., Epha5, Nrg1, Klhl14) or obvious candidate genes (e.g., Adcy3, Prlhr). There were strong phenotypic and genetic correlations among obesity-related traits, and there was extensive pleiotropy at individual loci. CONCLUSIONS This study demonstrates the utility of HS rats for investigating the genetics of obesity-related traits across institutions and identify several candidate genes for future functional testing.
Collapse
Affiliation(s)
- Apurva S Chitre
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Katie Holl
- Human and Molecular Genetic Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jianjun Gao
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Hannah Bimschleger
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Angel Garcia Martinez
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tony George
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, New York, USA
| | - Alexander F Gileta
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Department of Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Wenyan Han
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Aidan Horvath
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Alesa Hughson
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Keita Ishiwari
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, New York, USA
| | | | | | | | - Connor Martin
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, New York, USA
| | | | - Jordan A Tripi
- Department of Psychology, University at Buffalo, Buffalo, New York, USA
| | - Tengfei Wang
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Hao Chen
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Shelly B Flagel
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul Meyer
- Department of Psychology, University at Buffalo, Buffalo, New York, USA
| | - Jerry Richards
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, New York, USA
| | - Terry E Robinson
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
9
|
Epigenetic Regulation of Neuregulin-1 Tunes White Adipose Stem Cell Differentiation. Cells 2020; 9:cells9051148. [PMID: 32392729 PMCID: PMC7290571 DOI: 10.3390/cells9051148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 11/17/2022] Open
Abstract
Expansion of subcutaneous adipose tissue by differentiation of new adipocytes has been linked to improvements in metabolic health. However, an expandability limit has been observed wherein new adipocytes cannot be produced, the existing adipocytes become enlarged (hypertrophic) and lipids spill over into ectopic sites. Inappropriate ectopic storage of these surplus lipids in liver, muscle, and visceral depots has been linked with metabolic dysfunction. Here we show that Neuregulin-1 (NRG1) serves as a regulator of adipogenic differentiation in subcutaneous primary human stem cells. We further demonstrate that DNA methylation modulates NRG1 expression in these cells, and a 3-day exposure of stem cells to a recombinant NRG1 peptide fragment is sufficient to reprogram adipogenic cellular differentiation to higher levels. These results define a novel molecular adipogenic rheostat with potential implications for the expansion of adipose tissue in vivo.
Collapse
|
10
|
Zhao M, Jung Y, Jiang Z, Svensson KJ. Regulation of Energy Metabolism by Receptor Tyrosine Kinase Ligands. Front Physiol 2020; 11:354. [PMID: 32372975 PMCID: PMC7186430 DOI: 10.3389/fphys.2020.00354] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic diseases, such as diabetes, obesity, and fatty liver disease, have now reached epidemic proportions. Receptor tyrosine kinases (RTKs) are a family of cell surface receptors responding to growth factors, hormones, and cytokines to mediate a diverse set of fundamental cellular and metabolic signaling pathways. These ligands signal by endocrine, paracrine, or autocrine means in peripheral organs and in the central nervous system to control cellular and tissue-specific metabolic processes. Interestingly, the expression of many RTKs and their ligands are controlled by changes in metabolic demand, for example, during starvation, feeding, or obesity. In addition, studies of RTKs and their ligands in regulating energy homeostasis have revealed unexpected diversity in the mechanisms of action and their specific metabolic functions. Our current understanding of the molecular, biochemical and genetic control of energy homeostasis by the endocrine RTK ligands insulin, FGF21 and FGF19 are now relatively well understood. In addition to these classical endocrine signals, non-endocrine ligands can govern local energy regulation, and the intriguing crosstalk between the RTK family and the TGFβ receptor family demonstrates a signaling network that diversifies metabolic process between tissues. Thus, there is a need to increase our molecular and mechanistic understanding of signal diversification of RTK actions in metabolic disease. Here we review the known and emerging molecular mechanisms of RTK signaling that regulate systemic glucose and lipid metabolism, as well as highlighting unexpected roles of non-classical RTK ligands that crosstalk with other receptor pathways.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Yunshin Jung
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Zewen Jiang
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| | - Katrin J Svensson
- Department of Pathology, Stanford University, Stanford, CA, United States.,Stanford Diabetes Research Center, Stanford, CA, United States
| |
Collapse
|
11
|
De Keulenaer GW, Feyen E, Dugaucquier L, Shakeri H, Shchendrygina A, Belenkov YN, Brink M, Vermeulen Z, Segers VFM. Mechanisms of the Multitasking Endothelial Protein NRG-1 as a Compensatory Factor During Chronic Heart Failure. Circ Heart Fail 2019; 12:e006288. [DOI: 10.1161/circheartfailure.119.006288] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1—a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.
Collapse
Affiliation(s)
- Gilles W. De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium (G.W.D.K.)
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Lindsey Dugaucquier
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Anastasia Shchendrygina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Yury N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Marijke Brink
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland (M.B.)
| | - Zarha Vermeulen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Vincent F. M. Segers
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, University Hospital Antwerp, Edegem, Belgium (V.F.M.S.)
| |
Collapse
|
12
|
Zhang P, Kuang H, He Y, Idiga SO, Li S, Chen Z, Yang Z, Cai X, Zhang K, Potthoff MJ, Xu Y, Lin JD. NRG1-Fc improves metabolic health via dual hepatic and central action. JCI Insight 2018. [PMID: 29515030 DOI: 10.1172/jci.insight.98522] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neuregulins (NRGs) are emerging as an important family of signaling ligands that regulate glucose and lipid homeostasis. NRG1 lowers blood glucose levels in obese mice, whereas the brown fat-enriched secreted factor NRG4 protects mice from high-fat diet-induced insulin resistance and hepatic steatosis. However, the therapeutic potential of NRGs remains elusive, given the poor plasma half-life of the native ligands. Here, we engineered a fusion protein using human NRG1 and the Fc domain of human IgG1 (NRG1-Fc) that exhibited extended half-life in circulation and improved potency in receptor signaling. We evaluated its efficacy in improving metabolic parameters and dissected the mechanisms of action. NRG1-Fc treatment triggered potent AKT activation in the liver, lowered blood glucose, improved insulin sensitivity, and suppressed food intake in obese mice. NRG1-Fc acted as a potent secretagogue for the metabolic hormone FGF21; however, the latter was largely dispensable for its metabolic effects. NRG1-Fc directly targeted the hypothalamic POMC neurons to promote membrane depolarization and increase firing rate. Together, NRG1-Fc exhibits improved pharmacokinetic properties and exerts metabolic benefits through dual inhibition of hepatic gluconeogenesis and caloric intake.
Collapse
Affiliation(s)
- Peng Zhang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Henry Kuang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sharon O Idiga
- Department of Pharmacology and Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Siming Li
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Department of Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Department of Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Matthew J Potthoff
- Department of Pharmacology and Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Declerck K, Remy S, Wohlfahrt-Veje C, Main KM, Van Camp G, Schoeters G, Vanden Berghe W, Andersen HR. Interaction between prenatal pesticide exposure and a common polymorphism in the PON1 gene on DNA methylation in genes associated with cardio-metabolic disease risk-an exploratory study. Clin Epigenetics 2017; 9:35. [PMID: 28396702 PMCID: PMC5382380 DOI: 10.1186/s13148-017-0336-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/30/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Prenatal environmental conditions may influence disease risk in later life. We previously found a gene-environment interaction between the paraoxonase 1 (PON1) Q192R genotype and prenatal pesticide exposure leading to an adverse cardio-metabolic risk profile at school age. However, the molecular mechanisms involved have not yet been resolved. It was hypothesized that epigenetics might be involved. The aim of the present study was therefore to investigate whether DNA methylation patterns in blood cells were related to prenatal pesticide exposure level, PON1 Q192R genotype, and associated metabolic effects observed in the children. METHODS Whole blood DNA methylation patterns in 48 children (6-11 years of age), whose mothers were occupationally unexposed or exposed to pesticides early in pregnancy, were determined by Illumina 450 K methylation arrays. RESULTS A specific methylation profile was observed in prenatally pesticide exposed children carrying the PON1 192R-allele. Differentially methylated genes were enriched in several neuroendocrine signaling pathways including dopamine-DARPP32 feedback (appetite, reward pathways), corticotrophin releasing hormone signaling, nNOS, neuregulin signaling, mTOR signaling, and type II diabetes mellitus signaling. Furthermore, we were able to identify possible candidate genes which mediated the associations between pesticide exposure and increased leptin level, body fat percentage, and difference in BMI Z score between birth and school age. CONCLUSIONS DNA methylation may be an underlying mechanism explaining an adverse cardio-metabolic health profile in children carrying the PON1 192R-allele and prenatally exposed to pesticides.
Collapse
Affiliation(s)
- Ken Declerck
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
| | - Sylvie Remy
- Department of Epidemiology and Social Medicine, Antwerp University, Universiteitsplein 1, Antwerp, Belgium.,Flemish Institute for Technological Research (VITO), Unit Environmental Risk and Health, Boeretang 200, Mol, Belgium
| | - Christine Wohlfahrt-Veje
- Department of Growth and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Greet Schoeters
- Flemish Institute for Technological Research (VITO), Unit Environmental Risk and Health, Boeretang 200, Mol, Belgium.,Department of Biomedical Sciences, Antwerp University, Universiteitsplein 1, Antwerp, Belgium.,Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
| | - Helle R Andersen
- Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
14
|
Ennequin G, Boisseau N, Caillaud K, Chavanelle V, Etienne M, Li X, Sirvent P. Neuregulin 1 Improves Glucose Tolerance in db/db Mice. PLoS One 2015; 10:e0130568. [PMID: 26230680 PMCID: PMC4521942 DOI: 10.1371/journal.pone.0130568] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 05/21/2015] [Indexed: 01/14/2023] Open
Abstract
In vitro experiments using rodent skeletal muscle cells suggest that neuregulin 1 (NRG1) is involved in glucose metabolism regulation, although no study has evaluated the role of NRG1 in systemic glucose homeostasis. The purpose of this study was to investigate the effect of chronic and acute NRG1 treatment on glucose homeostasis in db/db mice. To this aim, glucose tolerance tests were performed in 8-week-old male db/db mice after treatment with NRG1 (50μg.kg-1) or saline 3 times per week for 8 weeks. In other experiments, glucose tolerance and pyruvate tolerance tests were performed in db/db mice 15 minutes after a single NRG1 (50μg.kg-1) or saline injection. Liver, adipose tissue, hypothalamus and skeletal muscle were also collected 30 minutes after acute NRG1 (50μg.kg-1) or saline treatment, and the phosphorylation status of the ERBB receptors, AKT (on Ser473) and FOXO1 (on Ser256) was assessed by western blotting. Chronic treatment (8 weeks) with NRG1 improved glucose tolerance in db/db mice. Acute treatment also lowered glycemia and insulinemia during glucose or pyruvate tolerance tests. NRG1 acute injection induced activation of ERBB3 receptors and phosphorylation of AKT and FOXO1 only in liver. Altogether, this study shows that acute and chronic NRG1 treatments improve glucose tolerance in db/db mice. This effect could be mediated through inhibition of hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Gaël Ennequin
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Nathalie Boisseau
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Kevin Caillaud
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Vivien Chavanelle
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Monique Etienne
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Xinyan Li
- Zensun Sci & Tech Ltd., Shanghai, China
| | - Pascal Sirvent
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l’Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France
- * E-mail:
| |
Collapse
|
15
|
Ennequin G, Boisseau N, Caillaud K, Chavanelle V, Gerbaix M, Metz L, Etienne M, Walrand S, Masgrau A, Guillet C, Courteix D, Niu A, Li YP, Capel F, Sirvent P. Exercise training and return to a well-balanced diet activate the neuregulin 1/ErbB pathway in skeletal muscle of obese rats. J Physiol 2015; 593:2665-77. [PMID: 25820551 DOI: 10.1113/jp270026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 03/17/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Some studies suggest that neuregulin 1 (NRG1) could be involved in the regulation of skeletal muscle energy metabolism in rodents. Here we assessed whether unbalanced diet is associated with alterations of the NRG1 signalling pathway and whether exercise and diet might restore NRG1 signalling in skeletal muscle of obese rats. We show that diet-induced obesity does not impair NRG1 signalling in rat skeletal muscle. We also report that endurance training and a well-balanced diet activate the NRG1 signalling in skeletal muscle of obese rats, possibly via a new mechanism mediated by the protease ADAM17. These results suggest that some beneficial effects of physical activity and diet in obese rats could be partly explained by stimulation of the NRG1 signalling pathway. ABSTRACT Some studies suggest that the signalling pathway of neuregulin 1 (NRG1), a protein involved in the regulation of skeletal muscle metabolism, could be altered by nutritional and exercise interventions. We hypothesized that diet-induced obesity could lead to alterations of the NRG1 signalling pathway and that chronic exercise could improve NRG1 signalling in rat skeletal muscle. To test this hypothesis, male Wistar rats received a high fat/high sucrose (HF/HS) diet for 16 weeks. At the end of this period, NRG1 and ErbB expression/activity in skeletal muscle was assessed. The obese rats then continued the HF/HS diet or were switched to a well-balanced diet. Moreover, in both groups, half of the animals also performed low intensity treadmill exercise training. After another 8 weeks, NRG1 and ErbB expression/activity in skeletal muscle were tested again. The 16 week HF/HS diet induced obesity, but did not significantly affect the NRG1/ErbB signalling pathway in rat skeletal muscle. Conversely, after the switch to a well-balanced diet, NRG1 cleavage ratio and ErbB4 amount were increased. Chronic exercise training also promoted NRG1 cleavage, resulting in increased ErbB4 phosphorylation. This result was associated with increased protein expression and phosphorylation ratio of the metalloprotease ADAM17, which is involved in NRG1 shedding. Similarly, in vitro stretch-induced activation of ADAM17 in rat myoblasts induced NRG1 cleavage and ErbB4 activation. These results show that low intensity endurance training and well-balanced diet activate the NRG1-ErbB4 pathway, possibly via the metalloprotease ADAM17, in skeletal muscle of diet-induced obese rats.
Collapse
Affiliation(s)
- Gaël Ennequin
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Nathalie Boisseau
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Kevin Caillaud
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Vivien Chavanelle
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Maude Gerbaix
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Lore Metz
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Monique Etienne
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Stéphane Walrand
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France.,INRA, UMR 1019, Clermont-Ferrand, F-63001, France.,University Clermont 1, UFR Médecine, Clermont-Ferrand, F-63001, France
| | - Aurélie Masgrau
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France.,INRA, UMR 1019, Clermont-Ferrand, F-63001, France.,University Clermont 1, UFR Médecine, Clermont-Ferrand, F-63001, France
| | - Christelle Guillet
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France.,INRA, UMR 1019, Clermont-Ferrand, F-63001, France.,University Clermont 1, UFR Médecine, Clermont-Ferrand, F-63001, France
| | - Daniel Courteix
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| | - Airu Niu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Centre, Houston, TX, 77030, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Centre, Houston, TX, 77030, USA
| | - Fréderic Capel
- CRNH-Auvergne, Clermont-Ferrand, F-63001, France.,INRA, UMR 1019, Clermont-Ferrand, F-63001, France.,University Clermont 1, UFR Médecine, Clermont-Ferrand, F-63001, France
| | - Pascal Sirvent
- Université Clermont Auvergne, Université Blaise Pascal, EA 3533, Laboratoire des Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P), BP 80026, F-63171, Aubière Cedex, France.,CRNH-Auvergne, Clermont-Ferrand, F-63001, France
| |
Collapse
|