1
|
Barbut D, Kinney WA, Chen HH, Stewart AFR, Hecksher-Sørensen J, Zhang C, Fleming A, Zemel M, Zasloff M. A novel, centrally acting mammalian aminosterol, ENT-03, induces weight loss in obese and lean rodents. Diabetes Obes Metab 2024; 26:5701-5712. [PMID: 39307948 DOI: 10.1111/dom.15940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 11/05/2024]
Abstract
ENT-03, a spermine bile acid we recently discovered in the brain of newborn mice acts centrally to regulate energy and metabolism. Obese, diabetic (ob/ob) mice treated with five doses of ENT-03 over 2 weeks, demonstrated a rapid decrease in blood glucose levels into the range seen in non-obese animals, prior to any significant weight loss. Weight fell substantially thereafter as food intake decreased, and serum biochemical parameters normalized compared with both vehicle and pair-fed controls. To determine whether ENT-03 could be acting centrally, we injected a single dose of ENT-03 intracerebroventricularly to Sprague-Dawley rats. Weight fell significantly and remained below vehicle injected controls for an extended period. By autoradiography, ENT-03 localized to the arcuate nucleus of the hypothalamus, the choroid plexus and cerebrospinal fluid. Significant cFos activation occurred in multiple anatomical regions within the hypothalamus and brainstem involved in appetite suppression, food-entrained circadian rhythmicity, autonomic function, and growth. These data support a role for ENT-03 in the treatment of type 2 diabetes and obesity. Phase 1 studies in subjects with obesity and diabetes are currently in progress.
Collapse
Affiliation(s)
- Denise Barbut
- Enterin, Inc., Philadelphia, Pennsylvania, USA
- Enterin Research Institute, Philadelphia, Pennsylvania, USA
| | - William A Kinney
- Enterin, Inc., Philadelphia, Pennsylvania, USA
- Enterin Research Institute, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | - Michael Zasloff
- Enterin, Inc., Philadelphia, Pennsylvania, USA
- Enterin Research Institute, Philadelphia, Pennsylvania, USA
- MedStar Georgetown Transplant Institute, Washington, District of Columbia, USA
| |
Collapse
|
2
|
Singh H, Almabhouh FA, Alshaikhli HSI, Hassan MJM, Daud S, Othman R, Md Salleh MFRR. Leptin in reproduction and hypertension in pregnancy. Reprod Fertil Dev 2024; 36:RD24060. [PMID: 39038160 DOI: 10.1071/rd24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024] Open
Abstract
Leptin has important roles in numerous physiological functions, including those in the regulation of energy balance, and in immune and reproductive systems. However, in the recent years, evidence has implicated it in a number of obesity-related diseases, where its concentrations in serum are significantly elevated. Elevated serum leptin concentrations and increased placental leptin secretion have been reported in women with hypertensive disorders of pregnancy. Whether leptin is responsible for this disorder remains to be established. Leptin injections in healthy rats and mice during pregnancy result in endothelial activation, increased blood pressure and proteinuria. A potential role for leptin in the pathogenesis of pre-eclampsia is hypothesised, particularly in women who are overweight or obese where serum leptin concentrations are often elevated. This review summarises pertinent information in the literature on the role of leptin in puberty, pregnancy, and hypertensive disorders of pregnancy. In particular, the possible mechanism that may be involved in leptin-induced increase in blood pressure and proteinuria during pregnancy and the potential role of marinobufagenin in this disease entity. We hypothesise a significant role for oxidative stress in this, and propose a conceptual framework on the events that lead to endothelial activation, raised blood pressure and proteinuria following leptin administration.
Collapse
Affiliation(s)
- Harbindarjeet Singh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Fayez A Almabhouh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia; and Department of Biology and Biotechnology, Faculty of Science Islamic University of Gaza, Gaza Strip, Palestine
| | | | | | - Suzanna Daud
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Rosfayati Othman
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| | - Muhd Fakh Rur Razi Md Salleh
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| |
Collapse
|
3
|
Chen SM, Hsiao CW, Chen YJ, Hong CJ, Lin JC, Yang CP, Chang YH. Interleukin-4 inhibits the hypothalamic appetite control by modulating the insulin-AKT and JAK-STAT signaling in leptin mutant mice. ENVIRONMENTAL TOXICOLOGY 2024; 39:3980-3990. [PMID: 38597583 DOI: 10.1002/tox.24264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/19/2024] [Accepted: 03/23/2024] [Indexed: 04/11/2024]
Abstract
Our previous research identified interleukin-4 (IL-4) as a key regulator of glucose/lipid metabolism, circulatory leptin levels, and insulin action, suggesting its potential as a therapeutic target for obesity and related complications. This study aimed to further elucidate the role of IL-4 in regulating hypothalamic appetite-controlling neuropeptides using leptin dysfunctional Leptin145E/145E mice as the experimental model. IL-4 significantly reduces body weight, food intake, and serum glucose levels. Our data demonstrated that IL-4 exhibits multiple functions in regulating hypothalamic appetite control, including downregulating orexigenic agouti-related peptide and neuropeptide Y levels, promoting expression of anorexigenic proopiomelanocortin, alleviating microenvironmental hypothalamic inflammation, enhancing leptin and insulin pathway, and attenuating insulin resistance. Furthermore, IL-4 promotes uncoupling protein 1 expression of white adipose tissue (WAT), suggesting its role in triggering WAT-beige switch. In summary, this study uncovers novel function of IL-4 in mediating food-intake behaviors and metabolic efficiency by regulating hypothalamic appetite-control and WAT browning activities. These findings support the therapeutic potential of targeting hypothalamic inflammation and reducing adiposity through IL-4 intervention for tackling the pandemic increasing prevalence of obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Shu-Mei Chen
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Chiao-Wan Hsiao
- Institute of Brain Science of National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Ju Chen
- Research Assistant Center, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan, Taiwan
| | - Chen-Jee Hong
- Section of Psychosomatic Medicine, Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jung-Chun Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Ping Yang
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
4
|
Zhang X, Liu Y, Sun H, Chen S, Tang P, Hu Q, He M, Tang N, Li Z, Chen D. Long-term dietary exposure to 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) reduced feeding in common carp (Cyprinus carpio): Via the JAK-STAT signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 349:123966. [PMID: 38621451 DOI: 10.1016/j.envpol.2024.123966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely present in water ecosystems where they pose a significant threat to aquatic life, but our knowledge about how PBDEs affect feeding is limited. Therefore, this study explored the effects of continuous dietary exposure to 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) (40 and 4000 ng/g) on the feeding in common carp (Cyprinus carpio) and the underlying mechanism. BDE-47 significantly decreased the food intake of carp. Transcriptome analysis of brain tissue showed that BDE-47 mainly affected the nervous, immune, and endocrine systems. Further examination of the expression levels of appetite factors in the brain revealed that BDE-47 caused dysregulation of appetite factors expressions such as agrp, pomc, cart, etc. In addition, the JAK-STAT signaling pathway was activated under BDE-47 exposure. It can be concluded from these findings that BDE-47 activated the JAK-STAT signaling pathway, causing imbalanced expression of appetite factors, leading to disordered feeding behavior and decreased food intake in carp. These results provide an important reference for a more comprehensive understanding of the hazards posed by BDE-47 on animal feeding and the associated mechanisms.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Youlian Liu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huimin Sun
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shuhuang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Peng Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qiang Hu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mengxuan He
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ni Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
5
|
Firth W, Pye KR, Weightman Potter PG. Astrocytes at the intersection of ageing, obesity, and neurodegeneration. Clin Sci (Lond) 2024; 138:515-536. [PMID: 38652065 DOI: 10.1042/cs20230148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Once considered passive cells of the central nervous system (CNS), glia are now known to actively maintain the CNS parenchyma; in recent years, the evidence for glial functions in CNS physiology and pathophysiology has only grown. Astrocytes, a heterogeneous group of glial cells, play key roles in regulating the metabolic and inflammatory landscape of the CNS and have emerged as potential therapeutic targets for a variety of disorders. This review will outline astrocyte functions in the CNS in healthy ageing, obesity, and neurodegeneration, with a focus on the inflammatory responses and mitochondrial function, and will address therapeutic outlooks.
Collapse
Affiliation(s)
- Wyn Firth
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, U.K
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Paul G Weightman Potter
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
6
|
Xu X, Hu X, Ma G, Wang T, Wu J, Zhu X, Chen G, Zhao L, Chen J. Detecting fa leptin receptor mutation in Zucker rats with tetra-primer amplification-refractory mutation system (ARMS)-PCR. Heliyon 2023; 9:e20159. [PMID: 37809507 PMCID: PMC10559934 DOI: 10.1016/j.heliyon.2023.e20159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the genetic mutation (fa) in the gene encoding for leptin receptor, homozygous Zucker rats (fa-/-) develop excessive adiposity and become an experimental animal model in obesity and metabolic-related diseases research. Based on tetra-primer amplification refractory mutation system-polymerase chain reaction (ARMS-PCR), we developed a method to quickly genotype Zucker rats with a mutated fa allele from their wildtype littermates. The three genotypes are clearly discriminated on 2.0% agarose gel. Our method can be used as a reliable tool to set up and maintain the breeding colony in animal facilities as well as assign animals to control and treatment groups based on their genotypes for animal studies.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xinge Hu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guodong Ma
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Tiannan Wang
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jayne Wu
- Department of Electrical Engineering and Computer Science, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xiaojuan Zhu
- Office of Information Technology, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guoxun Chen
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee Knoxville, TN, 37996, United States
| |
Collapse
|
7
|
Doyle ME, Premathilake HU, Yao Q, Mazucanti CH, Egan JM. Physiology of the tongue with emphasis on taste transduction. Physiol Rev 2023; 103:1193-1246. [PMID: 36422992 PMCID: PMC9942923 DOI: 10.1152/physrev.00012.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The tongue is a complex multifunctional organ that interacts and senses both interoceptively and exteroceptively. Although it is easily visible to almost all of us, it is relatively understudied and what is in the literature is often contradictory or is not comprehensively reported. The tongue is both a motor and a sensory organ: motor in that it is required for speech and mastication, and sensory in that it receives information to be relayed to the central nervous system pertaining to the safety and quality of the contents of the oral cavity. Additionally, the tongue and its taste apparatus form part of an innate immune surveillance system. For example, loss or alteration in taste perception can be an early indication of infection as became evident during the present global SARS-CoV-2 pandemic. Here, we particularly emphasize the latest updates in the mechanisms of taste perception, taste bud formation and adult taste bud renewal, and the presence and effects of hormones on taste perception, review the understudied lingual immune system with specific reference to SARS-CoV-2, discuss nascent work on tongue microbiome, as well as address the effect of systemic disease on tongue structure and function, especially in relation to taste.
Collapse
Affiliation(s)
- Máire E Doyle
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hasitha U Premathilake
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Qin Yao
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Caio H Mazucanti
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
8
|
Tawfik MK, Badran DI, Keshawy MM, Makary S, Abdo M. Alternate-day fat diet and exenatide modulate the brain leptin JAK2/STAT3/SOCS3 pathway in a fat diet-induced obesity and insulin resistance mouse model. Arch Med Sci 2023; 19:1508-1519. [PMID: 37732053 PMCID: PMC10507768 DOI: 10.5114/aoms/158534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/24/2022] [Indexed: 09/22/2023] Open
Abstract
Introduction Obesity is one of the most burdensome health problems and is closely linked to leptin resistance. The study examined whether an alternate-day high-fat diet (ADF) and/or GLP-1 agonist (exenatide) modulate brain leptin resistance caused by a high-fat diet (HFD). Material and methods Sixty adult male mice were divided into 6 groups: (i) normal palatable diet (NPD), (ii) exenatide control (NPD received exenatide) (iii) HFD, (iv) ADF treated, (v) exenatide treated, (vi) ADF and exenatide treated. All animal groups were fed a HFD for 8 weeks, before they received treatment (ADF and/or exenatide) for 8 additional weeks. Body weight was assessed at the start and at the end of the experiment. Lipid profile, brain leptin and its receptor expression with the leptin-sensitive pathway, JAK2/STAT3/SOCS3/PTP1B, fasting blood glucose (FBG), serum insulin, liver metabolic handling via its regulators IRS1/PI3K/GLUT4 for hyperinsulinemia/obesity-induced PDK3/NAFLD2 modification, and liver enzymes were determined at the end of the experiment. Results ADF and exenatide reduced body weight and FBG in HFD-obese mice (p < 0.05). The combined ADF and exenatide regimen enhanced the brain anorexic leptin/JAK2/STAT3 and attenuated the SOCS3/PTP1B pathway (p < 0.05). The ADF/exenatide anorexigenic brain effect also modulated liver glucose via IRS1/PI3K/GLUT4 expression (p < 0.05), attenuating NAFLD2 and PDK3 expression (p < 0.05). Liver enzymes and the histopathological profile confirmed the improvement. Conclusions In HFD caloric consumption, a combination of ADF and GLP-1 agonist enhances the brain leptin anorexigenic effect with the improvement of the metabolic sequelae of hyperinsulinemia, hyperlipidemia and liver steatosis.
Collapse
Affiliation(s)
- Mona K. Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Dahlia I. Badran
- Department of Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Department of Biochemistry, Faculty of Medicine, Badr University, Cairo, Egypt
| | - Mohammed M. Keshawy
- Department of Internal Medicine, Nephrology Division, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Samy Makary
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mohamed Abdo
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
9
|
Liu J, Lai F, Hou Y, Zheng R. Leptin signaling and leptin resistance. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:363-384. [PMID: 37724323 PMCID: PMC10388810 DOI: 10.1515/mr-2022-0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/12/2022] [Indexed: 09/20/2023]
Abstract
With the prevalence of obesity and associated comorbidities, studies aimed at revealing mechanisms that regulate energy homeostasis have gained increasing interest. In 1994, the cloning of leptin was a milestone in metabolic research. As an adipocytokine, leptin governs food intake and energy homeostasis through leptin receptors (LepR) in the brain. The failure of increased leptin levels to suppress feeding and elevate energy expenditure is referred to as leptin resistance, which encompasses complex pathophysiological processes. Within the brain, LepR-expressing neurons are distributed in hypothalamus and other brain areas, and each population of the LepR-expressing neurons may mediate particular aspects of leptin effects. In LepR-expressing neurons, the binding of leptin to LepR initiates multiple signaling cascades including janus kinase (JAK)-signal transducers and activators of transcription (STAT) phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), extracellular regulated protein kinase (ERK), and AMP-activated protein kinase (AMPK) signaling, etc., mediating leptin actions. These findings place leptin at the intersection of metabolic and neuroendocrine regulations, and render leptin a key target for treating obesity and associated comorbidities. This review highlights the main discoveries that shaped the field of leptin for better understanding of the mechanism governing metabolic homeostasis, and guides the development of safe and effective interventions to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Futing Lai
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing 100191, China
| |
Collapse
|
10
|
Ghosh-Swaby OR, Reichelt AC, Sheppard PAS, Davies J, Bussey TJ, Saksida LM. Metabolic hormones mediate cognition. Front Neuroendocrinol 2022; 66:101009. [PMID: 35679900 DOI: 10.1016/j.yfrne.2022.101009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Recent biochemical and behavioural evidence indicates that metabolic hormones not only regulate energy intake and nutrient content, but also modulate plasticity and cognition in the central nervous system. Disruptions in metabolic hormone signalling may provide a link between metabolic syndromes like obesity and diabetes, and cognitive impairment. For example, altered metabolic homeostasis in obesity is a strong determinant of the severity of age-related cognitive decline and neurodegenerative disease. Here we review the evidence that eating behaviours and metabolic hormones-particularly ghrelin, leptin, and insulin-are key players in the delicate regulation of neural plasticity and cognition. Caloric restriction and antidiabetic therapies, both of which affect metabolic hormone levels can restore metabolic homeostasis and enhance cognitive function. Thus, metabolic hormone pathways provide a promising target for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Olivia R Ghosh-Swaby
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada
| | - Amy C Reichelt
- Faculty of Health and Medical Sciences, Adelaide Medical School, Adelaide, Australia
| | - Paul A S Sheppard
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Jeffrey Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Timothy J Bussey
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Lisa M Saksida
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
11
|
Zhu F, Zhang D, Shen F, Xu K, Huang X, Liu J, Zhang J, Teng Y. Maternal Socs3 knockdown attenuates postnatal obesity caused by an early life environment of maternal obesity and intrauterine overnutrition in progeny mice. IUBMB Life 2021; 73:1210-1221. [PMID: 34184397 DOI: 10.1002/iub.2526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/29/2021] [Accepted: 06/20/2021] [Indexed: 11/07/2022]
Abstract
Pathological states in the early life environment of mammalian offspring, including maternal obesity and intrauterine overnutrition, can induce obesity and metabolic disorder later in life. Leptin resistance caused by upregulation of Socs3 in the hypothalamus of offspring was believed to be the main mechanism of this effect. In this study, obese mother (OM) and lean mother (LM) models were generated by feeding C57BL/6N female mice a high-fat diet or standard lean diet, respectively. Additionally, an obese mother with intervention (OMI) model was generated by injecting the high-fat diet group with Socs3-shRNA lentivirus during early pregnancy. The offspring of the groups was correspondingly named OM-F1 , LM-F1 , and OMI-F1 , representing progeny mouse models of different early life environments. The offspring were fed a high-fat diet to test their propensity for obesity. The body weight, food intake and fat accumulation were higher, while glucose intolerance and insulin resistance were worse in the OM-F1 group than LM-F1 group. By contrast, the obesity phenotype, hyperphagia and metabolic disorder were alleviated in the OMI-F1 group compared with the OM-F1 group. The mechanism was identified that downregulation of hypothalamic SOCS3 resulted in an increased level of p-STAT3 and p-JAK2, which ameliorated the leptin resistance and restored the lean expression of appetite regulatory genes (Pomc and Agrp) in hypothalamus of OMI-F1 group. Taken together, these results indicate that reducing maternal Socs3 expression during pregnancy can attenuate obesity caused by the early life environment in mice, which may inspire therapies that enable obese mothers to bear metabolically healthy children.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Graduate, Bengbu Medical College, Bengbu, China
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Dawei Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Fangfang Shen
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ke Xu
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xin Huang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Jue Liu
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jin Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Yiqun Teng
- Department of Graduate, Bengbu Medical College, Bengbu, China
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
12
|
SRISUKSAI K, PARUNYAKUL K, PHAONAKROP N, ROYTAKUL S, FUNGFUANG W. The effect of cordycepin on brain oxidative stress and protein expression in streptozotocin-induced diabetic mice. J Vet Med Sci 2021; 83:1425-1434. [PMID: 34334512 PMCID: PMC8498841 DOI: 10.1292/jvms.21-0268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022] Open
Abstract
Diabetes mellitus (DM) is characterized by metabolic disorders and psychological deficits, including cognitive decline. Here, we investigated the effect of cordycepin on oxidative stress and protein expression in the brains of diabetic mice. Twenty-four mice were divided into four groups, one comprising untreated healthy mice (N); one comprising healthy mice treated with cordycepin (24 mg/kg body weight) (N+Cor); one comprising untreated DM mice; and one comprising DM mice treated with cordycepin (24 mg/kg body weight) (DM+Cor). After 14 days of treatment, cognitive behavior was assessed using the novel object recognition (NOR) test. The brain levels of oxidative stress markers (glutathione, catalase, and superoxide dismutase) were examined using the respective detection kits. Protein expression in brain tissues was assessed by liquid chromatography with tandem mass spectrometry (LC-MS/MS); the functions of the identified proteins were annotated by PANTHER, while major protein-protein interactions were assessed using STITCH. We found that cordycepin treatment significantly decreased body weight and food and water intake in the DM+Cor group compared with that in the DM group; however, no differences in blood glucose levels were found between the two groups. Cordycepin treatment significantly reversed cognitive decline in diabetic mice in the NOR test and ameliorated antioxidant defenses. Additionally, we identified ULK1 isoform 2, a protein associated with cognitive function via the activated AMPK and autophagic pathways, as being uniquely expressed in the DM+Cor group. Our findings provide novel insights into the cellular mechanisms underlying how cordycepin improves cognitive decline in diabetic mice.
Collapse
Affiliation(s)
- Krittika SRISUKSAI
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Kongphop PARUNYAKUL
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Narumon PHAONAKROP
- Functional Ingredient and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology
Development Agency, Pathum Thani 12120, Thailand
| | - Sittiruk ROYTAKUL
- Functional Ingredient and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology
Development Agency, Pathum Thani 12120, Thailand
| | - Wirasak FUNGFUANG
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food and Health, Kasetsart University (OmiKU), Bangkok 10900, Thailand
| |
Collapse
|
13
|
Decoding signaling pathways involved in prolactin-induced neuroprotection: A review. Front Neuroendocrinol 2021; 61:100913. [PMID: 33766566 DOI: 10.1016/j.yfrne.2021.100913] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 11/23/2022]
Abstract
It has been well recognized that prolactin (PRL), a pleiotropic hormone, has many functions in the brain, such as maternal behavior, neurogenesis, and neuronal plasticity, among others. Recently, it has been reported to have a significant role in neuroprotection against excitotoxicity. Glutamate excitotoxicity is a common alteration in many neurological and neurodegenerative diseases, leading to neuronal death. In this sense, several efforts have been made to decrease the progression of these pathologies. Despite various reports of PRL's neuroprotective effect against excitotoxicity, the signaling pathways that underlie this mechanism remain unclear. This review aims to describe the most recent and relevant studies on the molecular signaling pathways, particularly, PI3K/AKT, NF-κB, and JAK2/STAT5, which are currently under investigation and might be implicated in the molecular mechanisms that explain the PRL effects against excitotoxicity and neuroprotection. Remarkable neuroprotective effects of PRL might be useful in the treatment of some neurological diseases.
Collapse
|
14
|
In silico analysis of non-coding RNAs and putative target genes implicated in metabolic syndrome. Comput Biol Med 2021; 130:104229. [PMID: 33516961 DOI: 10.1016/j.compbiomed.2021.104229] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/12/2023]
Abstract
Regulation of gene expression is vital to maintain normal cellular functions and its dysregulation leads to molecular pathogenesis of many diseases and disorders. Non-coding RNAs regulate the expression of approximately 60% of protein-coding genes and their malfunction contribute to the development of numerous diseases. The involvement of variant forms of circulating non-coding RNAs in diseases has been established. However, their function as biomarkers or therapeutic targets in metabolic disorders are underexploited. The aim of this study was to predict therapeutic targets and construction of biomarker panel for early detection of metabolic syndrome (MS). Non-coding RNAs including circular RNAs (circRNAs), long chain non-coding RNAs (lncRNA) and micro RNAs (miRNAs) were extracted from intensive literature search and experimentally supported databases. Raw data of gene expression profiles of MS were obtained from the GEO dataset and analyzed to get differentially expressed genes (DEGs). Functional enrichment analysis, network illustration of non-coding RNAs and predicted target DEGs were performed. Furthermore, a few numbers of miRNAs targeted DEGs were subjected to homology study. The strong association of hsa-miR-548c-3p, hsa-miR-579-3p, hsa-miR-17-5p and hsa-miR-320a was observed with the pathogenesis of MS. It includes the regulation of genes in glucose and lipid homeostasis, MAPKK activity, regulation of inflammatory responses and many signaling pathways such as insulin resistance, JAK/STAT and mTOR. Finally, interactions of hsa-miR-17-5p:STAT3, hsa-miR-320:JAK2, hsa-miR-320:S6K and hsa-let-7:DVL hybrids were predicted. Results of this study suggest the designing of a biomarker panel to detect early onset and molecular approach for the management of MS.
Collapse
|
15
|
Contreras-Chavez GG, Estrada JA, Contreras I. Changes in Appetite Regulation-Related Signaling Pathways in the Brain of Mice Supplemented with Non-nutritive Sweeteners. J Mol Neurosci 2020; 71:1144-1155. [PMID: 33128194 DOI: 10.1007/s12031-020-01737-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Non-nutritive sweeteners (NNSs) are commonly used to prevent weight gain and development of metabolic diseases associated with consumption of high-energy diets. Recent studies have demonstrated that these compounds may have unwanted detrimental effects under specific circumstances in vivo. In particular, an association between NNS consumption and changes in signaling pathways involved in the hunger-satiety system in the brain has been reported. Nonetheless, the extent of alterations in brain signaling pathways associated with consumption of these compounds has not been determined. The objective of this study was to determine the effect of frequent consumption of NNSs on the expression of proteins involved in signaling pathways related to appetite control in the brain in vivo. Eight-week-old BALB/c mice were supplemented with sucrose, sucralose, or steviol glycosides in their daily drinking water for 6 weeks. Subsequently, total brain protein extracts were used to analyze the expression of total and phosphorylated JAK2, STAT5, ERK 1/2, JNK, as well as SHP-2 and POMC, by western blot. Serum concentrations of leptin and α-MSH were quantified by ELISA. Results show that consumption of NNSs promotes significant changes in these signaling pathways, reducing the expression of pSTAT5/STAT5, pERK 1/2, SHP-2, and pJNK/JNK in male mice supplemented with steviol glycosides. Furthermore, consumption of steviol glycosides induced a decrease of α-MSH in male mice. In contrast, steviol glycosides induced overexpression of pSTAT5, pERK, and SHP-2 in females. These data suggest that chronic consumption of NNSs promotes sex-specific changes in signaling pathways related to the central hunger-satiety system in vivo.
Collapse
Affiliation(s)
- Gerson G Contreras-Chavez
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico
| | - José A Estrada
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico
| | - Irazú Contreras
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico.
| |
Collapse
|
16
|
Epigenetic regulation of POMC; implications for nutritional programming, obesity and metabolic disease. Front Neuroendocrinol 2019; 54:100773. [PMID: 31344387 DOI: 10.1016/j.yfrne.2019.100773] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/07/2023]
Abstract
Proopiomelanocortin (POMC) is a key mediator of satiety. Epigenetic marks such as DNA methylation may modulate POMC expression and provide a biological link between early life exposures and later phenotype. Animal studies suggest epigenetic marks at POMC are influenced by maternal energy excess and restriction, prenatal stress and Triclosan exposure. Postnatal factors including energy excess, folate, vitamin A, conjugated linoleic acid and leptin may also affect POMC methylation. Recent human studies suggest POMC DNA methylation is influenced by maternal nutrition in early pregnancy and associated with childhood and adult obesity. Studies in children propose a link between POMC DNA methylation and elevated lipids and insulin, independent of body habitus. This review brings together evidence from animal and human studies and suggests that POMC is sensitive to nutritional programming and is associated with a wide range of weight-related and metabolic outcomes.
Collapse
|
17
|
Tsai CF, Chen JH, Wu CT, Chang PC, Wang SL, Yeh WL. Induction of osteoclast-like cell formation by leptin-induced soluble intercellular adhesion molecule secreted from cancer cells. Ther Adv Med Oncol 2019; 11:1758835919846806. [PMID: 31205504 PMCID: PMC6535721 DOI: 10.1177/1758835919846806] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 03/13/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Leptin is considered a tumorigenic adipokine, suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported as proportional to cancer stage and considered as a potential diagnosis biomarker. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. Material and methods: The expression of ICAM-1 and its regulatory signaling were examined by Western blot or flow cytometry. The effect of soluble ICAM-1 on osteoclast formation was investigated by tartrate-resistance acid phosphatase staining of RAW cells and tumor-induced osteolysis in vivo. Results: In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αβ signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with the receptor activator of nuclear factor kappa-B ligand (RANKL) in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. Conclusion: These findings suggest that soluble ICAM-1 produced under leptin treatment enhances osteoclast formation and is involved in tumor-induced osteolysis. Leptin plays an important role in physiology in health and diseases. Leptin affects immune responses that may induce inflammation and carcinogenesis. Leptin is also considered as a tumorigenic adipokine suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported to be proportional to cancer stage and considered as a potential diagnosis biomarker. It has been reported that soluble ICAM-1 allows tumor cells to escape from immune recognition and stimulates angiogenesis and tumor growth. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αβ signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with receptor activator of nuclear factor-kappa B ligand in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. These findings suggest that soluble ICAM-1 produced under leptin treatment is possibly involved in lung and breast cancer bone metastasis.
Collapse
Affiliation(s)
- Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, China
| | - Jia-Hong Chen
- Department of General Surgery, Buddhist Tzu Chi Medical Foundation, Taichung, China
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, Taichung, China
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, China
| | - Shu-Lin Wang
- Institute of New Drug Development, China Medical University, Taichung, China
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402 China
| |
Collapse
|
18
|
Gustafson P, Ladyman SR, Brown RSE. Suppression of Leptin Transport Into the Brain Contributes to Leptin Resistance During Pregnancy in the Mouse. Endocrinology 2019; 160:880-890. [PMID: 30840056 DOI: 10.1210/en.2018-01065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 02/27/2019] [Indexed: 11/19/2022]
Abstract
During pregnancy, when both food intake and circulating leptin concentrations increase, the brain becomes insensitive to leptin. The mechanism by which central leptin resistance during pregnancy emerges remains poorly understood. We investigated whether structural changes in the blood-brain barrier (BBB) or changes in carrier-mediated transport of leptin into the brain might contribute to pregnancy-induced leptin resistance. Immunohistochemical evaluation of the BBB at the level of the arcuate nucleus and median eminence in virgin, pregnant, and lactating mice was undertaken by labeling for tanycytes (vimentin), tight junction protein (zona occludens-1), and a marker of fenestrated endothelial capillaries (MECA-32). There were no changes in these BBB markers between virgin, pregnant or lactating mice. Transport of iodine 125-labeled leptin from the peripheral circulation into the brain was completely suppressed during pregnancy, however (days 14 through 16), compared with virgin and lactating (days 7 through 11) mice. This was accompanied by a suppression of leptin clearance from the blood in pregnant mice. We also investigated in virgin mice whether competition with other hormones for transport might contribute to suppression of leptin transport into the brain. Although leptin was able to compete with prolactin transport into the brain, prolactin did not compete with leptin transport. These data demonstrate that suppression of the transport of leptin into the brain during pregnancy, in the absence of structural changes in the BBB, is an important contributor to the insensitivity of the hypothalamus to leptin at this time.
Collapse
Affiliation(s)
- Papillon Gustafson
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Rosemary S E Brown
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Neuronal Signal Transduction-Involved Genes in Pig Hypothalamus Affect Feed Efficiency as Revealed by Transcriptome Analysis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5862571. [PMID: 30687750 PMCID: PMC6327278 DOI: 10.1155/2018/5862571] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023]
Abstract
Feed efficiency (FE) is an important trait affecting costs in swine industry. Investigation on FE-related genes in different tissues is valuable for molecular breeding. Hypothalamus is a convergent and integrated centre for multiple nutrient-related signals. The present study identified 363 differentially expressed (DE) genes and 14 DE lincRNAs in the hypothalamus of high- and low-FE Yorkshire pigs. Furthermore, 983 significantly correlated DE gene-lincRNA pairs were identified through weighted correlation network analysis (WGCNA) and Pearson correlation analysis. These DE genes were primarily enriched in the neuronal signal transduction process containing the upregulated genes of VIPR1, CCR1, CCR5, LEPR, INSR, ADRA1A, CCKAR, and ADORA3 and the downregulated genes of GRM1, GRM4, GRM5, and VIPR2, which were located in the cell membrane. These signal receptors were mainly connected to downstream Jak-STAT signaling that involved the increased genes (JAK2, STAT3, and POMC) and mTOR signaling pathway, including the decreased genes (CAMKK2, AMPK, and MTOR). STAT3 and AMPK genes also played a role in two major hypothalamic neurons of POMC and NPY/AGRP. A total of eight DE lincRNAs also participated in the potential network. In conclusion, neuronal signaling transduction-involved genes and lincRNAs were related to FE variation in pig hypothalamus.
Collapse
|
20
|
Jin S, Diano S. Mitochondrial Dynamics and Hypothalamic Regulation of Metabolism. Endocrinology 2018; 159:3596-3604. [PMID: 30203064 PMCID: PMC6157417 DOI: 10.1210/en.2018-00667] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/02/2018] [Indexed: 01/22/2023]
Abstract
Mitochondria are cellular organelles that play an important role in bioenergetic processes. In the central nervous system, high energy-demanding neurons are critically dependent on mitochondria to fulfill their appropriate functions. The hypothalamus is a key brain area for maintaining glucose and energy homeostasis via the ability of hypothalamic neurons to sense, integrate, and respond to numerous metabolic signals. Mitochondrial function has emerged as an important component in the regulation of hypothalamic neurons controlling glucose and energy homeostasis. Although the underlying mechanisms are not fully understood, emerging evidence indicates that mitochondrial dysfunction in hypothalamic neurons may contribute to the development of various metabolic diseases, including obesity and type 2 diabetes mellitus (T2DM). In this review, we summarize recent studies demonstrating the link between mitochondria and hypothalamic neural control of glucose and energy homeostasis. Finally, this review provides an insight to understand how mitochondria in hypothalamic neurons may contribute to the development of metabolic disorders, such as T2DM and obesity.
Collapse
Affiliation(s)
- Sungho Jin
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Clinical Medicine and Surgery, University of Naples “Federico II,” Naples, Italy
| |
Collapse
|
21
|
Qurania KR, Ikeda K, Wardhana DA, Barinda AJ, Nugroho DB, Kuribayashi Y, Rahardini EP, Rinastiti P, Ryanto GRT, Yagi K, Hirata KI, Emoto N. Systemic inhibition of Janus kinase induces browning of white adipose tissue and ameliorates obesity-related metabolic disorders. Biochem Biophys Res Commun 2018; 502:123-128. [DOI: 10.1016/j.bbrc.2018.05.131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 01/25/2023]
|
22
|
Haissaguerre M, Ferriere A, Clark S, Guzman-Quevedo O, Tabarin A, Cota D. NPV-BSK805, an Antineoplastic Jak2 Inhibitor Effective in Myeloproliferative Disorders, Causes Adiposity in Mice by Interfering With the Action of Leptin. Front Pharmacol 2018; 9:527. [PMID: 29867515 PMCID: PMC5962752 DOI: 10.3389/fphar.2018.00527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/02/2018] [Indexed: 11/23/2022] Open
Abstract
The pathophysiology of body weight gain that is observed in patients suffering from myeloproliferative neoplasms treated with inhibitors of the janus kinase (Jak) 1 and 2 pathway remains unknown. Here we hypothesized that this class of drugs interferes with the metabolic actions of leptin, as this hormone requires functional Jak2 signaling. To test this, C57BL/6J chow-fed mice received either chronic intraperitoneal (ip) or repeated intracerebroventricular (icv) administration of the selective Jak2 inhibitor NVP-BSK805, which was proven efficacious in treating polycythemia in rodents. Changes in food intake, body weight and body composition were recorded. Icv NVP-BSK805 was combined with ip leptin to evaluate ability to interfere with the action of this hormone on food intake and on induction of hypothalamic phosphorylation of signal transducer and activator of transcription 3 (STAT3). We found that chronic peripheral administration of NVP-BSK805 did not alter food intake, but increased fat mass and feed efficiency. The increase in fat mass was more pronounced during repeated icv administration of the compound, suggesting that metabolic effects were related to molecular interference in brain structures regulating energy balance. Accordingly, acute icv administration of NVP-BSK805 prevented the ability of leptin to decrease food intake and body weight by impeding STAT3 phosphorylation within the hypothalamus. Consequently, acute icv administration of NVP-BSK805 at higher dose induced hyperphagia and body weight gain. Our results provide evidence for a specific anabolic effect exerted by antineoplastic drugs targeting the Jak2 pathway, which is due to interference with the actions of leptin. Consequently, assessment of metabolic variables related to increased fat mass gain should be performed in patients treated with Jak2 inhibitors.
Collapse
Affiliation(s)
- Magalie Haissaguerre
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Amandine Ferriere
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
| | - Samantha Clark
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
| | - Omar Guzman-Quevedo
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Facultad de Químico-Farmacobiología, Universidad Michoacána de San Nicolás de Hidalgo, Morelia, Mexico
| | - Antoine Tabarin
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- Service d’Endocrinologie, Diabétologie et Nutrition, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- *Correspondence: Antoine Tabarin, Daniela Cota,
| |
Collapse
|
23
|
Guénard F, Bouchard-Mercier A, Rudkowska I, Lemieux S, Couture P, Vohl MC. Genome-Wide Association Study of Dietary Pattern Scores. Nutrients 2017; 9:E649. [PMID: 28644415 PMCID: PMC5537769 DOI: 10.3390/nu9070649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 01/08/2023] Open
Abstract
Dietary patterns, representing global food supplies rather than specific nutrients or food intakes, have been associated with cardiovascular disease (CVD) incidence and mortality. The contribution of genetic factors in the determination of food intakes, preferences and dietary patterns has been previously established. The current study aimed to identify novel genetic factors associated with reported dietary pattern scores. Reported dietary patterns scores were derived from reported dietary intakes for the preceding month and were obtained through a food frequency questionnaire and genome-wide association study (GWAS) conducted in a study sample of 141 individuals. Reported Prudent and Western dietary patterns demonstrated nominal associations (p < 1 × 10-5) with 78 and 27 single nucleotide polymorphisms (SNPs), respectively. Among these, SNPs annotated to genes previously associated with neurological disorders, CVD risk factors and obesity were identified. Further assessment of SNPs demonstrated an impact on gene expression levels in blood for SNPs located within/near BCKDHB (p = 0.02) and the hypothalamic glucosensor PFKFB3 (p = 0.0004) genes, potentially mediated through an impact on the binding of transcription factors (TFs). Overrepresentations of glucose/energy homeostasis and hormone response TFs were also observed from SNP-surrounding sequences. Results from the current GWAS study suggest an interplay of genes involved in the metabolic response to dietary patterns on obesity, glucose metabolism and food-induced response in the brain in the adoption of dietary patterns.
Collapse
Affiliation(s)
- Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), School of Nutrition, Laval University, Québec, QC G1V 0A6, Canada.
| | - Annie Bouchard-Mercier
- Institute of Nutrition and Functional Foods (INAF), School of Nutrition, Laval University, Québec, QC G1V 0A6, Canada.
| | - Iwona Rudkowska
- Endocrinology and Nephrology Unit, Centre de recherche du CHU de Québec, Laval University, Québec, QC G1V 4G2, Canada.
| | - Simone Lemieux
- Institute of Nutrition and Functional Foods (INAF), School of Nutrition, Laval University, Québec, QC G1V 0A6, Canada.
| | - Patrick Couture
- Institute of Nutrition and Functional Foods (INAF), Endocrinology and Nephrology Unit, Centre de recherche du CHU de Québec, Laval University, Québec, QC G1V 4G2, Canada.
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), School of Nutrition, Laval University, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
24
|
Russell AL, Grimes JM, Cruthirds DF, Westerfield J, Wooten L, Keil M, Weiser MJ, Landauer MR, Handa RJ, Wu TJ, Larco DO. Dietary Isoflavone-Dependent and Estradiol Replacement Effects on Body Weight in the Ovariectomized (OVX) Rat. Horm Metab Res 2017; 49:457-465. [PMID: 28482370 PMCID: PMC5820000 DOI: 10.1055/s-0043-108250] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
17β-Estradiol is known to regulate energy metabolism and body weight. Ovariectomy results in body weight gain while estradiol administration results in a reversal of weight gain. Isoflavones, found in rodent chow, can mimic estrogenic effects making it crucial to understand the role of these compounds on metabolic regulation. The goal of this study is to examine the effect of dietary isoflavones on body weight regulation in the ovariectomized rat. This study will examine how dietary isoflavones can interact with estradiol treatment to affect body weight. Consistent with previous findings, animals fed an isoflavone-rich diet had decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin levels (p<0.05) compared to animals fed an isoflavone-free diet. Estradiol replacement resulted in decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin (p<0.05). Current literature suggests the involvement of cytokines in the inflammatory response of body weight gain. We screened a host of cytokines and chemokines that may be altered by dietary isoflavones or estradiol replacement. Serum cytokine analysis revealed significant (p<0.05) diet-dependent increases in inflammatory cytokines (keratinocyte-derived chemokine). The isoflavone-free diet in OVX rats resulted in the regulation of the following cytokines and chemokines: interleukin-10, interleukin-18, serum regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 (p<0.05). Overall, these results reveal that estradiol treatment can have differential effects on energy metabolism and body weight regulation depending on the presence of isoflavones in rodent chow.
Collapse
Affiliation(s)
- Ashley L. Russell
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jamie Moran Grimes
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Danette F. Cruthirds
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joanna Westerfield
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lawren Wooten
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Michael J. Weiser
- DSM Nutritional Products Inc., Human Nutrition & Health, Boulder, Colorado, USA
| | - Michael R. Landauer
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert J. Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - T. John Wu
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Darwin O. Larco
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
25
|
Adiponectin deficiency rescues high-fat diet-induced hepatic injury, apoptosis and autophagy loss despite persistent steatosis. Int J Obes (Lond) 2017; 41:1403-1412. [PMID: 28559541 DOI: 10.1038/ijo.2017.128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/01/2017] [Accepted: 05/16/2017] [Indexed: 02/06/2023]
Abstract
Background &aims:Low levels of adiponectin (APN), an adipose-derived adipokine, are associated with obesity and non-alcoholic steatohepatitis although its role in high-fat diet-induced hepatic injury and steatosis remains unclear. Here we hypothesized that APN deficiency alters fat diet-induced hepatic function. To this end, we examined the effect of APN deficiency on high-fat diet-induced hepatic injury, apoptosis and steatosis. METHODS Adult wild type and APN knockout mice were fed a low- or high-fat diet for 20 weeks. Serum levels of liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), cholesterol, hepatic triglycerides, steatosis, pro-inflammatory cytokines, apoptosis and autophagy were examined. RESULTS High-fat feeding led to elevated body (48.2%) and liver weights (18.8%), increased levels of ALT (87.8%), serum cholesterol (104.4%), hepatic triglycerides (305.6%) and hepatic fat deposition as evidenced by Oil Red O staining, along with a reduced AST/ALT ratio and unchanged AST. Although APN knockout itself did not affect hepatic function and morphology, it reconciled fat diet-induced hepatic injury (P<0.05 vs WT-HF group) without reversing changes in body and liver weights, serum cholesterol and hepatic steatosis. In addition, fat diet intake promoted AMPK phosphorylation, p62 accumulation and apoptosis, including elevated Bax and cleaved Caspase-3 and downregulated Bcl-2, along with suppressed phosphorylation of Akt, STAT3 and JNK, and the autophagy makers Atg7, Beclin-1 and LC3B (P<0.05 vs WT-LF group) without affecting hepatic interlelukin-6 and tumor necrosis factor-α levels, the effects were reversed or significantly attenuated by APN knockout (P<0.05 vs WT-HF group). In vitro study using HepG2 cells revealed that STAT3 activation rescued palmitic acid-induced cell injury whereas STAT3 inhibition nullified APN knockdown-offered beneficial effects. CONCLUSIONS Our results revealed that high-fat diet intake promotes hepatic steatosis, apoptosis and interrupted autophagy. APN knockout elicits protective effect against hepatic injury possibly associated with autophagy regulation despite persistent hepatic steatosis.
Collapse
|
26
|
Jia Y, Liu T, Zhou L, Zhu J, Wu J, Sun D, Xu J, Wang Q, Chen H, Xu F, Zhang Y, Zhang T, Liu H, Ye L. Effects of Di-(2-ethylhexyl) Phthalate on Lipid Metabolism by the JAK/STAT Pathway in Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13111085. [PMID: 27827939 PMCID: PMC5129295 DOI: 10.3390/ijerph13111085] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/20/2016] [Accepted: 11/01/2016] [Indexed: 02/07/2023]
Abstract
The most widely used plasticizer, di-(2-ethylhexyl) phthalate (DEHP), is known to affect lipid metabolism and adipogenesis. We studied the effects of dietary DEHP exposure on metabolism in rats as well as the role of the JAK/STAT pathway in this process. Eighty rats were exposed to DEHP (0, 5, 50 and 500 mg/kg/d) through dietary intake for 4 weeks. We then collected blood samples, liver, and adipose tissues to detect modifications in the levels of serum lipids, leptin, adiponectin and insulin. JAK3, STAT5a and PPARγ expression were detected at both the gene and protein levels. The activation of JAK3 and STAT5a was also detected. The DEHP-exposed rats had increased body weight, serum lipid, insulin, and leptin levels. Moreover, the JAK3/STAT5a pathway was activated in the adipose tissue; however, this pathway was not activated in the liver. The mRNA of SREBP-1c in the liver was increased significantly among each of the groups, in contrast to the levels found in the mature SREBP-1c protein form. Furthermore, the expression of FABP4, Acox and FASn was decreased in the liver, but increased in adipose tissue. Thus, we conclude that exposure to DEHP reduces the hydrolysis of lipid and promotes triglyceride accumulation by oppositely regulating the activation state of JAK/STAT pathway in the liver and adipose tissue, resulting in the disorder of body lipid metabolism and obesity.
Collapse
Affiliation(s)
- Yiyang Jia
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Te Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Jian Zhu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Juan Wu
- Cancer Center, Tumor Hospital of Jiangxi Province, Nanchang 330029, China.
| | - Di Sun
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Jin Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Qi Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Huaiji Chen
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Feng Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Yuezhu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Tianrong Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Hongbo Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
27
|
Camer D, Yu Y, Szabo A, Wang H, Dinh CHL, Huang XF. Bardoxolone methyl prevents obesity and hypothalamic dysfunction. Chem Biol Interact 2016; 256:178-87. [PMID: 27417254 DOI: 10.1016/j.cbi.2016.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/18/2016] [Accepted: 07/10/2016] [Indexed: 01/28/2023]
Abstract
High-fat (HF) diet-induced obesity is associated with hypothalamic leptin resistance and low grade chronic inflammation, which largely impairs the neuroregulation of negative energy balance. Neuroregulation of negative energy balance is largely controlled by the mediobasal and paraventricular nuclei regions of the hypothalamus via leptin signal transduction. Recently, a derivative of oleanolic acid, bardoxolone methyl (BM), has been shown to have anti-inflammatory effects. We tested the hypothesis that BM would prevent HF diet-induced obesity, hypothalamic leptin resistance, and inflammation in mice fed a HF diet. Oral administration of BM via drinking water (10 mg/kg daily) for 21 weeks significantly prevented an increase in body weight, energy intake, hyperleptinemia, and peripheral fat accumulation in mice fed a HF diet. Furthermore, BM treatment prevented HF diet-induced decreases in the anorexigenic effects of peripheral leptin administration. In the mediobasal and paraventricular nuclei regions of the hypothalamus, BM administration prevented HF diet-induced impairments of the downstream protein kinase b (Akt) pathway of hypothalamic leptin signalling. BM treatment also prevented an increase in inflammatory cytokines, tumour necrosis factor alpha (TNFα) and interleukin 6 (IL-6) in these two hypothalamic regions. These results identify a potential novel neuropharmacological application for BM in preventing HF diet-induced obesity, hypothalamic leptin resistance, and inflammation.
Collapse
Affiliation(s)
- Danielle Camer
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.
| | - Yinghua Yu
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Alexander Szabo
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia; ANSTO Life Sciences, Australian Nuclear Science and Technology Organisation, NSW, 2234, Australia
| | - Hongqin Wang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Chi H L Dinh
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong and Illawarra Health and Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| |
Collapse
|
28
|
Zimmers TA, Fishel ML, Bonetto A. STAT3 in the systemic inflammation of cancer cachexia. Semin Cell Dev Biol 2016; 54:28-41. [PMID: 26860754 PMCID: PMC4867234 DOI: 10.1016/j.semcdb.2016.02.009] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
Weight loss is diagnostic of cachexia, a debilitating syndrome contributing mightily to morbidity and mortality in cancer. Most research has probed mechanisms leading to muscle atrophy and adipose wasting in cachexia; however cachexia is a truly systemic phenomenon. Presence of the tumor elicits an inflammatory response and profound metabolic derangements involving not only muscle and fat, but also the hypothalamus, liver, heart, blood, spleen and likely other organs. This global response is orchestrated in part through circulating cytokines that rise in conditions of cachexia. Exogenous Interleukin-6 (IL6) and related cytokines can induce most cachexia symptomatology, including muscle and fat wasting, the acute phase response and anemia, while IL-6 inhibition reduces muscle loss in cancer. Although mechanistic studies are ongoing, certain of these cachexia phenotypes have been causally linked to the cytokine-activated transcription factor, STAT3, including skeletal muscle wasting, cardiac dysfunction and hypothalamic inflammation. Correlative studies implicate STAT3 in fat wasting and the acute phase response in cancer cachexia. Parallel data in non-cancer models and disease states suggest both pathological and protective functions for STAT3 in other organs during cachexia. STAT3 also contributes to cancer cachexia through enhancing tumorigenesis, metastasis and immune suppression, particularly in tumors associated with high prevalence of cachexia. This review examines the evidence linking STAT3 to multi-organ manifestations of cachexia and the potential and perils for targeting STAT3 to reduce cachexia and prolong survival in cancer patients.
Collapse
Affiliation(s)
- Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Melissa L Fishel
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
29
|
Gurzov EN, Stanley WJ, Pappas EG, Thomas HE, Gough DJ. The JAK/STAT pathway in obesity and diabetes. FEBS J 2016; 283:3002-15. [PMID: 26972840 DOI: 10.1111/febs.13709] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/14/2016] [Accepted: 03/08/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus are complex, multi-organ metabolic pathologies characterized by hyperglycemia. Emerging evidence shows that the highly conserved and potent JAK/STAT signaling pathway is required for normal homeostasis, and, when dysregulated, contributes to the development of obesity and diabetes. In this review, we analyze the role of JAK/STAT activation in the brain, liver, muscle, fat and pancreas, and how this affects the course of the disease. We also consider the therapeutic implications of targeting the JAK/STAT pathway in treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Esteban N Gurzov
- St Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
| | - William J Stanley
- St Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
| | - Evan G Pappas
- St Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
| | - Helen E Thomas
- St Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
| | - Daniel J Gough
- Hudson Institute of Medical Research, Clayton, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Australia
| |
Collapse
|
30
|
Whitfield JF, Chiarini A, Dal Prà I, Armato U, Chakravarthy B. The Possible Roles of the Dentate Granule Cell's Leptin and Other Ciliary Receptors in Alzheimer's Neuropathology. Cells 2015; 4:253-74. [PMID: 26184316 PMCID: PMC4588035 DOI: 10.3390/cells4030253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/18/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
Dentate-gyral granule cells in the hippocampus plus dentate gyrus memory-recording/retrieving machine, unlike most other neurons in the brain, are continuously being generated in the adult brain with the important task of separating overlapping patterns of data streaming in from the outside world via the entorhinal cortex. This "adult neurogenesis" is driven by tools in the mature granule cell's cilium. Here we report our discovery of leptin's LepRb receptor in this cilium. In addition, we discuss how ciliary LepRb signaling might be involved with ciliary p75NTR and SSTR3 receptors in adult neurogenesis and memory formation as well as attenuation of Alzheimer's neuropathology by reducing the production of its toxic amyloid-β-derived drivers.
Collapse
Affiliation(s)
- James F Whitfield
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada.
| | - Anna Chiarini
- Histology & Embryology Unit, Department of Life & Reproduction Sciences, University of Verona Medical School, 8 Strada Le Grazie, Verona, Venetia 37134, Italy.
| | - Ilaria Dal Prà
- Histology & Embryology Unit, Department of Life & Reproduction Sciences, University of Verona Medical School, 8 Strada Le Grazie, Verona, Venetia 37134, Italy.
| | - Ubaldo Armato
- Histology & Embryology Unit, Department of Life & Reproduction Sciences, University of Verona Medical School, 8 Strada Le Grazie, Verona, Venetia 37134, Italy.
| | - Balu Chakravarthy
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada.
| |
Collapse
|
31
|
Breit A, Besik V, Solinski HJ, Muehlich S, Glas E, Yarwood SJ, Gudermann T. Serine-727 phosphorylation activates hypothalamic STAT-3 independently from tyrosine-705 phosphorylation. Mol Endocrinol 2015; 29:445-59. [PMID: 25584415 DOI: 10.1210/me.2014-1300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transcriptional activity of signal transducer and activator of transcription-3 (STAT-3) is a key element in the central regulation of appetite and energy homeostasis. Activation of hypothalamic STAT-3 has been attributed to cytokine-promoted phosphorylation at tyrosine-705 (Tyr-705). In nonhypothalamic cells, STAT-3 is also phosphorylated at serine-727 (Ser-727), but the functional significance of Ser-727 in the regulation of hypothalamic STAT-3 is not known. We used 2 hypothalamic cell lines and analyzed the effects of various hormones on STAT-3-dependent reporter gene activity and observed that IFN-γ, epidermal growth factor (EGF), and bradykinin (BK) induce similar STAT-3 reporter activation. EGF and BK solely increased Ser-727 and IFN-γ increased Tyr-705 phosphorylation of STAT-3. Specific inhibition of ERK-1/2 activity blocked EGF- and BK-induced STAT-3 activation and Ser-727 phosphorylation. BK-induced ERK-1/2 activation occurred via EGF receptor transactivation. Consequently, the BK-mediated effects on STAT-3 were blocked by a specific EGF receptor antagonist. Next, we analyzed the effects of IFN-γ and EGF on the expression of the STAT-3-dependent genes thyroliberin-releasing hormone and suppressors of cytokine signaling-3. EGF but not IFN-γ enhanced thyroliberin-releasing hormone expression via STAT-3. With regard to suppressors of cytokine signaling-3, we observed prolonged expression induced by IFN-γ and a transient effect of EGF that required coactivation of the activator protein-1. Thus, EGF-promoted Ser-727 phosphorylation by ERK-1/2 is not only sufficient to fully activate hypothalamic STAT-3, but, in terms of targeted genes and required cofactors, entails distinct modes of STAT-3 actions compared with IFN-γ-induced Tyr-705 phosphorylation.
Collapse
Affiliation(s)
- Andreas Breit
- Walther-Straub-Institut für Pharmakologie und Toxikologie (A.B., V.B., H.J.S., S.M., E.G., T.G.), Ludwig-Maximilians-Universität München, München, Germany 80336; and The Institute of Molecular, Cell and Systems Biology (S.J.Y.), College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow GC12 8QQ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The ability of an organism to convert organic molecules from the environment into energy is essential for the development of cellular structures, cell differentiation and growth. Mitochondria have a fundamental role in regulating metabolic pathways, and tight control of mitochondrial functions and dynamics is critical to maintaining adequate energy balance. In complex organisms, such as mammals, it is also essential that the metabolic demands of various tissues are coordinated to ensure that the energy needs of the whole body are effectively met. Within the arcuate nucleus of the hypothalamus, the NPY-AgRP and POMC neurons have a crucial role in orchestrating the regulation of hunger and satiety. Emerging findings from animal studies have revealed an important function for mitochondrial dynamics within these two neuronal populations, which facilitates the correct adaptive responses of the whole body to changes in the metabolic milieu. The main proteins implicated in these studies are the mitofusins, Mfn1 and Mfn2, which are regulators of mitochondrial dynamics. In this Review, we provide an overview of the mechanisms by which mitochondria are involved in the central regulation of energy balance and discuss the implications of mitochondrial dysfunction for metabolic disorders.
Collapse
Affiliation(s)
- Carole M Nasrallah
- Program in Integrative Cell Signalling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, SHM L-200, PO Box 208074, New Haven, CT 06520-8074, USA
| | - Tamas L Horvath
- Program in Integrative Cell Signalling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, SHM L-200, PO Box 208074, New Haven, CT 06520-8074, USA
| |
Collapse
|
33
|
Varco-Merth B, Rotwein P. Differential effects of STAT proteins on growth hormone-mediated IGF-I gene expression. Am J Physiol Endocrinol Metab 2014; 307:E847-55. [PMID: 25205818 PMCID: PMC4216947 DOI: 10.1152/ajpendo.00324.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Growth hormone (GH) plays a key role regulating somatic growth and in controlling metabolism and other physiological processes in humans and other animal species. GH acts by binding to the extracellular part of its transmembrane receptor, leading to induction of multiple intracellular signal transduction pathways that culminate in changes in gene and protein expression. A key agent in GH-stimulated growth is the latent transcription factor signal transducer and activator of transcription (STAT) 5B, one of four STAT proteins induced by the GH receptor in cultured cells and in vivo. As shown by genetic and biochemical studies, GH-activated STAT5B promotes transcription of the gene encoding the critical growth peptide, insulin-like growth factor-I (IGF-I), and natural null mutations of STAT5B in humans lead to growth failure accompanied by diminished IGF-I expression. Here we have examined the possibility that other GH-activated STATs can enhance IGF-I gene transcription, and thus potentially contribute to GH-regulated somatic growth. We find that human STAT5A is nearly identical to STAT5B in its biochemical and functional responses to GH but that STAT1 and STAT3 show a weaker profile of in vitro binding to STAT DNA elements from the IGF-I gene than STAT5B, and are less potent inducers of gene transcription through these elements. Taken together, our results offer a molecular explanation for why STAT5B is a key in vivo mediator of GH-activated IGF-I gene transcription and thus of GH-regulated somatic growth.
Collapse
Affiliation(s)
- Ben Varco-Merth
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|