51
|
Du ZD, Yu S, Qi Y, Qu TF, He L, Wei W, Liu K, Gong SS. NADPH oxidase inhibitor apocynin decreases mitochondrial dysfunction and apoptosis in the ventral cochlear nucleus of D-galactose-induced aging model in rats. Neurochem Int 2018; 124:31-40. [PMID: 30578839 DOI: 10.1016/j.neuint.2018.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/01/2018] [Accepted: 12/17/2018] [Indexed: 01/17/2023]
Abstract
Presbycusis has become a common sensory deficit in humans. Oxidative damage to mitochondrial DNA and mitochondrial dysfunction is strongly associated with the aging of the auditory system. A previous study established a mimetic rat model of aging using D-galactose (D-gal) and first reported that NADPH oxidase-dependent mitochondrial oxidative damage and apoptosis in the ventral cochlear nucleus (VCN) might contribute to D-gal-induced central presbycusis. In this study, we investigated the effects of apocynin, an NADPH oxidase inhibitor, on mitochondrial dysfunction and mitochondria-dependent apoptosis in the VCN of D-gal-induced aging model in rats. Our data showed that apocynin decreased NADPH oxidase activity, H2O2 levels, mitochondrial DNA common deletion, and 8-hydroxy-2-deoxyguanosine (8-OHdG) expression and increased total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px) activity in the VCN of D-gal-induced aging model in rats. Moreover, apocynin also decreased the protein levels of phospho-p47phox (p-p47phox), tumor necrosis factor alpha (TNFα), and uncoupling protein 2 (UCP2) in the VCN of D-gal-induced aging model in rats. Meanwhile, apocynin alleviated mitochondrial ultrastructure damage and enhanced ATP production and mitochondrial membrane potential (MMP) levels in the VCN of D-gal-induced aging model in rats. Furthermore, apocynin inhibited cytochrome c (Cyt c) translocation from mitochondria to the cytoplasm and suppressed caspase 3-dependent apoptosis in the VCN of D-gal-induced aging model in rats. Consequently, our findings suggest that neuronal survival promoted by an NADPH oxidase inhibitor is a potentially effective method to enhance the resistance of neurons to central presbycusis.
Collapse
Affiliation(s)
- Zheng-De Du
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Shukui Yu
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Yue Qi
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Teng-Fei Qu
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Lu He
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Wei Wei
- Department of Otology, Shengjing Hospital, China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Ke Liu
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China.
| | - Shu-Sheng Gong
- Department of Otorhinolaryngology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
52
|
Abstract
The hypothalamus is the central neural site governing food intake and energy expenditure. During the past 25 years, understanding of the hypothalamic cell types, hormones, and circuitry involved in the regulation of energy metabolism has dramatically increased. It is now well established that the adipocyte-derived hormone, leptin, acts upon two distinct groups of hypothalamic neurons that comprise opposing arms of the central melanocortin system. These two cell populations are anorexigenic neurons expressing proopiomelanocortin (POMC) and orexigenic neurons that express agouti-related peptide (AGRP). Several important studies have demonstrated that reactive oxygen species and endoplasmic reticulum stress significantly impact these hypothalamic neuronal populations that regulate global energy metabolism. Reactive oxygen species and redox homeostasis are influenced by selenoproteins, an essential class of proteins that incorporate selenium co-translationally in the form of the 21st amino acid, selenocysteine. Levels of these proteins are regulated by dietary selenium intake and they are widely expressed in the brain. Of additional relevance, selenium supplementation has been linked to metabolic alterations in both animal and human studies. Recent evidence also indicates that hypothalamic selenoproteins are significant modulators of energy metabolism in both neurons and tanycytes, a population of glial-like cells lining the floor of the 3rd ventricle within the hypothalamus. This review article will summarize current understanding of the regulatory influence of redox status on hypothalamic nutrient sensing and highlight recent work revealing the importance of selenoproteins in the hypothalamus.
Collapse
Affiliation(s)
- Ting Gong
- Department of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI 96813, USA
| | - Daniel J Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Marla J Berry
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Matthew W Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
53
|
Bai Y, Bai Y, Wang S, Wu F, Wang DH, Chen J, Huang J, Li H, Li Y, Wu S, Wang Y, Yang Y. Targeted upregulation of uncoupling protein 2 within the basal ganglia output structure ameliorates dyskinesia after severe liver failure. Free Radic Biol Med 2018; 124:40-50. [PMID: 29857139 DOI: 10.1016/j.freeradbiomed.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 02/06/2023]
Abstract
Impaired motor function, due to the dysfunction of the basal ganglia, is the most common syndrome of hepatic encephalopathy (HE), and its etiology remains poorly understood. Neural oxidative stress is shown to be the major cellular defects contributing to HE pathogenesis. Mitochondrial uncoupling protein 2 (UCP2) has been implicated in neuroprotection in several neurological disorders. We explored the neuroprotective role of UCP2 within the substantia nigra pars reticulate (SNr), the output structure of the basal ganglia, in HE. The toxin thioacetamide (TAA) induced HE mice showed hypolocomotion, which was associated with decreased ATP levels and loss of antioxidant substances SOD and GSH within the SNr. Stable overexpression of UCP2 via AAV-UCP2 under the control of the UCP2 promoter in bilateral SNr preserved local ATP level, increased antioxidant substances, and ameliorated locomotion defects after severe liver failure. Contrary to UCP2 overexpression, targeted knockdown of UCP2 within bilateral SNr via AAV-UCP2 shRNA exacerbated the impaired mitochondrial dysfunction and hypokinesia in HE mice. The modulatory effects of UCP2 was due to mediation of K+-Cl- cotransporter-2 (KCC2) expression on GABAergic neurons of SNr. Taken together, our results demonstrate that UCP2 exerts a neural protective role at the subcortical level by increasing the resistance of neurons to oxidative stress, which may offer a novel therapeutic target for the treatment of motor dysfunction diseases.
Collapse
Affiliation(s)
- Yunhu Bai
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Yang Bai
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Shengming Wang
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Feifei Wu
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Dong Hui Wang
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Jing Chen
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Jing Huang
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Hui Li
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Yunqing Li
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Shengxi Wu
- Department of Neurobiology, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China
| | - Yayun Wang
- Department of Anatomy and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200000, China.
| | - Yanling Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
54
|
Corona-Herrera GA, Arranz SE, Martínez-Palacios CA, Navarrete-Ramírez P, Toledo-Cuevas EM, Valdez-Alarcón JJ, Martínez-Chávez CC. Experimental evidence of masculinization by continuous illumination in a temperature sex determination teleost (Atherinopsidae) model: is oxidative stress involved? JOURNAL OF FISH BIOLOGY 2018; 93:229-237. [PMID: 29931822 DOI: 10.1111/jfb.13651] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
The present study evaluates the influence of continuous light on phenotypic sex ratios in Chirostoma estor, a temperature sex determination animal model. Relative gene expression levels of 5 day old larvae were performed on two early gonad differentiation genes (sox9 and foxl2), two stress axis activation genes (gcr1 and crf) and four reactive oxygen species (ROS) antagonist effector genes (sod2, ucp2, gsr and cat). Two light treatments were applied from fertilization; control (12L:12D) simulated natural photoperiod and a continuous illumination photoperiod. By the end of the trial (12 weeks after hatching), differentiated and normal gonads were clearly identifiable in both treatments by histological observations. Regarding sex ratio, 73% of phenotypic males were found in continuous illumination compared with 40% in controls. Consistently, the sox9 gene (involved in early testis differentiation) showed an over expression in 64% of the individual larvae analysed compared with foxl2 (ovarian differentiation) suggesting a masculinization tendency in continuous illumination. On the other hand, only 36% of individuals showed the same tendency in the control treatment consistent with phenotypic sex ratios found under normal culture conditions. Relative gene expression results did not show significant difference in sod2, ucp2 and gcr1 levels, but cat, gsr and crf showed significantly higher expression levels in the continuous illumination treatment suggesting that both, the stress axis and ROS response mechanisms were activated at this time. This study suggests, a link between continuous light, oxidative stress and environmental sex determination in vertebrates. However, further research is necessary to describe this possible upstream mechanism that may drive some aspects of sexual plasticity in vertebrates.
Collapse
Affiliation(s)
- Guillermo A Corona-Herrera
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Silvia E Arranz
- Laboratorio de Biotecnología Acuática, Universidad Nacional de Rosario, Santa Fe, Argentina
| | - Carlos A Martínez-Palacios
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Pamela Navarrete-Ramírez
- CONACyT-Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Elva M Toledo-Cuevas
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Juan J Valdez-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo. Km. 9.5 carretera Morelia-Zinapecuaro, Tarimbaro, Mexico
| | - Carlos C Martínez-Chávez
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
55
|
Hasan-Olive MM, Lauritzen KH, Ali M, Rasmussen LJ, Storm-Mathisen J, Bergersen LH. A Ketogenic Diet Improves Mitochondrial Biogenesis and Bioenergetics via the PGC1α-SIRT3-UCP2 Axis. Neurochem Res 2018; 44:22-37. [PMID: 30027365 DOI: 10.1007/s11064-018-2588-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 11/30/2022]
Abstract
A ketogenic diet (KD; high-fat, low-carbohydrate) can benefit refractory epilepsy, but underlying mechanisms are unknown. We used mice inducibly expressing a mutated form of the mitochondrial DNA repair enzyme UNG1 (mutUNG1) to cause progressive mitochondrial dysfunction selectively in forebrain neurons. We examined the levels of mRNAs and proteins crucial for mitochondrial biogenesis and dynamics. We show that hippocampal pyramidal neurons in mutUNG1 mice, as well as cultured rat hippocampal neurons and human fibroblasts with H2O2 induced oxidative stress, improve markers of mitochondrial biogenesis, dynamics and function when fed on a KD, and when exposed to the ketone body β-hydroxybutyrate, respectively, by upregulating PGC1α, SIRT3 and UCP2, and (in cultured cells) increasing the oxygen consumption rate (OCR) and the NAD+/NADH ratio. The mitochondrial level of UCP2 was significantly higher in the perikarya and axon terminals of hippocampus CA1 pyramidal neurons in KD treated mutUNG1 mice compared with mutUNG1 mice fed a standard diet. The β-hydroxybutyrate receptor GPR109a (HCAR2), but not the structurally closely related lactate receptor GPR81 (HCAR1), was upregulated in mutUNG1 mice on a KD, suggesting a selective influence of KD on ketone body receptor mechanisms. We conclude that progressive mitochondrial dysfunction in mutUNG1 expressing mice causes oxidative stress, and that exposure of animals to KD, or of cells to ketone body in vitro, elicits compensatory mechanisms acting to augment mitochondrial mass and bioenergetics via the PGC1α-SIRT3-UCP2 axis (The compensatory processes are overwhelmed in the mutUNG1 mice by all the newly formed mitochondria being dysfunctional).
Collapse
Affiliation(s)
- Md Mahdi Hasan-Olive
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway. .,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway. .,Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Knut H Lauritzen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mohammad Ali
- Department of Biochemistry, Sir Salimullah Medical College & Mitford Hospital, Dhaka, Bangladesh
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jon Storm-Mathisen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Linda H Bergersen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway. .,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway. .,Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
56
|
Sosa-Gutiérrez JA, Valdéz-Solana MA, Forbes-Hernández TY, Avitia-Domínguez CI, Garcia-Vargas GG, Salas-Pacheco JM, Flores-Herrera O, Téllez-Valencia A, Battino M, Sierra-Campos E. Effects of Moringa oleifera Leaves Extract on High Glucose-Induced Metabolic Changes in HepG2 Cells. BIOLOGY 2018; 7:biology7030037. [PMID: 29949946 PMCID: PMC6164683 DOI: 10.3390/biology7030037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/08/2018] [Accepted: 06/25/2018] [Indexed: 12/29/2022]
Abstract
Mitochondrial dysfunction is a hallmark of diabetes, but the metabolic alterations during early stages of the disease remain unknown. The ability of liver cells to rearrange their metabolism plays an important role in compensating the energy shortage and may provide cell survival. Moringa oleifera leaves have been studied for its health properties against diabetes, insulin resistance, and non-alcoholic liver disease. We postulated that M. oleifera executes a protective function on mitochondrial functionality in HepG2 treated with high glucose. We evaluated the effect of high glucose treatment on the mitochondrial function of HepG2 cells using a Seahorse extracellular flux analyzer (Agilent, Santa Clara, CA, USA), blue native polyacrylamide gel electrophoresis (BN-PAGE), and western blot analysis. For assessment of mitochondrial abnormalities, we measured the activity of mitochondrial Complex I and IV as well as uncoupling protein 2, and sirtuin 3 protein contents. Our results demonstrate that, under conditions mimicking the hyperglycemia, Complex I activity, UCP2, Complex III and IV subunits content, supercomplex formation, and acetylation levels are modified with respect to the control condition. However, basal oxygen consumption rate was not affected and mitochondrial reactive oxygen species production remained unchanged in all groups. Treatment of HepG2 cells with M. oleifera extract significantly increased both protein content and mitochondrial complexes activities. Nonetheless, control cells’ respiratory control ratio (RCR) was 4.37 compared to high glucose treated cells’ RCR of 15.3, and glucose plus M. oleifera treated cells’ RCR of 5.2, this indicates high-quality mitochondria and efficient oxidative phosphorylation coupling. Additionally, the state app was not altered between different treatments, suggesting no alteration in respiratory fluxes. These findings enhance understanding of the actions of M. oleifera and suggest that the known antidiabetic property of this plant, at least in part, is mediated through modulating the mitochondrial respiratory chain.
Collapse
Affiliation(s)
- Jorge A Sosa-Gutiérrez
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Campus Gómez Palacio, Avenida Artículo 123 S/N, Fracc, Filadelfia, 35010 Gómez Palacio, Mexico.
| | - Mónica A Valdéz-Solana
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Campus Gómez Palacio, Avenida Artículo 123 S/N, Fracc, Filadelfia, 35010 Gómez Palacio, Mexico.
| | - Tamara Y Forbes-Hernández
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO)-Sez. Biochimica, Facoltà di Medicina, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - Claudia I Avitia-Domínguez
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango Campus Durango, Avenida Universidad y Fanny Anitúa S/N, 34000 Durango, Mexico.
| | - Gonzalo G Garcia-Vargas
- Facultad de Ciencias de la Salud, Universidad Juárez del Estado de Durango Campus Gómez Palacio, Calzada Palmas 1, Colonia Revolución, 35050 Gómez Palacio, Mexico.
| | - José M Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Avenida Universidad S/N, 34000 Durango, Mexico.
| | - Oscar Flores-Herrera
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico.
| | - Alfredo Téllez-Valencia
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango Campus Durango, Avenida Universidad y Fanny Anitúa S/N, 34000 Durango, Mexico.
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO)-Sez. Biochimica, Facoltà di Medicina, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - Erick Sierra-Campos
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Campus Gómez Palacio, Avenida Artículo 123 S/N, Fracc, Filadelfia, 35010 Gómez Palacio, Mexico.
| |
Collapse
|
57
|
Rajamani U, Gross AR, Hjelm BE, Sequeira A, Vawter MP, Tang J, Gangalapudi V, Wang Y, Andres AM, Gottlieb RA, Sareen D. Super-Obese Patient-Derived iPSC Hypothalamic Neurons Exhibit Obesogenic Signatures and Hormone Responses. Cell Stem Cell 2018; 22:698-712.e9. [PMID: 29681516 PMCID: PMC6398951 DOI: 10.1016/j.stem.2018.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 02/06/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022]
Abstract
The hypothalamus contains neurons that integrate hunger and satiety endocrine signals from the periphery and are implicated in the pathophysiology of obesity. The limited availability of human hypothalamic neurons hampers our understanding of obesity disease mechanisms. To address this, we generated human induced pluripotent stem cells (hiPSCs) from multiple normal body mass index (BMI; BMI ≤ 25) subjects and super-obese (OBS) donors (BMI ≥ 50) with polygenic coding variants in obesity-associated genes. We developed a method to reliably differentiate hiPSCs into hypothalamic-like neurons (iHTNs) capable of secreting orexigenic and anorexigenic neuropeptides. Transcriptomic profiling revealed that, although iHTNs maintain a fetal identity, they respond appropriately to metabolic hormones ghrelin and leptin. Notably, OBS iHTNs retained disease signatures and phenotypes of high BMI, exhibiting dysregulated respiratory function, ghrelin-leptin signaling, axonal guidance, glutamate receptors, and endoplasmic reticulum (ER) stress pathways. Thus, human iHTNs provide a powerful platform to study obesity and gene-environment interactions.
Collapse
Affiliation(s)
- Uthra Rajamani
- Board of Governors, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew R Gross
- Board of Governors, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Brooke E Hjelm
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA 92868, USA
| | - Adolfo Sequeira
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA 92868, USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA 92868, USA
| | - Jie Tang
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Yizhou Wang
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Allen M Andres
- Metabolism and Mitochondrial Research Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Roberta A Gottlieb
- Metabolism and Mitochondrial Research Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dhruv Sareen
- Board of Governors, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; iPSC Core, The David Janet Polak Foundation Stem Cell Core Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
58
|
Yang H, Sun R, Ma N, Liu Q, Sun X, Zi P, Wang J, Chao K, Yu L. Inhibition of nuclear factor-κB signal by pyrrolidine dithiocarbamate alleviates lipopolysaccharide-induced acute lung injury. Oncotarget 2018; 8:47296-47304. [PMID: 28521300 PMCID: PMC5564565 DOI: 10.18632/oncotarget.17624] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022] Open
Abstract
This study mainly studied the effect of inhibition of nuclear factor-κB (NF-κB) signal by pyrrolidine dithiocarbamate (PDTC) on lipopolysaccharide (LPS)-induced inflammatory response, oxidative stress, and mitochondrial dysfunction in a murine acute lung injury model. The results showed that LPS exposure activated NF-κB and its upstream proteins and caused lung inflammation, oxidative stress, and mitochondrial dysfunction in mice. While inhibition of NF-κB by PDTC adminstration alleviated LPS-induced generation of lymphocytes, IL-1β, and TNF-α. Malondialdehyde, a common oxidative product, was markedly reduced after PDTC treatment in LPS-challenged mice. Furthermore, PDTC alleviated LPS-induced mitochondrial dysfunction via improving ATP synthesis and uncoupling protein 2 expression. In conclusion, inhibition of NF-κB by PDTC alleviated LPS-induced acute lung injury via maintaining inflammatory status, oxidative balance, and mitochondrial function in mice.
Collapse
Affiliation(s)
- Hongfu Yang
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Rongqing Sun
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Ning Ma
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Qilong Liu
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Xiaoge Sun
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Panpan Zi
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Junsheng Wang
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Ke Chao
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Lei Yu
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| |
Collapse
|
59
|
Wang M, Li G, Yang Z, Wang L, Zhang L, Wang T, Zhang Y, Zhang S, Han Y, Jia L. Uncoupling protein 2 downregulation by hypoxia through repression of peroxisome proliferator-activated receptor γ promotes chemoresistance of non-small cell lung cancer. Oncotarget 2018; 8:8083-8094. [PMID: 28042952 PMCID: PMC5352384 DOI: 10.18632/oncotarget.14097] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022] Open
Abstract
Hypoxic microenvironment is critically involved in the response of non-small cell lung cancer (NSCLC) to chemotherapy, the mechanisms of which remain largely unknown. Here, we found that NSCLC patients exhibited increased chemotherapeutic resistance when complicated by chronic obstructive pulmonary disease (COPD), a critical cause of chronic hypoxemia. The downregulation of uncoupling protein 2 (UCP2), which is attributed to hypoxia-inducible factor 1 (HIF-1)-mediated suppression of the transcriptional factor peroxisome proliferator-activated receptor γ (PPARγ), was involved in NSCLC chemoresistance, and predicted a poor survival rate of patients receiving routine chemotherapy. UCP2 suppression induced reactive oxygen species production and upregulation of the ABC transporter protein ABCG2, which leads to chemoresistance by promoting drug efflux. UCP2 downregulation also altered metabolic rates as shown by elevated glucose uptake and reduced oxygen consumption. These data suggest that UCP2 is a key mediator of hypoxia-triggered chemoresistance of NSCLCs, which can be potentially targeted in clinical treatment of chemo-refractory NSCLCs.
Collapse
Affiliation(s)
- Mingxing Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Guoyin Li
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Zhiwei Yang
- Department of Applied Physics, Xi'an Jiaotong University, Xi'an, China
| | - Lei Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Applied Physics, Xi'an Jiaotong University, Xi'an, China
| | - Ting Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Yimeng Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Shengli Zhang
- Department of Applied Physics, Xi'an Jiaotong University, Xi'an, China
| | - Yong Han
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
60
|
mRNA m 6A plays opposite role in regulating UCP2 and PNPLA2 protein expression in adipocytes. Int J Obes (Lond) 2018; 42:1912-1924. [PMID: 29487348 DOI: 10.1038/s41366-018-0027-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/19/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND/OBJECTIVE N6-methyladenosine (m6A) modification of mRNA plays an important role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. SUBJECTS/METHODS Using Jinhua and Landrace pigs as fat and lean models, we presented a comprehensive transcriptome-wide m6A profiling in adipose tissues from these two pig breeds. Two differentially methylated genes were selected to explore the mechanisms of m6A-mediated regulation of gene function. RESULTS The ratio of m6A/A in the layer of backfat (LB) was significantly higher in Landrace than that in Jinhua. Transcriptome-wide m6A profiling revealed that m6A modification on mRNA occurs in the conserved sequence motif of RRACH and that the pig transcriptome contains 0.53-0.91 peak per actively expressed transcript. The relative density of m6A peaks in the 3'UTR were higher than in 5'UTR. Genes with common m6A peaks from both Landrace (L-LB) and Jinhua (J-LB) were enriched in RNA splicing and cellular lipid metabolic process. The unique m6A peak genes (UMGs) from L-LB were mainly enriched in the extracellular matrix (ECM) and collagen catabolic process, whereas the UMGs from J-LB are mainly involved in RNA splicing, etc. Lipid metabolism processes were not significantly enriched in the UMGs from L-LB or J-LB. Uncoupling protein-2 (UCP2) and patatin-like phospholipase domain containing 2 (PNPLA2) were two of the UMGs in L-LB. Synonymous mutations (MUT) were conducted to reduce m6A level of UCP2 and PNPLA2 mRNAs. Adipogenesis test showed that UCP2-MUT further inhibited adipogenesis, while PNPLA2-MUT promoted lipid accumulation compared with UCP2-WT and PNPLA2-WT, respectively. Further study showed m6A negatively mediates UCP2 protein expression and positively mediates PNPLA2 protein expression. m6A modification affects the translation of PNPLA2 most likely through YTHDF1, whereas UCP2 is likely neither the target of YTHDF2 nor the target of YTHDF1. CONCLUSION Our data demonstrated a conserved and yet dynamically regulated m6A methylome in pig transcriptomes and provided an important resource for studying the function of m6A epitranscriptomic modification in obesity development.
Collapse
|
61
|
Quinhoneiro DCG, Nicoletti CF, Pinhel MAS, Noronha NY, Braga CBM, Oliveira BAP, Cortes-Oliveira C, Oliveira WP, Salgado Junior W, Marchini JS, Nonino CB. Green tea supplementation upregulates uncoupling protein 3 expression in severe obese women adipose tissue but does not promote weight loss. Int J Food Sci Nutr 2018; 69:995-1002. [PMID: 29482377 DOI: 10.1080/09637486.2018.1442819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This study aims (i) to verify expression of the UCPs, PLIN1, PPARG2, and ADRB3 genes in the abdominal subcutaneous adipose tissue of obese women at baseline and after 8 weeks of supplementation with decaffeinated green tea extract, and (ii) to associate findings with clinical parameters. This is a longitudinal study during which 11 women with obesity grade III were submitted to supplementation with 450 mg of (-)-epigallocatechin gallate (EGCG) (intervention group); the control group consisted of 10 eutrophic women. Anthropometric parameters [weight, height, and body mass index (BMI)], resting metabolic rate (RMR, measured by indirect calorimetry), and gene expression (measured by real-time PCR, RT-qPCR) were determined before and after supplementation. After 8 weeks, clinical parameters and UCP1, PLIN1, PPARG2, and ADRB3 expression remained unaltered in the intervention group (p > .05). Genetic analysis also showed that the UCP3 gene was upregulated (p = .026), but its upregulation did not promote weight loss.
Collapse
Affiliation(s)
| | - Carolina Ferreira Nicoletti
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Marcela Augusta Souza Pinhel
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Natália Yumi Noronha
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Camila Bitu Moreno Braga
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | | | - Cristiana Cortes-Oliveira
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Wanderley Pereira Oliveira
- b Department of Pharmaceutical Sciences School of Pharmaceutical Sciences of Ribeirão Preto , University of Sao Paulo - USP , Ribeirao Preto , Brazil
| | - Wilson Salgado Junior
- c Department of Surgery and Anatomy , Ribeirao Preto Medical School University of São Paulo , Ribeirão Preto , Brazil
| | - Júlio Sergio Marchini
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Carla Barbosa Nonino
- a Department of Internal Medicine , Ribeirao Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| |
Collapse
|
62
|
Yang C, Zhao D, Liu G, Zheng H, Yang H, Yang S, Yang P. Atorvastatin Attenuates Metabolic Remodeling in Ischemic Myocardium through the Downregulation of UCP2 Expression. Int J Med Sci 2018; 15:517-527. [PMID: 29559841 PMCID: PMC5859775 DOI: 10.7150/ijms.22454] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Uncoupling protein 2 (UCP2) is primarily expressed in the myocardium and is closely related to myocardial ischemia/reperfusion injury and myocardial metabolism. To explore the effects and the mechanisms of UCP2 on atorvastatin-mediated myocardium protection, the rat model of myocardial ischemia was established by ligation of the left anterior descending coronary arteries (LADs). The rats were divided into the sham operation (SO) group, myocardial infarction (MI) group and MI-atorvastatin group. The study that atorvastatin reduced myocardial remodeling and improved the disturbed myocardial energy metabolism after MI. Furthermore, the mechanisms of myocardial metabolic remodeling affected by atorvastatin were explored. The atorvastatin group showed a significantly decreased expression of UCP2 mRNA and protein. Furthermore, the primary rat cardiomyocytes were cultured and treated with angiotensin II (Ang II) to induce cardiomyocyte hypertrophy. The results showed that in the atorvastatin group, the surface area of the cardiomyocytes, the total protein content per unit of cells, and the expression of the UCP2 protein were significantly decreased. These data suggested that atorvastatin significantly attenuated the myocardial remodeling by downregulating the expression of UCP2 that was found to improve the myocardial energy metabolism, inhibit myocardial hypertrophy, and eventually reduce myocardial remodeling.
Collapse
Affiliation(s)
- Chunyan Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Dongming Zhao
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.,Department of Cardiology, the affiliated hospital of Beihua University, Jilin, China
| | - Guohui Liu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Haikuo Zheng
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Hongliang Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Sibao Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| |
Collapse
|
63
|
Yu C, Xi L, Chen J, Jiang Q, Yi H, Wang Y, Wang X. PAM, OLA, and LNA are Differentially Taken Up and Trafficked Via Different Metabolic Pathways in Porcine Adipocytes. Lipids 2017; 52:929-938. [PMID: 29058170 DOI: 10.1007/s11745-017-4302-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Dietary fatty acids have different effects on fat deposition in pigs. To clarify the underlying mechanisms of this difference, we compared the metabolism of palmitic (PAM, saturated), oleic (OLA, monounsaturated) and linoleic acid (LNA, polyunsaturated) in porcine adipocytes treated with 100 μM PAM, OLA or LNA. We observed that the adipocytes incubated with LNA accumulated more lipids compared with those treated with PAM and OLA. We then probed the metabolism of these fatty acids in porcine adipocytes by using isotope-labelled fatty acids. The results showed that 42% of the [1-14C] LNA, 34% of the [1-14C] PAM and 28% of the [1-14C] OLA were recovered in the cellular lipids. The gene expression analyses showed that LNA significantly increased the expression of adipogenesis- and oxidation-related genes including PPARγ, C/EBPα, ap2 and NRF1. In addition, the cells incubated with LNA showed a decreased Ser112 phosphorylation in PPARγ compared to those incubated with PAM and OLA. Furthermore, when PPARγ Ser112 phosphorylation was inhibited, no significant difference in the triacylglycerol contents in the adipocytes was observed. These results showed the dietary fatty acids had different metabolism pathways in porcine adipocytes, and LNA significantly promoted lipid accumulation, probably by regulating PPARγ phosphorylation in adipocytes.
Collapse
Affiliation(s)
- Caihua Yu
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Lingling Xi
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Jin Chen
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Qin Jiang
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Hongbo Yi
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Xinxia Wang
- Key Laboratory of Animal Nutrition and Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China.
| |
Collapse
|
64
|
Abstract
The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.
Collapse
Affiliation(s)
- Chitoku Toda
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anna Santoro
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jung Dae Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520.,Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
65
|
Jourdan T, Nicoloro SM, Zhou Z, Shen Y, Liu J, Coffey NJ, Cinar R, Godlewski G, Gao B, Aouadi M, Czech MP, Kunos G. Decreasing CB 1 receptor signaling in Kupffer cells improves insulin sensitivity in obese mice. Mol Metab 2017; 6:1517-1528. [PMID: 29107297 PMCID: PMC5681272 DOI: 10.1016/j.molmet.2017.08.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/15/2022] Open
Abstract
Objective Obesity-induced accumulation of ectopic fat in the liver is thought to contribute to the development of insulin resistance, and increased activity of hepatic CB1R has been shown to promote both processes. However, lipid accumulation in liver can be experimentally dissociated from insulin resistance under certain conditions, suggesting the involvement of additional mechanisms. Obesity is also associated with pro-inflammatory changes which, in turn, can promote insulin resistance. Kupffer cells (KCs), the liver's resident macrophages, are the major source of pro-inflammatory cytokines in the liver, such as TNF-α, which has been shown to inhibit insulin signaling in multiple cell types, including hepatocytes. Here, we sought to identify the role of CB1R in KCs in obesity-induced hepatic insulin resistance. Methods We used intravenously administered β-D-glucan-encapsulated siRNA to knock-down CB1R gene expression selectively in KCs. Results We demonstrate that a robust knock-down of the expression of Cnr1, the gene encoding CB1R, results in improved glucose tolerance and insulin sensitivity in diet-induced obese mice, without affecting hepatic lipid content or body weight. Moreover, Cnr1 knock-down in KCs was associated with a shift from pro-inflammatory M1 to anti-inflammatory M2 cytokine profile and improved insulin signaling as reflected by increased insulin-induced Akt phosphorylation. Conclusion These findings suggest that CB1R expressed in KCs plays a critical role in obesity-related hepatic insulin resistance via a pro-inflammatory mechanism. CB1R signaling promotes hepatic insulin resistance by promoting hepatic steatosis and hepatic inflammation. CB1R knock-down in liver macrophages (Kupffer cells, KCs) improves global insulin resistance and glucose homeostasis. CB1R expressed in KCs play a critical role in hepatic insulin resistance independent of ectopic fat in the liver or adipose tissue inflammation.
Collapse
Affiliation(s)
- Tony Jourdan
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA.
| | - Sarah M Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Zhou Zhou
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Yuefei Shen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jie Liu
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Nathan J Coffey
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Grzegorz Godlewski
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA
| | - Myriam Aouadi
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD 20852, USA.
| |
Collapse
|
66
|
Michael NJ, Simonds SE, van den Top M, Cowley MA, Spanswick D. Mitochondrial uncoupling in the melanocortin system differentially regulates NPY and POMC neurons to promote weight-loss. Mol Metab 2017; 6:1103-1112. [PMID: 29031712 PMCID: PMC5641603 DOI: 10.1016/j.molmet.2017.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Objective The mitochondrial uncoupling agent 2,4-dinitrophenol (DNP), historically used as a treatment for obesity, is known to cross the blood-brain-barrier, but its effects on central neural circuits controlling body weight are largely unknown. As hypothalamic melanocortin neuropeptide Y/agouti-related protein (NPY/AgRP) and pro-opiomelanocortin (POMC) neurons represent key central regulators of food intake and energy expenditure we investigated the effects of DNP on these neurons, food intake and energy expenditure. Method C57BL/6 and melanocortin-4 receptor (MC4R) knock-out mice were administered DNP intracerebroventricularly (ICV) and the metabolic changes were characterized. The specific role of NPY and POMC neurons and the ionic mechanisms mediating the effects of uncoupling were examined with in vitro electrophysiology performed on NPY hrGFP or POMC eGFP mice. Results Here we show DNP-induced differential effects on melanocortin neurons including inhibiting orexigenic NPY and activating anorexigenic POMC neurons through independent ionic mechanisms coupled to mitochondrial function, consistent with an anorexigenic central effect. Central administration of DNP induced weight-loss, increased BAT thermogenesis and browning of white adipose tissue, and decreased food intake, effects that were absent in MC4R knock-out mice and blocked by the MC4R antagonist, AgRP. Conclusion These data show a novel central anti-obesity mechanism of action of DNP and highlight the potential for selective melanocortin mitochondrial uncoupling to target metabolic disorders. Mitochondrial uncoupling of the melanocortin system with DNP induced weight-loss. DNP inhibited NPY neurones via activation of ATP-sensitive potassium channels. DNP activated POMC neurones via block of inwardly rectifying potassium channels. Central DNP reduced food intake and increased WAT browning and BAT thermogenesis.
Collapse
Affiliation(s)
- Natalie Jane Michael
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | - Stephanie Elise Simonds
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | | | - Michael Alexander Cowley
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Australia(5).
| | - David Spanswick
- Neuroscience Program, Biomedicine Discovery Institute, Monash University, Australia(5); Neurosolutions, Coventry, P.O. 3517, UK; Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
67
|
Cortes de Oliveira C, Nicoletti CF, Pinhel MADS, de Oliveira BAP, Quinhoneiro DCG, Noronha NY, Fassini PG, Marchini JS, da Silva Júnior WA, Salgado Júnior W, Nonino CB. Influence of expression of UCP3, PLIN1 and PPARG2 on the oxidation of substrates after hypocaloric dietary intervention. Clin Nutr 2017; 37:1383-1388. [PMID: 28651828 DOI: 10.1016/j.clnu.2017.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS In addition to environmental and psychosocial factors, it is known that genetic factors can also influence the regulation of energy metabolism, body composition and determination of excess weight. The objective of this study was to evaluate the influence of UCP3, PLIN1 and PPARG2 genes on the substrates oxidation in women with grade III obesity after hypocaloric dietary intervention. SUBJECTS/METHODS This is a longitudinal study with 21 women, divided into two groups: Intervention Group (G1): 11 obese women (Body Mass Index (BMI) ≥40 kg/m2), and Control Group (G2): 10 eutrophic women (BMI between 18.5 kg/m2 and 24.9 kg/m2). Weight (kg), height (m), BMI (kg/m2), substrate oxidation (by Indirect Calorimetry) and abdominal subcutaneous adipose tissue were collected before and after the intervention. For the dietary intervention, the patients were hospitalized for 6 weeks receiving 1200 kcal/day. RESULTS There was a significant weight loss (8.4 ± 4.3 kg - 5.2 ± 1.8%) and reduction of UCP3 expression after hypocaloric dietary intervention. There was a positive correlation between carbohydrate oxidation and UCP3 (r = 0.609; p = 0.04), PLIN1 (r = 0.882; p = 0.00) and PPARG2 (r = 0.791; p = 0.00) expression before dietary intervention and with UCP3 (r = 0.682; p = 0.02) and PLIN1 (r = 0.745; p = 0.00) genes after 6 weeks of intervention. There was a negative correlation between lipid oxidation and PLIN1 (r = -0.755; p = 0.00) and PPARG2 (r = 0.664; p = 0.02) expression before dietary intervention and negative correlation with PLIN1 (r = 0.730; p = 0.02) expression after 6 weeks of hypocaloric diet. CONCLUSION Hypocaloric diet reduces UCP3 expression in individuals with obesity and the UCP3, PLIN1 and PPARG2 expression correlate positively with carbohydrate oxidation and negatively with lipid oxidation.
Collapse
Affiliation(s)
- Cristiana Cortes de Oliveira
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Carolina Ferreira Nicoletti
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Marcela Augusta de Souza Pinhel
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | | | | | - Natália Yumi Noronha
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Priscila Giacomo Fassini
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Júlio Sérgio Marchini
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Wilson Araújo da Silva Júnior
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Wilson Salgado Júnior
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Carla Barbosa Nonino
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
68
|
Glucose-Insulin-Potassium Alleviates Intestinal Mucosal Barrier Injuries Involving Decreased Expression of Uncoupling Protein 2 and NLR Family-Pyrin Domain-Containing 3 Inflammasome in Polymicrobial Sepsis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4702067. [PMID: 28428961 PMCID: PMC5385915 DOI: 10.1155/2017/4702067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/05/2017] [Indexed: 12/18/2022]
Abstract
Uncoupling protein 2 (UCP2) may be critical for intestinal barrier function which may play a key role in the development of sepsis, and insulin has been reported to have anti-inflammatory effects. Male Sprague-Dawley rats were randomly allocated into five groups: control group, cecal ligation and puncture (CLP) group, sham surgery group, CLP plus glucose-insulin-potassium (GIK) group, and CLP plus glucose and potassium (GK) group. Ileum tissues were collected at 24 h after surgery. Histological and cytokine analyses, intestinal permeability tests, and western blots of intestinal epithelial tight junction component proteins and UCP2 were performed. Compared with CLP group, the CLP + GIK group had milder histological damage, lower levels of cytokines in the serum and ileum tissue samples, and lower UCP2 expression, whereas the CLP + GK group had no such effects. Moreover, the CLP + GIK group exhibited decreased epithelial permeability of the ileum and increased expression of zonula occludens-1, occludin, and claudin-1 in the ileum. The findings demonstrated that the UCP2 and NLR family-pyrin domain-containing 3/caspase 1/interleukin 1β signaling pathway may be involved in intestinal barrier injury and that GIK treatment decreased intestinal barrier permeability. Thus, GIK may be a useful treatment for intestinal barrier injury during sepsis.
Collapse
|
69
|
Vazquez-Anaya G, Martinez B, Soñanez-Organis JG, Nakano D, Nishiyama A, Ortiz RM. Exogenous thyroxine improves glucose intolerance in insulin-resistant rats. J Endocrinol 2017; 232:501-511. [PMID: 27980001 PMCID: PMC5419047 DOI: 10.1530/joe-16-0428] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022]
Abstract
Both hypothyroidism and hyperthyroidism are associated with glucose intolerance, calling into question the contribution of thyroid hormones (TH) on glucose regulation. TH analogues and derivatives may be effective treatment options for glucose intolerance and insulin resistance (IR), but their potential glucoregulatory effects during conditions of impaired metabolism are not well described. To assess the effects of thyroxine (T4) on glucose intolerance in a model of insulin resistance, an oral glucose tolerance test (oGTT) was performed on three groups of rats (n = 8): (1) lean, Long Evans Tokushima Otsuka (LETO), (2) obese, Otsuka Long Evans Tokushima Fatty (OLETF) and (3) OLETF + T4 (8.0 µg/100 g BM/day × 5 weeks). T4 attenuated glucose intolerance by 15% and decreased IR index (IRI) by 34% in T4-treated OLETF compared to untreated OLETF despite a 31% decrease in muscle Glut4 mRNA expression. T4 increased the mRNA expressions of muscle monocarboxylate transporter 10 (Mct10), deiodinase type 2 (Di2), sirtuin 1 (Sirt1) and uncoupling protein 2 (Ucp2) by 1.8-, 2.2-, 2.7- and 1.4-fold, respectively, compared to OLETF. Activation of AMP-activated protein kinase (AMPK) and insulin receptor were not significantly altered suggesting that the improvements in glucose intolerance and IR were independent of enhanced insulin-mediated signaling. The results suggest that T4 treatment increased the influx of T4 in skeletal muscle and, with an increase of DI2, increased the availability of the biologically active T3 to upregulate key factors such SIRT1 and UCP2 involved in cellular metabolism and glucose homeostasis.
Collapse
Affiliation(s)
| | - Bridget Martinez
- Department of Molecular & Cellular BiologyUniversity of California, Merced, California, USA
| | - José G Soñanez-Organis
- Division of Science and EngineeringDepartment of Chemical Biological and Agropecuary Sciences, University of Sonora, Navojoa, Sonora, Mexico
| | - Daisuke Nakano
- Department of PharmacologyFaculty of Medicine, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of PharmacologyFaculty of Medicine, Kagawa University, Kagawa, Japan
| | - Rudy M Ortiz
- Department of Molecular & Cellular BiologyUniversity of California, Merced, California, USA
| |
Collapse
|
70
|
Brandi J, Cecconi D, Cordani M, Torrens-Mas M, Pacchiana R, Dalla Pozza E, Butera G, Manfredi M, Marengo E, Oliver J, Roca P, Dando I, Donadelli M. The antioxidant uncoupling protein 2 stimulates hnRNPA2/B1, GLUT1 and PKM2 expression and sensitizes pancreas cancer cells to glycolysis inhibition. Free Radic Biol Med 2016; 101:305-316. [PMID: 27989750 DOI: 10.1016/j.freeradbiomed.2016.10.499] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/12/2016] [Accepted: 10/24/2016] [Indexed: 01/09/2023]
Abstract
Several evidence indicate that metabolic alterations play a pivotal role in cancer development. Here, we report that the mitochondrial uncoupling protein 2 (UCP2) sustains the metabolic shift from mitochondrial oxidative phosphorylation (mtOXPHOS) to glycolysis in pancreas cancer cells. Indeed, we show that UCP2 sensitizes pancreas cancer cells to the treatment with the glycolytic inhibitor 2-deoxy-D-glucose. Through a bidimensional electrophoresis analysis, we identify 19 protein species differentially expressed after treatment with the UCP2 inhibitor genipin and, by bioinformatic analyses, we show that these proteins are mainly involved in metabolic processes. In particular, we demonstrate that the antioxidant UCP2 induces the expression of hnRNPA2/B1, which is involved in the regulation of both GLUT1 and PKM2 mRNAs, and of lactate dehydrogenase (LDH) increasing the secretion of L-lactic acid. We further demonstrate that the radical scavenger N-acetyl-L-cysteine reverts hnRNPA2/B1 and PKM2 inhibition by genipin indicating a role for reactive oxygen species in the metabolic reprogramming of cancer cells mediated by UCP2. We also observe an UCP2-dependent decrease in mtOXPHOS complex I (NADH dehydrogenase), complex IV (cytochrome c oxidase), complex V (ATPase) and in mitochondrial oxygen consumption, suggesting a role for UCP2 in the counteraction of pancreatic cancer cellular respiration. All these results reveal novel mechanisms through which UCP2 promotes cancer cell proliferation with the concomitant metabolic shift from mtOXPHOS to the glycolytic pathway.
Collapse
Affiliation(s)
- Jessica Brandi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona, Italy
| | - Daniela Cecconi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona, Italy
| | - Marco Cordani
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Margalida Torrens-Mas
- Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute (ISCIII), Madrid, Spain; Palma Institute for Health Research (IdISPa), E07010 Palma, Spain; Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma, Spain
| | - Raffaella Pacchiana
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Elisa Dalla Pozza
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Giovanna Butera
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Marcello Manfredi
- Department of Sciences and Technological Innovation, University of Eastern Piedmont, Alessandria, Italy; ISALIT, Novara, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Eastern Piedmont, Alessandria, Italy
| | - Jordi Oliver
- Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute (ISCIII), Madrid, Spain; Palma Institute for Health Research (IdISPa), E07010 Palma, Spain; Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma, Spain
| | - Pilar Roca
- Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute (ISCIII), Madrid, Spain; Palma Institute for Health Research (IdISPa), E07010 Palma, Spain; Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma, Spain
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| | - Massimo Donadelli
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
71
|
Cortes-Oliveira C, Nicoletti CF, de Souza Pinhel MA, de Oliveira BAP, Quinhoneiro DCG, Noronha NY, Marchini JS, da Silva Júnior WA, Júnior WS, Nonino CB. UCP2 expression is associated with weight loss after hypocaloric diet intervention. Eur J Clin Nutr 2016; 71:402-406. [PMID: 27759071 DOI: 10.1038/ejcn.2016.185] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/09/2016] [Accepted: 08/12/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES Although energy restriction contributes to weight loss, it may also reduce energy expenditure, limiting the success of weight loss in the long term. Studies have described how genetics contributes to the development of obesity, and uncoupling proteins 1 and 2 (UCP1 and UCP2) and beta-3-adrenoceptor (ADRB3) have been implicated in the metabolic pathways that culminate in this condition. This study aimed to evaluate how the UCP1, UCP2 and ADRB3 genes influence weight loss in severely obese women submitted to hypocaloric dietary intervention. SUBJECTS/METHODS This longitudinal study included 21 women divided into two groups: Group 1 (Dietary intervention (G1)) consisted of 11 individuals with severe obesity (body mass index (BMI) ⩾40 kg/m2), selected for dietary intervention and Group 2 (Control (G2)) consisted of 10 normal-weight women (BMI between 18.5 and 24.9 kg/m2). Evaluation included weight (kg), height (m), waist circumference (cm), body composition, resting metabolic rate (RMR, kcal) and abdominal subcutaneous adipose tissue collection. The dietary intervention required that G1 patients remained hospitalized in the university hospital for 6 weeks receiving a hypocaloric diet (1200 kcal per day). The statistical analyses included t-test for paired samples, Spearman correlation and multivariate linear regressions, with the level of significance set at P<0.05. RESULTS Weight (155.0±31.4-146.5±27.8 kg), BMI (58.5±10.5-55.3±9.2 kg/m2), fat-free mass (65.4±8.6-63.1±7.1 kg), fat mass (89.5±23.0-83.4±21.0 kg) and RMR (2511.6±386.1-2324.0±416.4 kcal per day) decreased significantly after dietary intervention. Multiple regression analyses showed that UCP2 expression contributed to weight loss after dietary intervention (P=0.05). CONCLUSIONS UCP2 expression is associated with weight loss after hypocaloric diet intervention.
Collapse
Affiliation(s)
- C Cortes-Oliveira
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - C F Nicoletti
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - M A de Souza Pinhel
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - B A P de Oliveira
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - D C G Quinhoneiro
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - N Y Noronha
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - J S Marchini
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - W A da Silva Júnior
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - W S Júnior
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - C B Nonino
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
72
|
In vitro evidence in rainbow trout supporting glucosensing mediated by sweet taste receptor, LXR, and mitochondrial activity in Brockmann bodies, and sweet taste receptor in liver. Comp Biochem Physiol B Biochem Mol Biol 2016; 200:6-16. [DOI: 10.1016/j.cbpb.2016.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 12/31/2022]
|
73
|
Du RH, Wu FF, Lu M, Shu XD, Ding JH, Wu G, Hu G. Uncoupling protein 2 modulation of the NLRP3 inflammasome in astrocytes and its implications in depression. Redox Biol 2016; 9:178-187. [PMID: 27566281 PMCID: PMC5007434 DOI: 10.1016/j.redox.2016.08.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial uncoupling protein 2 (UCP2) has been well characterized to control the production of reactive oxygen species (ROS) and astrocytes are the major cells responsible for the ROS production and the inflammatory responses in the brain. However, the function of UCP2 in astrocytes and the contribution of astrocytic UCP2 to depression remain undefined. Herein, we demonstrated that UCP2 knockout (KO) mice displayed aggravated depressive-like behaviors, impaired neurogenesis, and enhanced loss of astrocytes in the chronic mild stress (CMS)-induced anhedonia model of depression. We further found that UCP2 ablation significantly enhanced the activation of the nod-like receptor protein 3 (NLRP3) inflammasome in the hippocampus and in astrocytes. Furthermore, UCP2 deficiency promoted the injury of mitochondria, the generation of ROS and the physical association between thioredoxin-interacting protein (TXNIP) and NLRP3 in astrocytes. Moreover, transiently expressing exogenous UCP2 partially rescued the deleterious effects of UCP2 ablation on the astrocytes. These data indicate that UCP2 negatively regulates the activation of NLRP3 inflammasome and inhibited the ROS-TXNIP-NLRP3 pathway in astrocytes. Collectively, our findings reveal that UCP2 regulates inflammation responses in astrocytes and plays an important role in the pathogenesis of depression and that UCP2 may be a promising therapeutic target for depression.
Collapse
Affiliation(s)
- Ren-Hong Du
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China
| | - Fang-Fang Wu
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China
| | - Xiao-Dong Shu
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents, 1459 Laney Walker Blvd., Augusta, GA 30912, United States
| | - Gang Hu
- Jiangsu Key Laboratory of Neurogeneration, Department of Pharmacology, Nanjing Medical University, 101 Nongmian Road, Nanjing, Jiangsu 210029, PR China; Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, PR China.
| |
Collapse
|
74
|
Nicoletti CF, Kimura BM, de Oliveira BAP, de Pinhel MAS, Salgado W, Marchini JS, Nonino CB. Role of UCP2 polymorphisms on dietary intake of obese patients who underwent bariatric surgery. Clin Obes 2016; 6:354-8. [PMID: 27256164 DOI: 10.1111/cob.12148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/05/2016] [Accepted: 04/12/2016] [Indexed: 12/26/2022]
Abstract
Uncoupling protein 2 ( UCP2 ) plays an important role in body weight and energy metabolism and may be related to the control of food consumption. This study aimed to investigate the contribution of UCP2 gene variants on the dietary intake on a population after bariatric surgery. This study enrolled 150 obese patients (body mass index ≥ 35kg m(-2) ) who submitted to Roux-en-Y gastric bypass. Weight (kg), BMI (kg m(-2) ), energy (kcal d(-1) ) and macronutrients intake (g d(-1) ) of preoperative and 1-year postoperative period were collected from medical records. Ala55Val and -866G>A polymorphisms in the UCP2 gene were genotyped through allelic discrimination method in real-time polymerase chain reaction using the TaqMan pre-designed SNP Genotyping Assays kits. Hardy-Weinberg equilibrium, t-test and regression models were performed in statistical analysis (P<0.05).We found an allelic frequency of 0.44 for allele Val and 0.41 for allele A. In the postoperative period, patients with at least one rare allele for polymorphisms and with at least one rare allele for both polymorphisms together (haplotype) present a greater energy and carbohydrate intake, even after adjusting for gender, age and weight. Genetic variants in UCP2 gene were associated with the dietary consumption after Roux-En-Y gastric bypass.
Collapse
Affiliation(s)
- C F Nicoletti
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - B M Kimura
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - B A P de Oliveira
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - M A S de Pinhel
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - W Salgado
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - J S Marchini
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil
| | - C B Nonino
- Department of Internal Medicine, Faculty of Medicine of Ribeirao Preto, University of São Paulo, RibeirãoPreto, Brazil.
| |
Collapse
|
75
|
Cell Death and Heart Failure in Obesity: Role of Uncoupling Proteins. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9340654. [PMID: 27642497 PMCID: PMC5011521 DOI: 10.1155/2016/9340654] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
Abstract
Metabolic diseases such as obesity, metabolic syndrome, and type II diabetes are often characterized by increased reactive oxygen species (ROS) generation in mitochondrial respiratory complexes, associated with fat accumulation in cardiomyocytes, skeletal muscle, and hepatocytes. Several rodents studies showed that lipid accumulation in cardiac myocytes produces lipotoxicity that causes apoptosis and leads to heart failure, a dynamic pathological process. Meanwhile, several tissues including cardiac tissue develop an adaptive mechanism against oxidative stress and lipotoxicity by overexpressing uncoupling proteins (UCPs), specific mitochondrial membrane proteins. In heart from rodent and human with obesity, UCP2 and UCP3 may protect cardiomyocytes from death and from a state progressing to heart failure by downregulating programmed cell death. UCP activation may affect cytochrome c and proapoptotic protein release from mitochondria by reducing ROS generation and apoptotic cell death. Therefore the aim of this review is to discuss recent findings regarding the role that UCPs play in cardiomyocyte survival by protecting against ROS generation and maintaining bioenergetic metabolism homeostasis to promote heart protection.
Collapse
|
76
|
Chen GG, Yan JB, Wang XM, Zheng MZ, Jiang JP, Zhou XM, Cai B, Shen YL. Mechanism of uncoupling protein 2‑mediated myocardial injury in hypothermic preserved rat hearts. Mol Med Rep 2016; 14:1857-64. [PMID: 27356851 DOI: 10.3892/mmr.2016.5436] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/10/2016] [Indexed: 11/05/2022] Open
Abstract
In the present study, the alterations in uncoupling protein 2 (UCP2) expression following hypothermic preservation in rat hearts were investigated. Isolated rat hearts were preserved in Celsior solution for 3‑12 h followed by 60 min of reperfusion. The cardiac function was evaluated using the Langendorff perfusion system. UCP2 and silent mating type information regulation 2 homolog 1 (SIRT1) proteins were detected by western blot analysis. The ATP production and mitochondrial reactive oxygen species (ROS) levels were assessed. Subsequent to preservation in ice‑cold Celsior solution for 3‑12 h, the UCP2 protein expression in rat hearts was observed to increase in a time‑dependent manner. The UCP2 inhibitor genipin inhibited the hypothermic preservation‑induced cardiac dysfunction, prevented a decline in ATP production induced by 9 h of preservation, however had no effect on the hypothermic preservation‑induced increase in mitochondrial ROS levels. Compared with the control group, the SIRT1 protein expression in rat hearts reduced following hypothermic preservation. Compared with the 9‑h preservation group, Celsior solution supplemented with the SIRT1 activator resveratrol (20 or 40 µmol/l) inhibited UCP2 protein overexpression, prevented the decline in ATP production and resulted in an improvement cardiac function. The SIRT1 inhibitor EX‑527 abolished the resveratrol‑induced inhibition of UCP2 overexpression and cardiac protection in the hypothermic preserved rat heart. These observations suggest that downregulation of UCP2 expression in the hypothermic preserved rat heart in part initiated the protective mechanism via the SIRT1 pathway.
Collapse
Affiliation(s)
- Gai-Ge Chen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Jin-Bin Yan
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Xu-Ming Wang
- Department of Pharmacology, Medicine School, Taizhou University, Taizhou, Zhejiang 317000, P.R. China
| | - Ming-Zhi Zheng
- Department of Pharmacology, Zhejiang Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Jian-Ping Jiang
- Department of Clinical Medicine, Zhejiang Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Xin-Mei Zhou
- Department of Physiology, Jiaxing University College of Medicine, Jiaxing, Zhejiang 314001, P.R. China
| | - Bin Cai
- Department of Medical Affairs, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Yue-Liang Shen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
77
|
Yang L, Dong Z, Zhou J, Ma Y, Pu W, Zhao D, He H, Ji H, Yang Y, Wang X, Xu X, Pang Y, Zou H, Jin L, Yang C, Wang J. Common UCP2 variants contribute to serum urate concentrations and the risk of hyperuricemia. Sci Rep 2016; 6:27279. [PMID: 27273589 PMCID: PMC4897637 DOI: 10.1038/srep27279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022] Open
Abstract
Elevated serum urate, which is regulated at multiple levels including genetic variants, is a risk factor for gout and other metabolic diseases. This study aimed to investigate the association between UCP2 variants and serum urate as well as hyperuricemia in a Chinese population. In total, 4332 individuals were genotyped for two common UCP2 variants, -866G/A and Ala55Val. These loci were not associated either serum urate level or with a risk of hyperuricemia in the total group of subjects. However, in females, -866G/A and Ala55Val were associated with a lower serum urate (P = 0.006 and 0.014, seperately) and played a protective role against hyperuricemia (OR = 0.80, P = 0.018; OR = 0.79, P = 0.016). These associations were not observed in the males. After further stratification, the two loci were associated with serum urate in overweight, but not underweight females. The haplotype A-T (-866G/A-Ala55Val) was a protective factor for hyperuricemia in the female subgroup (OR = 0.80, P = 0.017). This present study identified a novel gene, UCP2, that influences the serum urate concentration and the risk of hyperuricemia, and the degree of association varies with gender and BMI levels.
Collapse
Affiliation(s)
- Luyu Yang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Zheng Dong
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jingru Zhou
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Dongbao Zhao
- Division of Rheumatology and Immunology, Changhai Hospital, Shanghai, China
| | - Hongjun He
- Division of Rheumatology, Taixing People's Hospital, Jiangsu Province, China
| | - Hengdong Ji
- Division of Rheumatology, Taizhou People's Hospital, Jiangsu Province, China
| | - Yajun Yang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China
| | - Xia Xu
- Division of Rheumatology and Immunology, Changhai Hospital, Shanghai, China
| | - Yafei Pang
- Division of Rheumatology and Immunology, Changhai Hospital, Shanghai, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China
| | - Chengde Yang
- Division of Rheumatology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
78
|
Roy M, Itoh K, Iijima M, Sesaki H. Parkin suppresses Drp1-independent mitochondrial division. Biochem Biophys Res Commun 2016; 475:283-8. [PMID: 27181353 DOI: 10.1016/j.bbrc.2016.05.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/08/2016] [Indexed: 12/12/2022]
Abstract
The cycle of mitochondrial division and fusion disconnect and reconnect individual mitochondria in cells to remodel this energy-producing organelle. Although dynamin-related protein 1 (Drp1) plays a major role in mitochondrial division in cells, a reduced level of mitochondrial division still persists even in the absence of Drp1. It is unknown how much Drp1-mediated mitochondrial division accounts for the connectivity of mitochondria. The role of a Parkinson's disease-associated protein-parkin, which biochemically and genetically interacts with Drp1-in mitochondrial connectivity also remains poorly understood. Here, we quantified the number and connectivity of mitochondria using mitochondria-targeted photoactivatable GFP in cells. We show that the loss of Drp1 increases the connectivity of mitochondria by 15-fold in mouse embryonic fibroblasts (MEFs). While a single loss of parkin does not affect the connectivity of mitochondria, the connectivity of mitochondria significantly decreased compared with a single loss of Drp1 when parkin was lost in the absence of Drp1. Furthermore, the loss of parkin decreased the frequency of depolarization of the mitochondrial inner membrane that is caused by increased mitochondrial connectivity in Drp1-knockout MEFs. Therefore, our data suggest that parkin negatively regulates Drp1-indendent mitochondrial division.
Collapse
Affiliation(s)
- Madhuparna Roy
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
79
|
Barnstable CJ, Reddy R, Li H, Horvath TL. Mitochondrial Uncoupling Protein 2 (UCP2) Regulates Retinal Ganglion Cell Number and Survival. J Mol Neurosci 2016; 58:461-9. [PMID: 26846222 PMCID: PMC4833669 DOI: 10.1007/s12031-016-0728-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/27/2016] [Indexed: 12/29/2022]
Abstract
In the brain, mitochondrial uncoupling protein 2 (UCP2) has emerged as a stress signal associated with neuronal survival. In the retina, UCP2 is expressed primarily by retinal ganglion cells. Here, we investigated the functional relevance of UCP2 in the mouse retina. Increased expression of UCP2 significantly reduced apoptosis during the critical developmental period resulting in elevated numbers of retinal ganglion cells in the adult. Elevated UCP2 levels also protected against excitotoxic cell death induced by intraocular injection of either NMDA or kainic acid. In monolayer cultures of retinal cells, elevated UCP2 levels increased cell survival and rendered the cells independent of the survival-promoting effects of the neurotrophic factors BDNF and CNTF. Taken together, these data implicate UCP2 as an important regulator of retinal neuron survival both during development and in adult animals.
Collapse
Affiliation(s)
- Colin J Barnstable
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Department of Neural and Behavioral Science, Penn State University College of Medicine, H109, 500 University Drive, Hershey, PA, 17033, USA.
| | - Rajini Reddy
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Hong Li
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neural and Behavioral Science, Penn State University College of Medicine, H109, 500 University Drive, Hershey, PA, 17033, USA
| | - Tamas L Horvath
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
80
|
Otero-Rodiño C, Velasco C, Álvarez-Otero R, López-Patiño MA, Míguez JM, Soengas JL. In vitro evidence supports the presence of glucokinase-independent glucosensing mechanisms in hypothalamus and hindbrain of rainbow trout. ACTA ACUST UNITED AC 2016; 219:1750-9. [PMID: 27026717 DOI: 10.1242/jeb.137737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/03/2016] [Indexed: 11/20/2022]
Abstract
We previously obtained evidence in rainbow trout for the presence and response to changes in circulating levels of glucose (induced by intraperitoneal hypoglycaemic and hyperglycaemic treatments) of glucosensing mechanisms based on liver X receptor (LXR), mitochondrial production of reactive oxygen species (ROS) leading to increased expression of uncoupling protein 2 (UCP2), and sweet taste receptor in the hypothalamus, and on sodium/glucose co-transporter 1 (SGLT-1) in hindbrain. However, these effects of glucose might be indirect. Therefore, we evaluated the response of parameters related to these glucosensing mechanisms in a first experiment using pooled sections of hypothalamus and hindbrain incubated for 6 h at 15°C in modified Hanks' medium containing 2, 4 or 8 mmol l(-1) d-glucose. The responses observed in some cases were consistent with glucosensing capacity. In a second experiment, pooled sections of hypothalamus and hindbrain were incubated for 6 h at 15°C in modified Hanks' medium with 8 mmol l(-1) d-glucose alone (control) or containing 1 mmol l(-1) phloridzin (SGLT-1 antagonist), 20 µmol l(-1) genipin (UCP2 inhibitor), 1 µmol l(-1) trolox (ROS scavenger), 100 µmol l(-1) bezafibrate (T1R3 inhibitor) and 50 µmol l(-1) geranyl-geranyl pyrophosphate (LXR inhibitor). The response observed in the presence of these specific inhibitors/antagonists further supports the proposal that critical components of the different glucosensing mechanisms are functioning in rainbow trout hypothalamus and hindbrain.
Collapse
Affiliation(s)
- Cristina Otero-Rodiño
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| | - Cristina Velasco
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| | - Rosa Álvarez-Otero
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| | - Marcos A López-Patiño
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| | - Jesús M Míguez
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| | - José L Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo E-36310, Spain
| |
Collapse
|
81
|
Oliveira BAP, Pinhel MAS, Nicoletti CF, Oliveira CC, Quinhoneiro DCG, Noronha NY, Marchini JS, Marchry AJ, Junior WS, Nonino CB. UCP1 and UCP3 Expression Is Associated with Lipid and Carbohydrate Oxidation and Body Composition. PLoS One 2016; 11:e0150811. [PMID: 26959981 PMCID: PMC4784729 DOI: 10.1371/journal.pone.0150811] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/19/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND/OBJECTIVE Uncoupling proteins (UCPs) are located in the inner membrane of mitochondria. These proteins participate in thermogenesis and energy expenditure. This study aimed to evaluate how UCP1 and UCP3 expression influences substrate oxidation and elicits possible changes in body composition in patients submitted to bariatric surgery. SUBJECTS/METHODS This is a longitudinal study comprising 13 women with obesity grade III that underwent bariatric surgery and 10 healthy weight individuals (control group). Body composition was assessed by bioelectrical impedance. Carbohydrate and fat oxidation was determined by indirect calorimetry. Subcutaneous adipose tissue was collected for gene expression analysis. QPCR was used to evaluate UCP1 and UCP3 expression. RESULTS Obese patients and the control group differed significantly in terms of lipid and carbohydrate oxidation. Six months after bariatric surgery, the differences disappeared. Lipid oxidation correlated with the percentage of fat mass in the postoperative period. Multiple linear regression analysis showed that the UCP1 and UCP3 genes contributed to lipid and carbohydrate oxidation. Additionally, UCP3 expression was associated with BMI, percentage of lean body mass, and percentage of mass in the postoperative period. CONCLUSIONS UCP1 and UCP3 expression is associated with lipid and carbohydrate oxidation in patients submitted to bariatric surgery. In addition, UCP3 participates in body composition modulation six months postoperatively.
Collapse
Affiliation(s)
- Bruno A. P. Oliveira
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Marcela A. S. Pinhel
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Carolina F. Nicoletti
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Cristiana C. Oliveira
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Driele C. G. Quinhoneiro
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Natália Y. Noronha
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Júlio S. Marchini
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Ana J. Marchry
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Wilson S. Junior
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Carla B. Nonino
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
- * E-mail:
| |
Collapse
|
82
|
Lehnen TE, da Silva MR, Camacho A, Marcadenti A, Lehnen AM. A review on effects of conjugated linoleic fatty acid (CLA) upon body composition and energetic metabolism. J Int Soc Sports Nutr 2015; 12:36. [PMID: 26388708 PMCID: PMC4574006 DOI: 10.1186/s12970-015-0097-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/04/2015] [Indexed: 01/01/2023] Open
Abstract
Conjugated linoleic acid (CLA) is highly found in fats from ruminants and it appears to favorably modify the body composition and cardiometabolic risk factors. The capacity of CLA to reduce the body fat levels as well as its benefic actions on glycemic profile, atherosclerosis and cancer has already been proved in experimental models. Furthermore, CLA supplementation may modulate the immune function, help re-synthetize of glycogen and potentiate the bone mineralization. CLA supplementation also could increase the lipolysis and reduce the accumulation of fatty acids on the adipose tissue; the putative mechanisms involved may be its action in reducing the lipase lipoprotein activity and to increase the carnitine-palmitoil-transferase-1 (CAT-1) activity, its interaction with PPARγ, and to raise the expression of UCP-1. Although studies made in human have shown some benefits of CLA supplementation as the weight loss, the results are still discordant. Moreover, some have shown adverse effects, such as negative effects on glucose metabolism and lipid profile. The purpose of this article is to review the available data regarding the benefits of CLA on the energetic metabolism and body composition, emphasizing action mechanisms.
Collapse
Affiliation(s)
- Tatiana Ederich Lehnen
- Faculdade Sogipa de Educação Física, Porto Alegre, Brazil ; Instituto de Cardiologia do Rio Grande do Sul, Av. Princesa Isabel, 395 Santana, 90620-001 Porto Alegre, RS Brazil
| | | | - Augusto Camacho
- Faculdade Sogipa de Educação Física, Porto Alegre, Brazil ; Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Porto Alegre, Brazil
| | - Aline Marcadenti
- Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Porto Alegre, Brazil ; Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Alexandre Machado Lehnen
- Faculdade Sogipa de Educação Física, Porto Alegre, Brazil ; Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Porto Alegre, Brazil
| |
Collapse
|
83
|
Zhou MC, Yu P, Sun Q, Li YX. Expression profiling analysis: Uncoupling protein 2 deficiency improves hepatic glucose, lipid profiles and insulin sensitivity in high-fat diet-fed mice by modulating expression of genes in peroxisome proliferator-activated receptor signaling pathway. J Diabetes Investig 2015; 7:179-89. [PMID: 27042269 PMCID: PMC4773664 DOI: 10.1111/jdi.12402] [Citation(s) in RCA: 15] [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: 04/06/2015] [Revised: 06/30/2015] [Accepted: 07/22/2015] [Indexed: 12/19/2022] Open
Abstract
Aims/Introduction Uncoupling protein 2 (UCP2), which was an important mitochondrial inner membrane protein associated with glucose and lipid metabolism, widely expresses in all kinds of tissues including hepatocytes. The present study aimed to explore the impact of UCP2 deficiency on glucose and lipid metabolism, insulin sensitivity and its effect on the liver‐associated signaling pathway by expression profiling analysis. Materials and Methods Four‐week‐old male UCP2−/− mice and UCP2+/+ mice were randomly assigned to four groups: UCP2−/− on a high‐fat diet, UCP2−/− on a normal chow diet, UCP2+/+ on a high‐fat diet and UCP2+/+ on a normal chow diet. The differentially expressed genes in the four groups on the 16th week were identified by Affymetrix gene array. Results The results of intraperitoneal glucose tolerance test and insulin tolerance showed that blood glucose and β‐cell function were improved in the UCP2−/− group on high‐fat diet. Enhanced insulin sensitivity was observed in the UCP2−/− group. The differentially expressed genes were mapped to 23 pathways (P < 0.05). We concentrated on the ‘peroxisome proliferator‐activated receptor (PPAR) signaling pathway’ (P = 3.19 × 10−11), because it is closely associated with the regulation of glucose and lipid profiles. In the PPAR signaling pathway, seven genes (PPARγ, Dbi, Acsl3, Lpl, Me1, Scd1, Fads2) in the UCP2−/− mice were significantly upregulated. Conclusions The present study used gene arrays to show that activity of the PPAR signaling pathway involved in the improvement of glucose and lipid metabolism in the liver of UCP2‐deficient mice on a long‐term high‐fat diet. The upregulation of genes in the PPAR signaling pathway could explain our finding that UCP2 deficiency ameliorated insulin sensitivity. The manipulation of UCP2 protein expression could represent a new strategy for the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Mei-Cen Zhou
- Department of Endocrinology Key Laboratory of Endocrinology Ministry of Health Peking Union Medical College Hospital Beijing China
| | - Ping Yu
- Department of Pharmacy the Third People's Hospital Qinhuangdao China
| | - Qi Sun
- Department of Endocrinology Key Laboratory of Endocrinology Ministry of Health Peking Union Medical College Hospital Beijing China
| | - Yu-Xiu Li
- Department of Endocrinology Key Laboratory of Endocrinology Ministry of Health Peking Union Medical College Hospital Beijing China
| |
Collapse
|
84
|
Wang C, Li Q, Wang W, Guo L, Guo C, Sun Y, Zhang J. GLP-1 contributes to increases in PGC-1α expression by downregulating miR-23a to reduce apoptosis. Biochem Biophys Res Commun 2015; 466:33-9. [PMID: 26315270 DOI: 10.1016/j.bbrc.2015.08.092] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 11/15/2022]
Abstract
GLP-1 can help to overcome problems of liver cells metabolism, not only pancreatic cell. But the explicit mechanism of this effect remains unclear. In recent years, microRNAs have received the attention of researchers and some microRNAs have important implications for diabetes. The mitochondrial protective gene PGC-1α is also closely related to diabetes, and UCP2 is related to anti-mitochondrial oxidative stress, but the mechanism of action of these genes is unclear. In this study, we used HepG2 cell line and used the cell counting kit (CCK) to measure the cell viability with GLP-1(7-36) and/or glucotoxicity. To investigate alterations in gene expression resulting from incubation with GLP-1 (7-36) or hyperglycaemia, the RNA expression levels of miR-23a, PGC-1α, Bak, Bax and UCP2 were quantified using real-time PCR. The protein levels of PGC-1α were determined by western blot. The role of miR-23a in the regulation of PGC-1α was further assessed through cell transfection to downregulate of miR-23a expression. In this study, the viability of HepG2 hepatocytes was decreased under hyperglycaemia, but incubation with 10 nmol/L GLP-1 (7-36) amide for 24 h significantly increased the viability and decreased the mRNA expression levels of Bax and Bak. Incubation with GLP-1(7-36) amide for 24 h attenuated the RNA expression of miR-23a and increased the mRNA and protein expression of PGC-1α. Inhibition of miR-23a expression by cell transfection led to increases in the mRNA and protein expression of PGC-1α. In addition, the mRNA expression of UCP2 increased after incubation with GLP-1(7-36) for 24 h. In conclusion, GLP-1 induced increased expression of mitochondrial protective gene PGC-1α by downregulating miR-23a to inhibit hepatocyte apoptosis and also enhanced UCP2 to reduce apoptosis.
Collapse
Affiliation(s)
- Chi Wang
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| | - Qiang Li
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China.
| | - Wei Wang
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| | - Lin Guo
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| | - Chang Guo
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| | - Yiqiong Sun
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| | - Jinchao Zhang
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Xuefu Road No. 246, Harbin, Heilongjiang 150086, China
| |
Collapse
|
85
|
Ould Hamouda H, Delplanque B, Benomar Y, Crépin D, Riffault L, LeRuyet P, Bonhomme C, Taouis M. Milk-soluble formula increases food intake and reduces Il6 expression in elderly rat hypothalami. J Endocrinol 2015; 226:67-80. [PMID: 25994005 DOI: 10.1530/joe-15-0076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2015] [Indexed: 01/17/2023]
Abstract
Malnutrition in the elderly is accompanied by several metabolic dysfunctions, especially alterations in energy homeostasis regulation and a loss of insulin responsiveness. Nutritional recommendations aim to enrich food with high protein and energy supplements, and protein composition and lipid quality have been widely studied. Despite the numerous studies that have examined attempts to overcome malnutrition in the elderly through such nutritional supplementation, it is still necessary to study the effects of a combination of protein, lipids, and vitamin D (VitD). This can be done in animal models of elderly malnutrition. In the present study, we investigated the effects of several diet formulae on insulin responsiveness, inflammation, and the hypothalamic expression of key genes that are involved in energy homeostasis control. To mimic elderly malnutrition in humans, elderly Wistar rats were food restricted (R, -50%) for 12 weeks and then refed for 4 weeks with one of four different isocaloric diets: a control diet; a diet where milk soluble protein (MSP) replaced casein; a blend of milk fat, rapeseed, and DHA (MRD); or a full formula (FF) diet that combined MSP and a blend of MRD (FF). All of the refeeding diets contained VitD. We concluded that: (i) food restriction led to the upregulation of insulin receptor in liver and adipose tissue accompanied by increased Tnfα in the hypothalamus; (ii) in all of the refed groups, refeeding led to similar body weight gain during the refeeding period; and (iii) refeeding with MSP and MRD diets induced higher food intake on the fourth week of refeeding, and this increase was associated with reduced hypothalamic interleukin 6 expression.
Collapse
Affiliation(s)
- Hassina Ould Hamouda
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Bernadette Delplanque
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Yacir Benomar
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Delphine Crépin
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Laure Riffault
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Pascale LeRuyet
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Cécile Bonhomme
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| | - Mohammed Taouis
- Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France Neuroendocrinologie Moléculaire de la Prise AlimentaireUniversity of Paris-Sud, UMR 8195, F-91405 Orsay, FranceNeuroendocrinologie Moléculaire de la Prise AlimentaireCNRS, Centre de Neurosciences Paris-Sud, UMR 8195, F-91405 Orsay, FranceService NutritionLactalis Recherche et Développement, 8 Fromy, CS 60082, 35240 Retiers, FranceLactalis Nutrition Parc d'Activité de Torcé-secteur Est35370 Torcé, France
| |
Collapse
|