1
|
Lin F, Gilbertson TA. Fat taste responsiveness, but not dietary fat intake, is affected in Adipor1 null mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642880. [PMID: 40161824 PMCID: PMC11952482 DOI: 10.1101/2025.03.12.642880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Taste is a major driving force that influences food choices and dietary intake. Adiponectin has been shown to selectively enhance cellular responses to fatty acids by mediating the activation of AMPK and translocation of CD36 in taste cells via its receptor AdipoR1. Whether Adipor1 gene knockout affects fat taste responsiveness and dietary fat intake in animals remains unclear. In the present study, we evaluated cellular, neural, and behavioral responses to fat, as well as the dietary fat intake in global Adipor1 knockout mice and their WT controls. Sex-specific changes in cellular and behavioral responses to fatty acid were observed in Adipor1 knockout mice. Linoleic acid (LA)-induced calcium responsiveness appears to be reduced in taste cells from Adipor1 -deficient males and increased in taste cells from Adipor1 -deficient females. Brief-access taste testing revealed a loss of fat taste behavioral responsiveness in naïve Adipor1 -/- animals. Fat taste loss found in Adipor1 -/- males was restored after fat exposure and showed no significant differences in taste behavioral responses to fatty acids with WT controls in two-bottle preference and conditioned taste aversion tests. Adipor1 -/- females were found to have diminished preference for LA in two-bottle preference tests, lower intralipid/water lick ratio in a brief-access assay, and reduced avoidance for LA in conditioned taste aversion assay. Furthermore, the taste nerve responses to intralipid and the dietary fat intakes appeared to be the same between Adipor1 -/- and WT mice. In the high-fat diet feeding study, Adipor1 -/- females gained more weight, while no differences in body weight gain were found in males. Together, we show that adiponectin/AdipoR1 signaling plays crucial sex-specific roles in the modulation of fat taste and the maintenance of healthy body weight primarily by regulating energy expenditure rather than dietary fat intake in mice.
Collapse
|
2
|
Lin Z, Lin J, Huang A, Zhang Z, Wu X, Yin G, Wei C, Xu W. Angiotensin (1-7) Improves Pancreatic Islet Function via Upregulating PDX-1 and GCK: A Dose-Dependent Study in Mice. Int J Endocrinol 2024; 2024:1672096. [PMID: 39734383 PMCID: PMC11671625 DOI: 10.1155/ije/1672096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose: This study aimed to verify the effect of angiotensin (1-7) on improving islet function and further explore the signaling pathway that may be involved in this improvement. It also aimed to explore the effects of angiotensin (1-7) on blood glucose levels, islet function, and morphological changes in db/db mice and its potential signal pathway. Methods: Forty-five db/db mice were divided randomly into a model control group and different doses of angiotensin (1-7) intervention groups (0, 150, 300, and 600 μg/kg/d), while seven db/m mice were assigned as the normal control group. The angiotensin (1-7) intervention groups received daily intraperitoneal administration for 8 weeks, whereas the normal control group was injected intraperitoneally with an equal volume of normal saline every day for 8 weeks. Changes in weight and food intake of mice were detected. Effect of angiotensin (1-7) on lipid metabolism, islet function, the morphology of pancreatic islets, and β-cell mass on mice were evaluated. The expression of PDX-1 and GCK in pancreatic tissue was verified. Results: The group receiving angiotensin (1-7) at a dosage of 600 μg/kg/d showed a significant decrease in body weight, triglyceride levels, and fasting blood glucose, along with an improvement in glucose tolerance. In the 300 μg/kg/d group, angiotensin (1-7) tended to increase the total volume of islets. Moreover, the intervention groups exhibited a significant increase in the ratio of β cells, small islets (30-80 μm in diameter), as well as the expression levels of PDX-1 and GCK in pancreatic tissue. Conclusion: Angiotensin (1-7) could improve glucose and lipid metabolism and islet function by promoting the expression of PDX-1 and GCK genes in the pancreas of db/db mice.
Collapse
Affiliation(s)
- Ziwei Lin
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jiaqi Lin
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Anqi Huang
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zixu Zhang
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xinyi Wu
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guoshu Yin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chiju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, China
| | - Wencan Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
3
|
Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024; 25:5277-5293. [PMID: 39558137 PMCID: PMC11624209 DOI: 10.1038/s44319-024-00321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The central nervous system and adipose tissue interact through complex communication. This bidirectional signaling regulates metabolic functions. The hypothalamus, a key homeostatic brain region, integrates exteroceptive and interoceptive signals to control appetite, energy expenditure, glucose, and lipid metabolism. This regulation is partly achieved via the nervous modulation of white (WAT) and brown (BAT) adipose tissue. In this review, we highlight the roles of sympathetic and parasympathetic innervation in regulating WAT and BAT activities, such as lipolysis and thermogenesis. Adipose tissue, in turn, plays a dual role as an energy reservoir and an endocrine organ, secreting hormones that influence brain function and metabolic health. In addition, this review focuses on recently uncovered communication pathways, including extracellular vesicles and neuro-mesenchymal units, which add new layers of regulation and complexity to the brain-adipose tissue interaction. Finally, we also examine the consequences of disrupted communication between the brain and adipose tissue in metabolic disorders like obesity and type-2 diabetes, emphasizing the potential for new therapeutic strategies targeting these pathways to improve metabolic health.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile
- Physiology Department, Biological Science Faculty, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- IBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Glasbrenner C, Höchsmann C, Pieper CF, Wasserfurth P, Dorling JL, Martin CK, Redman LM, Koehler K. Prediction of individual weight loss using supervised learning: findings from the CALERIE TM 2 study. Am J Clin Nutr 2024; 120:1233-1244. [PMID: 39270937 PMCID: PMC11600119 DOI: 10.1016/j.ajcnut.2024.09.003] [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: 05/17/2024] [Revised: 07/18/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Predicting individual weight loss (WL) responses to lifestyle interventions is challenging but might help practitioners and clinicians select the most promising approach for each individual. OBJECTIVE The primary aim of this study was to develop machine learning (ML) models to predict individual WL responses using only variables known before starting the intervention. In addition, we used ML to identify pre-intervention variables influencing the individual WL response. METHODS We used 12-mo data from the comprehensive assessment of long-term effects of reducing intake of energy (CALERIETM) phase 2 study, which aimed to analyze the long-term effects of caloric restriction on human longevity. On the basis of the data from 130 subjects in the intervention group, we developed classification models to predict binary ("Success" and "No/low success") or multiclass ("High success," "Medium success," and "Low/no success") WL outcomes. Additionally, regression models were developed to predict individual weight change (percent). Models were evaluated on the basis of accuracy, sensitivity, specificity (classification models), and root mean squared error (RMSE; regression models). RESULTS Best classification models used 20-40 predictors and achieved 89%-97% accuracy, 91%-100% sensitivity, and 56%-86% specificity for binary classification. For multiclass classification, accuracy (69%) and sensitivity (50%) tended to be lower. The best regression performance was obtained with 36 variables with an RMSE of 2.84%. Among the 21 variables predicting individual weight change most consistently, we identified 2 novel predictors, namely orgasm satisfaction and sexual behavior/experience. Other common predictors have previously been associated with WL (16) or are already used in traditional prediction models (3). CONCLUSIONS The prediction models could be implemented by practitioners and clinicians to support the decision of whether lifestyle interventions are sufficient or more aggressive interventions are needed for a given individual, thereby supporting better, faster, data-driven, and unbiased decisions. The CALERIETM phase 2 study was registered at clinicaltrials.gov as NCT00427193.
Collapse
Affiliation(s)
- Christina Glasbrenner
- TUM School of Medicine and Health, Department of Health and Sport Sciences, Technical University of Munich, Munich, Germany
| | - Christoph Höchsmann
- TUM School of Medicine and Health, Department of Health and Sport Sciences, Technical University of Munich, Munich, Germany
| | - Carl F Pieper
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Paulina Wasserfurth
- TUM School of Medicine and Health, Department of Health and Sport Sciences, Technical University of Munich, Munich, Germany
| | - James L Dorling
- Human Nutrition, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Corby K Martin
- Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Leanne M Redman
- Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Karsten Koehler
- TUM School of Medicine and Health, Department of Health and Sport Sciences, Technical University of Munich, Munich, Germany.
| |
Collapse
|
5
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
6
|
Rehman IU, Park JS, Choe K, Park HY, Park TJ, Kim MO. Overview of a novel osmotin abolishes abnormal metabolic-associated adiponectin mechanism in Alzheimer's disease: Peripheral and CNS insights. Ageing Res Rev 2024; 100:102447. [PMID: 39111409 DOI: 10.1016/j.arr.2024.102447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/19/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that affects millions of people worldwide. It is caused by abnormalities in cholinergic neurons, oxidative stress, and inflammatory cascades. The illness is accompanied by personality changes, memory issues, and dementia. Metabolic signaling pathways help with fundamental processes like DNA replication and RNA transcription. Being adaptable is essential for both surviving and treating illness. The body's metabolic signaling depends on adipokines, including adiponectin (APN) and other adipokines secreted by adipose tissues. Energy homeostasis is balanced by adipokines, and nutrients. Overconsumption of nutrients messes with irregular signaling of adipokines, such as APN in both peripheral and brain which leads to neurodegeneration, such as AD. Despite the failure of traditional treatments like memantine and cholinesterase inhibitors, natural plant bioactive substances like Osmotin (OSM) have been given a focus as potential therapeutics due to their antioxidant properties, better blood brain barrier (BBB) permeability, excellent cell viability, and especially nanoparticle approaches. The review highlights the published preclinical literature regarding the role of OSM in AD pathology while there is a need for more research to investigate the hidden therapeutic potential of OSM which may open a new gateway and further strengthen its healing role in the pathogenesis of neurodegeneration, especially AD.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands.
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht 6202 AZ, the Netherlands.
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, United Kingdom.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Alz-Dementia Korea Co., Jinju 52828, Republic of Korea.
| |
Collapse
|
7
|
Zheng J, Zhang W, Xu R, Liu L. The role of adiponectin and its receptor signaling in ocular inflammation-associated diseases. Biochem Biophys Res Commun 2024; 717:150041. [PMID: 38710142 DOI: 10.1016/j.bbrc.2024.150041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Abstract
Ocular inflammation-associated diseases are leading causes of global visual impairment, with limited treatment options. Adiponectin, a hormone primarily secreted by adipose tissue, binds to its receptors, which are widely distributed throughout the body, exerting powerful physiological regulatory effects. The protective role of adiponectin in various inflammatory diseases has gained increasing attention in recent years. Previous studies have confirmed the presence of adiponectin and its receptors in the eyes. Furthermore, adiponectin and its analogs have shown potential as novel drugs for the treatment of inflammatory eye diseases. This article summarizes the evidence for the interplay between adiponectin and inflammatory eye diseases and provides new perspectives on the diagnostic and therapeutic possibilities of adiponectin.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Afzal S, Sattar MA, Albokhadaim I, Attiq A, Kandeel M, Manap ASA, Alhojaily SM. Interaction between Nuclear Receptor and Alpha-Adrenergic Agonist Subtypes in Metabolism and Systemic Hemodynamics of Spontaneously Hypertensive Rats. PPAR Res 2024; 2024:5868010. [PMID: 38899161 PMCID: PMC11186691 DOI: 10.1155/2024/5868010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/22/2023] [Accepted: 05/03/2024] [Indexed: 06/21/2024] Open
Abstract
Partial and full PPAR-γ agonists have shown promising effects and antihypertensive and antidiabetic agents through increased plasma adiponectin concentration. This study is aimed at examining the role of PPAR-γ, alpha-adrenoceptors, and adiponectin receptors in the modulation of vasopressor responses to angiotensin II (Ang II) and adrenergic agonists, after a subset treatment of partial and full PPAR-γ agonists, each individually, and also when coupled with adiponectin in SHRs. The antioxidant potential and metabolic indices for these animals were also determined. Group I (WKY) and group II (SHR) were designated as normotensive control and hypertensive control, respectively. Groups III (SHR) and IV (SHR) received irbesartan (30 mg/kg) and pioglitazone (10 mg/kg) orally for 28 days, and groups V (SHR), VI (SHR), and VII (SHR) were treated with adiponectin (2.5 μg/kg) intraperitoneally alone, in combination with irbesartan, and in combination with pioglitazone, respectively, from days 21 to 28 only. On day 29, sodium pentobarbitone (60 mg/kg) was used to anesthetize all test animals, and systemic hemodynamic and plasma adiponectin concentrations and in vitro and in vivo antioxidant potential were measured. As compared to the WKY control, the SHR control group's noninvasive blood pressure and basal mean arterial pressure were significantly greater, along with increased arterial stiffness, lower plasma nitric oxide, adiponectin concentration, and antioxidant enzyme levels (all P < 0.05). However, they were gradually normalized by single drug treatments in all groups, and to a greater extent in the SHR + Irb + Adp group (P < 0.05). In the acute study, the dose dependant mean arterial pressure responses to intravenously administered adrenergic agonists and angiotensin-II were significantly larger in SHRs as compared to WKY by 20-25%. Adiponectin alone and in combination significantly blunted vasopressor responses to these alpha-adrenergic agonists in the SHR + Pio + Adp group by 63%, whereas attenuated responses to ANG-II administration to 70% in SHR + Irb + Adp. In conclusion, the combined treatment of adiponectin with PPAR-agonists reduced the systemic vascular responses to adrenergic agonists and improved arterial stiffness. This an evidence of the interaction of adiponectin receptors, PPAR-γ, alpha-adrenoceptors, and ANG-II in the systemic vasculature of SHRs. A significant level of synergism has also been proved among full PPAR-γ agonists and adiponectin receptors.
Collapse
Affiliation(s)
- Sheryar Afzal
- Department of Biomedical ScienceCollege of Veterinary MedicineKing Faisal University, Al Hofuf, Saudi Arabia
- Discipline of PharmacologySchool of Pharmaceutical SciencesUniversiti Sains Malaysia, Gelugor 11800, Penang, Malaysia
| | - Munavvar Abdul Sattar
- Discipline of PharmacologySchool of Pharmaceutical SciencesUniversiti Sains Malaysia, Gelugor 11800, Penang, Malaysia
| | - Ibrahim Albokhadaim
- Department of Biomedical ScienceCollege of Veterinary MedicineKing Faisal University, Al Hofuf, Saudi Arabia
| | - Ali Attiq
- Discipline of PharmacologySchool of Pharmaceutical SciencesUniversiti Sains Malaysia, Gelugor 11800, Penang, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical ScienceCollege of Veterinary MedicineKing Faisal University, Al Hofuf, Saudi Arabia
| | - Aimi Syamima Abdul Manap
- Department of Biomedical ScienceCollege of Veterinary MedicineKing Faisal University, Al Hofuf, Saudi Arabia
| | - Sameer M. Alhojaily
- Department of Biomedical ScienceCollege of Veterinary MedicineKing Faisal University, Al Hofuf, Saudi Arabia
| |
Collapse
|
9
|
Merabova N, Ugartemendia L, Edlow AG, Ibarra C, Darbinian N, Tatevosian G, Goetzl L. Maternal obesity: sex-specific in utero changes in fetal brain autophagy and mTOR. Obesity (Silver Spring) 2024; 32:1136-1143. [PMID: 38644654 DOI: 10.1002/oby.24017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 04/23/2024]
Abstract
OBJECTIVE Maternal obesity affects 39.7% of reproductive-age women in the United States. Emerging research has suggested that in utero exposure to maternal obesity is associated with adverse neurodevelopmental outcomes, but knowledge of underlying mechanisms in human samples is lacking. METHODS A matched case-control study was performed in women with singleton fetuses who were undergoing elective pregnancy termination at gestational ages 15 to 21 weeks. Maternal adiponectin levels from plasma were measured using ELISA kits. RNA was extracted from fetal brain tissue using RNeasy Mini Kit (QIAGEN). mRNA expression from ADIPOR1, ADIPOR2, MTOR, ATG5, ATG7, BECN1, and MAP1LC3B was quantified through the ΔΔCt method and using GAPDH as a housekeeping gene. RESULTS We have identified transcription patterns associated with inhibition of autophagy in male fetal brain tissue exposed to maternal obesity (↑MTOR, ↓ATG5, ↓ATG7, and ↓MAP1LC3B), with female fetuses demonstrating either no change in transcription or nonsignificant changes associated with increased autophagy. There was significant downregulation of the autophagy-associated gene BECN1 in both male and female individuals who were exposed to obesity in utero. CONCLUSIONS We present novel evidence suggesting that in utero exposure to maternal obesity in humans may significantly affect neurodevelopment, especially in male fetuses, through alterations in normal autophagy molecular mechanisms and with adiponectin as a potential mediator.
Collapse
Affiliation(s)
- Nana Merabova
- Department of Family Medicine, Medical College of Wisconsin-Prevea, Green Bay, Wisconsin, USA
| | - Lierni Ugartemendia
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, Massachusetts, USA
| | - Claudia Ibarra
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nune Darbinian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Gabriel Tatevosian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Laura Goetzl
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
10
|
Ozcan M, Ayar A. Endocrine Aspects of Pain Pathophysiology: Focus on Adipose Tissue. Neuroendocrinology 2024; 114:894-906. [PMID: 38801814 DOI: 10.1159/000539531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Multiple factors, including neurobiological, hormonal, psychological, and social/cultural norms, influence the manner in which individuals experience pain. Adipose tissue, once considered solely an energy storage site, has been recognized as a significant endocrine organ that produces and releases a range of hormones and cytokines. In recent years, research has highlighted the role of adipose tissue and its endocrine factors in the pathophysiology of pain. SUMMARY This narrative review aimed to provide a comprehensive overview of the current knowledge on the endocrine aspects of pain pathophysiology, with a specific focus on adipose tissue. We examine the role of adipokines released by adipose tissue, such as leptin, adiponectin, resistin, visfatin, asprosin in pain perception and response. We also explore the clinical implications of these findings, including the potential for personalized pain management based on endocrine factors and adipose tissue. KEY MESSAGES Overall, given this background, this review intended to highlight the importance of understanding the endocrine aspects of pain pathophysiology, particularly focusing on the role of adipose tissue, in the development of chronic pain and adipokines. Better understanding the role of adipokines in pain modulation might have therapeutic implications by providing novel targets for addressing underlying mechanism rather than directly focusing on symptoms for chronic pain, particularly in obese individuals.
Collapse
Affiliation(s)
- Mete Ozcan
- Department of Biophysics, Firat University Medical Faculty, Elazig, Turkey
| | - Ahmet Ayar
- Department of Physiology, Karadeniz Technical University Medical Faculty, Trabzon, Turkey
| |
Collapse
|
11
|
Chang HY, Chen SY, Lin JA, Chen YY, Chen YY, Liu YC, Yen GC. Phyllanthus emblica Fruit Improves Obesity by Reducing Appetite and Enhancing Mucosal Homeostasis via the Gut Microbiota-Brain-Liver Axis in HFD-Induced Leptin-Resistant Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10406-10419. [PMID: 38659208 PMCID: PMC11082930 DOI: 10.1021/acs.jafc.4c01226] [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: 02/07/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
The impact of leptin resistance on intestinal mucosal barrier integrity, appetite regulation, and hepatic lipid metabolism through the microbiota-gut-brain-liver axis has yet to be determined. Water extract of Phyllanthus emblica L. fruit (WEPE) and its bioactive compound gallic acid (GA) effectively alleviated methylglyoxal (MG)-triggered leptin resistance in vitro. Therefore, this study investigated how WEPE and GA intervention relieve leptin resistance-associated dysfunction in the intestinal mucosa, appetite, and lipid accumulation through the microbiota-gut-brain-liver axis in high-fat diet (HFD)-fed rats. The results showed that WEPE and GA significantly reduced tissues (jejunum, brain, and liver) MG-evoked leptin resistance, malondialdehyde (MDA), proinflammatory cytokines, SOCS3, orexigenic neuropeptides, and lipid accumulation through increasing leptin receptor, tight junction proteins, antimicrobial peptides, anorexigenic neuropeptides, excretion of fecal triglyceride (TG), and short-chain fatty acids (SCFAs) via a positive correlation with the Allobaculum and Bifidobacterium microbiota. These novel findings suggest that WEPE holds the potential as a functional food ingredient for alleviating obesity and its complications.
Collapse
Affiliation(s)
- Hsin-Yu Chang
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Sheng-Yi Chen
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Jer-An Lin
- Graduate
Institute of Food Safety, National Chung
Hsing University, 145
Xingda Road, Taichung 40227, Taiwan
| | - Ying-Yin Chen
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Ying-Ying Chen
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Yu-Chen Liu
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Gow-Chin Yen
- Department
of Food Science and Biotechnology, National
Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| |
Collapse
|
12
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
13
|
Liu Y, Qian SW, Tang Y, Tang QQ. The secretory function of adipose tissues in metabolic regulation. LIFE METABOLISM 2024; 3:loae003. [PMID: 39872218 PMCID: PMC11748999 DOI: 10.1093/lifemeta/loae003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2025]
Abstract
In addition to their pivotal roles in energy storage and expenditure, adipose tissues play a crucial part in the secretion of bioactive molecules, including peptides, lipids, metabolites, and extracellular vesicles, in response to physiological stimulation and metabolic stress. These secretory factors, through autocrine and paracrine mechanisms, regulate various processes within adipose tissues. These processes include adipogenesis, glucose and lipid metabolism, inflammation, and adaptive thermogenesis, all of which are essential for the maintenance of the balance and functionality of the adipose tissue micro-environment. A subset of these adipose-derived secretory factors can enter the circulation and target the distant tissues to regulate appetite, cognitive function, energy expenditure, insulin secretion and sensitivity, gluconeogenesis, cardiovascular remodeling, and exercise capacity. In this review, we highlight the role of adipose-derived secretory factors and their signaling pathways in modulating metabolic homeostasis. Furthermore, we delve into the alterations in both the content and secretion processes of these factors under various physiological and pathological conditions, shedding light on potential pharmacological treatment strategies for related diseases.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Eng PC, Phylactou M, Qayum A, Woods C, Lee H, Aziz S, Moore B, Miras AD, Comninos AN, Tan T, Franks S, Dhillo WS, Abbara A. Obesity-Related Hypogonadism in Women. Endocr Rev 2024; 45:171-189. [PMID: 37559411 PMCID: PMC10911953 DOI: 10.1210/endrev/bnad027] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/02/2023] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
Obesity-related hypogonadotropic hypogonadism is a well-characterized condition in men (termed male obesity-related secondary hypogonadism; MOSH); however, an equivalent condition has not been as clearly described in women. The prevalence of polycystic ovary syndrome (PCOS) is known to increase with obesity, but PCOS is more typically characterized by increased gonadotropin-releasing hormone (GnRH) (and by proxy luteinizing hormone; LH) pulsatility, rather than by the reduced gonadotropin levels observed in MOSH. Notably, LH levels and LH pulse amplitude are reduced with obesity, both in women with and without PCOS, suggesting that an obesity-related secondary hypogonadism may also exist in women akin to MOSH in men. Herein, we examine the evidence for the existence of a putative non-PCOS "female obesity-related secondary hypogonadism" (FOSH). We précis possible underlying mechanisms for the occurrence of hypogonadism in this context and consider how such mechanisms differ from MOSH in men, and from PCOS in women without obesity. In this review, we consider relevant etiological factors that are altered in obesity and that could impact on GnRH pulsatility to ascertain whether they could contribute to obesity-related secondary hypogonadism including: anti-Müllerian hormone, androgen, insulin, fatty acid, adiponectin, and leptin. More precise phenotyping of hypogonadism in women with obesity could provide further validation for non-PCOS FOSH and preface the ability to define/investigate such a condition.
Collapse
Affiliation(s)
- Pei Chia Eng
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, National University of Singapore, Singapore 117549
| | - Maria Phylactou
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Ambreen Qayum
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Casper Woods
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
| | - Hayoung Lee
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
| | - Sara Aziz
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
| | - Benedict Moore
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
| | - Alexander D Miras
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Tricia Tan
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Steve Franks
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W12 0NN, UK
| |
Collapse
|
15
|
Abbasi K, Zarezadeh R, Valizadeh A, Mehdizadeh A, Hamishehkar H, Nouri M, Darabi M. White-brown adipose tissue interplay in polycystic ovary syndrome: Therapeutic avenues. Biochem Pharmacol 2024; 220:116012. [PMID: 38159686 DOI: 10.1016/j.bcp.2023.116012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
This study highlights the therapeutic potential of activating brown adipose tissue (BAT) for managing polycystic ovary syndrome (PCOS), a prevalent endocrine disorder associated with metabolic and reproductive abnormalities. BAT plays a crucial role in regulating energy expenditure and systemic insulin sensitivity, making it an attractive target for the treatment of obesity and metabolic diseases. Recent research suggests that impaired BAT function and mass may contribute to the link between metabolic disturbances and reproductive issues in PCOS. Additionally, abnormal white adipose tissue (WAT) can exacerbate these conditions by releasing adipokines and nonesterified fatty acids. In this review, we explored the impact of WAT changes on BAT function in PCOS and discussed the potential of BAT activation as a therapeutic strategy to improve PCOS symptoms. We propose that BAT activation holds promise for managing PCOS; however, further research is needed to confirm its efficacy and to develop clinically feasible methods for BAT activation.
Collapse
Affiliation(s)
- Khadijeh Abbasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany.
| |
Collapse
|
16
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
17
|
Samad M, Ek J, Börchers S, Krieger JP, Stener-Victorin E, Skibicka KP, Asterholm IW, Benrick A. Elevated circulating adiponectin levels do not prevent anxiety-like behavior in a PCOS-like mouse model. Sci Rep 2024; 14:563. [PMID: 38177175 PMCID: PMC10766608 DOI: 10.1038/s41598-023-50503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with symptoms of moderate to severe anxiety and depression. Hyperandrogenism is a key feature together with lower levels of the adipocyte hormone adiponectin. Androgen exposure leads to anxiety-like behavior in female offspring while adiponectin is reported to be anxiolytic. Here we test the hypothesis that elevated adiponectin levels protect against the development of androgen-induced anxiety-like behavior. Pregnant mice overexpressing adiponectin (APNtg) and wildtypes were injected with vehicle or dihydrotestosterone to induce prenatal androgenization (PNA) in the offspring. Metabolic profiling and behavioral tests were performed in 4-month-old female offspring. PNA offspring spent more time in the closed arms of the elevated plus maze, indicating anxiety-like behavior. Intriguingly, neither maternal nor offspring adiponectin overexpression prevented an anxiety-like behavior in PNA-exposed offspring. However, adiponectin overexpression in dams had metabolic imprinting effects, shown as lower fat mass and glucose levels in their offspring. While serum adiponectin levels were elevated in APNtg mice, cerebrospinal fluid levels were similar between genotypes. Adiponectin overexpression improved metabolic functions but did not elicit anxiolytic effects in PNA-exposed offspring. These observations might be attributed to increased circulating but unchanged cerebrospinal fluid adiponectin levels in APNtg mice. Thus, increased adiponectin levels in the brain are likely needed to stimulate anxiolytic effects.
Collapse
Affiliation(s)
- Manisha Samad
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
| | - Stina Börchers
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
| | - Jean-Philippe Krieger
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-VetSuisse, 8057, Zurich, Switzerland
| | | | - Karolina P Skibicka
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ingrid Wernstedt Asterholm
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden
| | - Anna Benrick
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 423, 40530, Gothenburg, Sweden.
- School of Health Sciences, University of Skövde, 54128, Skövde, Sweden.
| |
Collapse
|
18
|
Zhao Y, Peng X, Wang Q, Zhang Z, Wang L, Xu Y, Yang H, Bai J, Geng D. Crosstalk Between the Neuroendocrine System and Bone Homeostasis. Endocr Rev 2024; 45:95-124. [PMID: 37459436 DOI: 10.1210/endrev/bnad025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Indexed: 01/05/2024]
Abstract
The homeostasis of bone microenvironment is the foundation of bone health and comprises 2 concerted events: bone formation by osteoblasts and bone resorption by osteoclasts. In the early 21st century, leptin, an adipocytes-derived hormone, was found to affect bone homeostasis through hypothalamic relay and the sympathetic nervous system, involving neurotransmitters like serotonin and norepinephrine. This discovery has provided a new perspective regarding the synergistic effects of endocrine and nervous systems on skeletal homeostasis. Since then, more studies have been conducted, gradually uncovering the complex neuroendocrine regulation underlying bone homeostasis. Intriguingly, bone is also considered as an endocrine organ that can produce regulatory factors that in turn exert effects on neuroendocrine activities. After decades of exploration into bone regulation mechanisms, separate bioactive factors have been extensively investigated, whereas few studies have systematically shown a global view of bone homeostasis regulation. Therefore, we summarized the previously studied regulatory patterns from the nervous system and endocrine system to bone. This review will provide readers with a panoramic view of the intimate relationship between the neuroendocrine system and bone, compensating for the current understanding of the regulation patterns of bone homeostasis, and probably developing new therapeutic strategies for its related disorders.
Collapse
Affiliation(s)
- Yuhu Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaole Peng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhiyu Zhang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Liangliang Wang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230022, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
19
|
Kim JD, Copperi F, Diano S. Microglia in Central Control of Metabolism. Physiology (Bethesda) 2024; 39:0. [PMID: 37962895 PMCID: PMC11283896 DOI: 10.1152/physiol.00021.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/12/2023] [Accepted: 11/12/2023] [Indexed: 11/15/2023] Open
Abstract
Beyond their role as brain immune cells, microglia act as metabolic sensors in response to changes in nutrient availability, thus playing a role in energy homeostasis. This review highlights the evidence and challenges of studying the role of microglia in metabolism regulation.
Collapse
Affiliation(s)
- Jung Dae Kim
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Francesca Copperi
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York, United States
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
20
|
Pathak MP, Patowary P, Chattopadhyay P, Barbhuiyan PA, Islam J, Gogoi J, Wankhar W. Obesity-associated Airway Hyperresponsiveness: Mechanisms Underlying Inflammatory Markers and Possible Pharmacological Interventions. Endocr Metab Immune Disord Drug Targets 2024; 24:1053-1068. [PMID: 37957906 DOI: 10.2174/0118715303256440231028072049] [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: 04/08/2023] [Revised: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 11/15/2023]
Abstract
Obesity is rapidly becoming a global health problem affecting about 13% of the world's population affecting women and children the most. Recent studies have stated that obese asthmatic subjects suffer from an increased risk of asthma, encounter severe symptoms, respond poorly to anti-asthmatic drugs, and ultimately their quality-of-life decreases. Although, the association between airway hyperresponsiveness (AHR) and obesity is a growing concern among the public due to lifestyle and environmental etiologies, however, the precise mechanism underlying this association is yet to establish. Apart from aiming at the conventional antiasthmatic targets, treatment should be directed towards ameliorating obesity pathogenesis too. Understanding the pathogenesis underlying the association between obesity and AHR is limited, however, a plethora of obesity pathologies have been reported viz., increased pro-inflammatory and decreased anti-inflammatory adipokines, depletion of ROS controller Nrf2/HO-1 axis, NLRP3 associated macrophage polarization, hypertrophy of WAT, and down-regulation of UCP1 in BAT following down-regulated AMPKα and melanocortin pathway that may be correlated with AHR. Increased waist circumference (WC) or central obesity was thought to be related to severe AHR, however, some recent reports suggest body mass index (BMI), not WC tends to exaggerate airway closure in AHR due to some unknown mechanisms. This review aims to co-relate the above-mentioned mechanisms that may explain the copious relation underlying obesity and AHR with the help of published reports. A proper understanding of these mechanisms discussed in this review will ensure an appropriate treatment plan for patients through advanced pharmacological interventions.
Collapse
Affiliation(s)
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India
| | | | | | - Johirul Islam
- Department of Pharmaceutical Sciences, School of Health Sciences, Assam Kaziranga University, Jorhat, India
| | - Jyotchna Gogoi
- Department of Biochemistry, Faculty of Science, Assam Down Town University, Guwahati, India
| | - Wankupar Wankhar
- Department of Dialysis, Faculty of Paramedical Science, Assam Down Town University, Guwahati, India
| |
Collapse
|
21
|
Cao M, Zheng S, Zhang W, Hu G. Progress in the study of nutritional status and selenium in dialysis patients. Ann Med 2023; 55:2197296. [PMID: 37038353 PMCID: PMC10101670 DOI: 10.1080/07853890.2023.2197296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/25/2023] [Indexed: 04/12/2023] Open
Abstract
Malnutrition is very common in patients with chronic kidney disease, especially in those on maintenance dialysis. Malnutrition is one of the major factors affecting survival and death of dialysis patients, and reducing their activity tolerance and immunity. There are numerous and interacting risk factors for malnutrition, such as reduced nutritional intake, increased energy expenditure, hormonal disorders, and inflammation. Selenium, in the form of selenoproteins, is involved in many physiological processes in the body and plays an important role in maintaining redox homeostasis. Oxidative stress and infection are very common in dialysis patients, and selenium levels in dialysis patients are significantly lower than those in the healthy population. It has been shown that there is a correlation between selenium levels in hemodialysis patients and their nutrition-related indicators, and that selenium supplementation may improve malnutrition in patients. However, further studies are needed to support this conclusion and there is a lack of basic research to further characterize the potential mechanisms by which selenium may improve malnutrition in dialysis patients. The purpose of this review is to provide a comprehensive overview of factors associated with malnutrition in dialysis patients and to describe the progress of research on nutritional status and selenium levels in dialysis patients.
Collapse
Affiliation(s)
- Meiran Cao
- Department of Nephrology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Shuai Zheng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Wenhua Zhang
- Department of Nephrology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Guicai Hu
- Department of Nephrology, Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
22
|
El-Seidy AMA, Elbaset MA, Ibrahim FAA, Abdelmottaleb Moussa SA, Bashandy SA. Nano cerium oxide and cerium/zinc nanocomposites characterization and therapeutic role in combating obesity via controlling oxidative stress and insulin resistance in rat model. J Trace Elem Med Biol 2023; 80:127312. [PMID: 37804595 DOI: 10.1016/j.jtemb.2023.127312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/18/2023] [Accepted: 09/21/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND CeO2NPs and ZnONPs can curb the increase of cholesterol and triglycerides observed in rats with non-alcoholic fatty liver disease. It was suggested that CeO2 NPs could potentially have an insulin-sensitizing effect, specifically on adipose tissue and skeletal muscle. It was reported that ZnONPs combat the increase of insulin resistance observed in obese rats and could be beneficial value in NAFLD. In our previous work, ZnO-NPs manifested valuable anti-obesity effects via lowering body weight gain, oxidative stress, BMI, lipids, and insulin resistance. METHODS In the present study, cerium oxide nanoparticles (A-1) and cerium/zinc nanocomposites (A-2 and A-3) were synthesized by solgel to investigate their role on oxidative stress, adipocyte hormones, and insulin resistance in an obese rat model. X-ray diffraction, HRTEM, SEM, and XPS were carried out to confirm the crystal structure, the particle size, the morphology of the nanoparticles and the oxidation states. RESULTS The Rietveld refinement has also been executed on A-1 (chi2 = 1.00; average Bragg = 2.92%; R-factor = 2.45%) and on A-2 (Rw = 9.87%, Rex= 9.68%, χ2 = 1.04, GoF = 1.02). The XPS spectra indicated the presence of Ce in + 4 and + 3 oxidation states and Zn as ZnO and ZnO.OH. Cerium oxide and ZnO crystal sizes lie in the range 40.53-45.01 and 40.53-45.01 nm, respectively. The results indicated that treating obese rats with any of the tested nano compounds (5 mg or 10 mg/Kg) lowered plasma cholesterol, triglycerides, LDL, insulin resistance, glucose, and BMI significantly relative to obese group values. On the other hand, HDL increased significantly in obese rats after treatment with either A-2 or A-3 compared to obese rats. The current investigation showed antioxidant activities for A-1, A-2, and A3 as evidenced by the significant increase in GSH level and a significant decrease in MDA. CONCLUSION It was found that A-1, A-2, and A-3 have an efficient therapeutic role in treating of obesity-related hyperlipidemia, oxidative stress and insulin resistance. The results of A-2 and A-3 were more pronounced than those of A-1. The use of Zn/Ce nanocomposite (that have positive characteristics) in combating obesity and its complications could be become a new trend in therapeutic application for a management of obesity.
Collapse
Affiliation(s)
- Ahmed M A El-Seidy
- Inorganic Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt.
| | - Marwan A Elbaset
- Pharmacology Department, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt
| | - Fatma A A Ibrahim
- Biophysics Laboratory, Biochemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt
| | - Sherif A Abdelmottaleb Moussa
- Biophysics Laboratory, Biochemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt
| | - Samir Ae Bashandy
- Pharmacology Department, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt
| |
Collapse
|
23
|
Wu X, Tao Y, Ren Y, Zhang Z, Zhao Y, Tian Y, Li Y, Hou M, Guo Y, Gong Y, Zhang Y, Li D, Li H, Jiang R, Li G, Liu X, Kang X, Tian Y. Adiponectin inhibits GnRH secretion via activating AMPK and PI3K signaling pathways in chicken hypothalamic neuron cells. Poult Sci 2023; 102:103028. [PMID: 37660449 PMCID: PMC10491727 DOI: 10.1016/j.psj.2023.103028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
It has been reported that adiponectin (AdipoQ), an adipokine secreted by white adipose tissue, plays an important role in the control of animal reproduction in addition to its function in energy homeostasis by binding to its receptors AdipoR1/2. However, the molecular mechanisms of AdipoQ in the regulation of animal reproduction remain elusive. In this study, we investigated the effects of AdipoQ on hypothalamic reproductive hormone (GnRH) secretion and reproduction-related receptor gene (estrogen receptor [ER] and progesterone receptor [PR]) expression in hypothalamic neuronal cells (HNCs) of chickens by using real-time fluorescent quantitative PCR (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), Western blot (WB) and cell counting kit-8 (CCK-8) assays and found that overexpression of AdipoQ could increase the expression levels of AdipoR1/2 and reproduction-related receptor genes (P < 0.05) while decreasing the expression level of GnRH. In contrast, interference with AdipoQ mRNA showed the opposite results in HNCs. Furthermore, we demonstrated that AdipoQ exerts its functions through the AMPK and PI3K signaling pathways. Finally, our in vitro experiments found that AdipoRon (a synthetic substitute for AdipoQ) treatment and AdipoR1/2 RNAi interference co-treatment resulted in no effect on GnRH secretion, suggesting that the inhibition of GnRH secretion by AdipoQ is mediated by the AdipoR1/2 signaling axis. In summary, we uncovered, for the first time, the molecular mechanism of AdipoQ in the regulation of reproductive hormone secretion in hypothalamic neurons in chickens.
Collapse
Affiliation(s)
- Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yiqing Tao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yijie Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Meng Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| |
Collapse
|
24
|
Zheng Y, Ye C, He M, Ko WKW, Chan YW, Wong AOL. Goldfish adiponectin: (I) molecular cloning, tissue distribution, recombinant protein expression, and novel function as a satiety factor in fish model. Front Endocrinol (Lausanne) 2023; 14:1283298. [PMID: 38027109 PMCID: PMC10643153 DOI: 10.3389/fendo.2023.1283298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Adiponectin (AdipoQ) is an adipokine involved in glucose homeostasis and lipid metabolism. In mammals, its role in appetite control is highly controversial. To shed light on the comparative aspects of AdipoQ in lower vertebrates, goldfish was used as a model to study feeding regulation by AdipoQ in fish species. As a first step, goldfish AdipoQ was cloned and found to be ubiquitously expressed at the tissue level. Using sequence alignment, protein modeling, phylogenetic analysis and comparative synteny, goldfish AdipoQ was shown to be evolutionarily related to its fish counterparts and structurally comparable with AdipoQ in higher vertebrates. In our study, recombinant goldfish AdipoQ was expressed in E. coli, purified by IMAC, and confirmed to be bioactive via activation of AdipoQ receptors expressed in HepG2 cells. Feeding in goldfish revealed that plasma levels of AdipoQ and its transcript expression in the liver and brain areas involved in appetite control including the telencephalon, optic tectum, and hypothalamus could be elevated by food intake. In parallel studies, IP and ICV injection of recombinant goldfish AdipoQ in goldfish was effective in reducing foraging behaviors and food consumption. Meanwhile, transcript expression of orexigenic factors (NPY, AgRP, orexin, and apelin) was suppressed with parallel rises in anorexigenic factors (POMC, CART, CCK, and MCH) in the telencephalon, optic tectum and/or hypothalamus. In these brain areas, transcript signals for leptin receptor were upregulated with concurrent drops in the NPY receptor and ghrelin receptors. In the experiment with IP injection of AdipoQ, transcript expression of leptin was also elevated with a parallel drop in ghrelin mRNA in the liver. These findings suggest that AdipoQ can act as a novel satiety factor in goldfish. In this case, AdipoQ signals (both central and peripheral) can be induced by feeding and act within the brain to inhibit feeding behaviors and food intake via differential regulation of orexigenic/anorexigenic factors and their receptors. The feeding inhibition observed may also involve the hepatic action of AdipoQ by modulation of feeding regulators expressed in the liver.
Collapse
Affiliation(s)
| | | | | | | | | | - Anderson O. L. Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Karava V, Kondou A, Dotis J, Christoforidis A, Taparkou A, Farmaki E, Kollios K, Liakopoulos V, Printza N. Association Between Adipokine Profile, Systemic Inflammation, Muscle and Protein Energy Wasting in Children With Chronic Kidney Disease. J Ren Nutr 2023; 33:629-638. [PMID: 37178774 DOI: 10.1053/j.jrn.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/07/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
OBJECTIVES This cross-sectional study explores the association of adipokines and interleukin-6 (IL-6) with muscle and protein energy wasting (PEW) in children with chronic kidney disease (CKD). METHODS We measured serum adiponectin, leptin, resistin and IL-6 in 53 patients with CKD stage 3-5. Lean tissue (LTI) and fat tissue index (FTI) were estimated by bioimpedance analysis spectroscopy. PEW was defined as muscle wasting [LTI adjusted to height age (LTI HA) z-score < -1.65 SD) and at least 2 of the following: reduced body mass [body mass index adjusted to height age (BMI HA) z-score < -1.65 SD), poor growth [height z-score < -1.88 SD], questionnaire-based decreased appetite, and serum albumin ≤3.8 g/dL. RESULTS PEW, observed in 8 (15.1%) patients, was more prevalent in CKD stage 5 (P = .010). Among the adipokines, adiponectin, and resistin levels were significantly higher in CKD stage 5 (P < .001, P = .005). Adiponectin was correlated to LTI HA z-score (Rs = -0.417, P = .002), leptin to FTI z-score (Rs = 0.620, P < .001), while no correlation was observed between resistin and body composition parameters. Resistin was the only adipokine correlated to IL-6 (Rs = 0.513, P < .001). After adjustment for CKD stage and patient age, PEW was associated with adiponectin and IL-6 rise by 1 μg/mL and 10 pg/mL respectively (odds ratio (OR) 1.240, 95% confidence interval (CI) 1.040, 1.478 and OR 1.405, 95% CI 1.075-1.836) but not with leptin, while resistin association with PEW lost its significance. CONCLUSIONS In pediatric CKD, adiponectin is associated with muscle wasting, leptin with adiposity and resistin with systemic inflammation. Adiponectin and cytokine IL-6 may serve as PEW biomarkers.
Collapse
Affiliation(s)
- Vasiliki Karava
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Antonia Kondou
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - John Dotis
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Christoforidis
- Pediatric Endocrinology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anna Taparkou
- Pediatric Immunology and Rheumatology Referral Center, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Farmaki
- Pediatric Immunology and Rheumatology Referral Center, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Kollios
- 3rd Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikoleta Printza
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
26
|
Lim JY, Kim E. The Role of Organokines in Obesity and Type 2 Diabetes and Their Functions as Molecular Transducers of Nutrition and Exercise. Metabolites 2023; 13:979. [PMID: 37755259 PMCID: PMC10537761 DOI: 10.3390/metabo13090979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Maintaining systemic homeostasis requires the coordination of different organs and tissues in the body. Our bodies rely on complex inter-organ communications to adapt to perturbations or changes in metabolic homeostasis. Consequently, the liver, muscle, and adipose tissues produce and secrete specific organokines such as hepatokines, myokines, and adipokines in response to nutritional and environmental stimuli. Emerging evidence suggests that dysregulation of the interplay of organokines between organs is associated with the pathophysiology of obesity and type 2 diabetes (T2D). Strategies aimed at remodeling organokines may be effective therapeutic interventions. Diet modification and exercise have been established as the first-line therapeutic intervention to prevent or treat metabolic diseases. This review summarizes the current knowledge on organokines secreted by the liver, muscle, and adipose tissues in obesity and T2D. Additionally, we highlighted the effects of diet/nutrition and exercise on the remodeling of organokines in obesity and T2D. Specifically, we investigated the ameliorative effects of caloric restriction, selective nutrients including ω3 PUFAs, selenium, vitamins, and metabolites of vitamins, and acute/chronic exercise on the dysregulation of organokines in obesity and T2D. Finally, this study dissected the underlying molecular mechanisms by which nutrition and exercise regulate the expression and secretion of organokines in specific tissues.
Collapse
Affiliation(s)
- Ji Ye Lim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| |
Collapse
|
27
|
Kuriyama T, Murata Y, Ohtani R, Yahara R, Nakashima S, Mori M, Ohe K, Mine K, Enjoji M. Modified activity-based anorexia paradigm dampens chronic food restriction-induced hyperadiponectinemia in adolescent female mice. PLoS One 2023; 18:e0289020. [PMID: 37478069 PMCID: PMC10361472 DOI: 10.1371/journal.pone.0289020] [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: 03/28/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023] Open
Abstract
Anorexia nervosa (AN) is a chronic, life-threatening disease with mental and physical components that include excessive weight loss, persistent food restriction, and altered body image. It is sometimes accompanied by hyperactivity, day-night reversal, and amenorrhea. No medications have been approved specific to the treatment of AN, partially due to its unclear etiopathogenesis. Because adiponectin is an appetite-regulating cytokine released by adipose tissue, we hypothesized that it could be useful as a specific biomarker that reflects the disease state of AN, so we developed a modified AN mouse model to test this hypothesis. Twenty-eight 3-week-old female C57BL/6J mice were randomly assigned to the following groups: 1) no intervention; 2) running wheel access; 3) food restriction (FR); and 4) activity-based anorexia (ABA) that included running wheel access plus FR. After a 10-day cage adaptation period, the mice of the FR and ABA groups were given 40% of their baseline food intake until 30% weight reduction (acute FR), then the body weight was maintained for 2.5 weeks (chronic FR). Running wheel activity and the incidence of the estrous cycle were assessed. Spontaneous food restriction and the plasma adiponectin level were evaluated at the end of the acute and chronic FR phases. An increase in running wheel activity was found in the light phase, and amenorrhea was found solely in the ABA group, which indicates that this is a good model of AN. This group showed a slight decrease in spontaneous food intake accompanied with an attenuated level of normally induced plasma adiponectin at the end of the chronic FR phase. These results indicate that the plasma adiponectin level may be a useful candidate biomarker for the status or stage of AN.
Collapse
Affiliation(s)
- Toru Kuriyama
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Yusuke Murata
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Reika Ohtani
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Rei Yahara
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Soichiro Nakashima
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kenji Ohe
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kazunori Mine
- Faculty of Neurology and Psychiatry, BOOCS Clinic Fukuoka, Fukuoka, Japan
| | - Munechika Enjoji
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
28
|
Pei X, Li H, Yu H, Wang W, Mao D. APN Expression in Serum and Corpus Luteum: Regulation of Luteal Steroidogenesis Is Possibly Dependent on the AdipoR2/AMPK Pathway in Goats. Cells 2023; 12:1393. [PMID: 37408227 DOI: 10.3390/cells12101393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/23/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
Adiponectin (APN) is an essential adipokine for a variety of reproductive processes. To investigate the role of APN in goat corpora lutea (CLs), CLs and sera from different luteal phases were collected for analysis. The results showed that the APN structure and content had no significant divergence in different luteal phases both in CLs and sera; however, high molecular weight APN was dominant in serum, while low molecular weight APN was more present in CLs. The luteal expression of both AdipoR1/2 and T-cadherin (T-Ca) increased on D11 and 17. APN and its receptors (AdipoR1/2 and T-Ca) were mainly expressed in goat luteal steroidogenic cells. The steroidogenesis and APN structure in pregnant CLs had a similar model as in the mid-cycle CLs. To further explore the effects and mechanisms of APN in CLs, steroidogenic cells from pregnant CLs were isolated to detect the AMPK-mediated pathway by the activation of APN (AdipoRon) and knockdown of APN receptors. The results revealed that P-AMPK in goat luteal cells increased after incubation with APN (1 μg/mL) or AdipoRon (25 μM) for 1 h, and progesterone (P4) and steroidogenic proteins levels (STAR/CYP11A1/HSD3B) decreased after 24 h. APN did not affect the steroidogenic protein expression when cells were pretreated with Compound C or SiAMPK. APN increased P-AMPK and reduced the CYP11A1 expression and P4 levels when cells were pretreated with SiAdipoR1 or SiT-Ca, while APN failed to affect P-AMPK, the CYP11A1 expression or the P4 levels when pretreated with SiAdipoR2. Therefore, the different structural forms of APN in CLs and sera may possess distinct functions; APN might regulate luteal steroidogenesis through AdipoR2 which is most likely dependent on AMPK.
Collapse
Affiliation(s)
- Xiaomeng Pei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haolin Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dagan Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
29
|
Zhang Z, Guo L, Yang F, Peng S, Wang D, Lai X, Su B, Xie H. Adiponectin Attenuates Splenectomy-Induced Cognitive Deficits by Neuroinflammation and Oxidative Stress via TLR4/MyD88/NF-κb Signaling Pathway in Aged Rats. ACS Chem Neurosci 2023; 14:1799-1809. [PMID: 37141577 DOI: 10.1021/acschemneuro.2c00744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common adverse event after surgical trauma in elderly patients. The pathogenesis of PND is still unclear. Adiponectin (APN) is a plasma protein secreted by adipose tissue. We have reported that a decreased APN expression is associated with PND patients. APN may be a promising therapeutic agent for PND. However, the neuroprotective mechanism of APN in PND is still unclear. In this study, 18 month old male Sprague-Dawley rats were assigned to six groups: the sham, sham + APN (intragastric (i.g.) administration of 10 μg/kg/day for 20 days before splenectomy), PND (splenectomy), PND + APN, PND + TAK-242 (intraperitoneal (i.p.) administration of 3 mg/kg TAK-242), and PND + APN + lipopolysaccharide (LPS) (i.p. administration of 2 mg/kg LPS). We first found that APN gastric infusion significantly improved learning and cognitive function in the Morris water maze (MWM) test after surgical trauma. Further experiments indicated that APN could inhibit the Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor kappa B (NF-κb) p65 pathway to decrease the degree of oxidative damage (malondialdehyde (MDA) and superoxide dismutase (SOD)), microglia-mediated neuroinflammation (ionized calcium binding adapter molecule 1 (IBA1), caspase-1, tumor necrosis factor (TNF)-α, interleukin-1β (IL-1β), and interleukin-6 (IL-6)), and apoptosis (p53, Bcl2, Bax, and caspase 3) in hippocampus. By using LPS-specific agonist and TAK-242-specific inhibitor, the involvement of TLR4 engagement was confirmed. APN intragastric administration exerts a neuroprotective effect against cognitive deficits induced by peripheral trauma, and the possible mechanisms include the inhibition of neuroinflammation, oxidative stress, and apoptosis, mediated by the suppression of the TLR4/MyD88/NF-κb signaling pathway. We propose that oral APN may be a promising candidate for PND treatment.
Collapse
Affiliation(s)
- Zhijing Zhang
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
| | - Lideng Guo
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
- Guangdong Medical University, No. 2 East Wenming Road, Xiashan District, 524000 Zhanjiang, China
| | - Fei Yang
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
- Southern Medical University, No. 1023, South Sha Tai Road, Jingxi Street, Baiyun District, 510000 Guangzhou, China
| | - Shanpan Peng
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
- Guangdong Medical University, No. 2 East Wenming Road, Xiashan District, 524000 Zhanjiang, China
| | - Di Wang
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
- Guangdong Medical University, No. 2 East Wenming Road, Xiashan District, 524000 Zhanjiang, China
| | - Xiawei Lai
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
- Southern Medical University, No. 1023, South Sha Tai Road, Jingxi Street, Baiyun District, 510000 Guangzhou, China
| | - Baiqin Su
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
| | - Haihui Xie
- Department of Anesthesiology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 523000 Dongguan, China
| |
Collapse
|
30
|
Madadi S, Hasasnpour S, Zendehdel M, Vazir B, Jahandideh A. Role of central Adiponectin and its interactions with NPY and GABAergic systems on food intake in neonatal layer chicken. Neurosci Lett 2023; 808:137283. [PMID: 37142113 DOI: 10.1016/j.neulet.2023.137283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 04/29/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND & AIM Adiponectin is a member of the adipokine family and contributes to regulating energy homeostasis, reproduction, and various biological functions, such as insulin receptor signaling pathway sensitivity, mitochondrial biogenesis, oxidative metabolism, neurogenesis, and suppression of inflammation. This study aimed to investigate the effects of intracerebroventricular (ICV) injection of adiponectin and its interaction with the neuropeptide Y (NPY) and GABAergic systems on central appetite regulation in neonatal layer-type chickens. MATERIALS & METHODS In this study, 6 experiments were conducted, each of which included 4 experimental groups. In the first experiment, the chickens were injected with saline and adiponectin (20.73, 41.45, and 62.18 nmol). In the second experiment, saline, adiponectin (62.18 nmol), B5063 (NPY1 receptor antagonist, 2.12 nmol), and simultaneous injections of adiponectin and B5063 were performed. Experiments 3 to 6 were done in the same way to experiment 1, but the chickens were injected with SF22 (NPY2 receptor antagonist, 2.66 nmol), SML0891 (NPY5 receptor antagonist, 2.89 nmol), picrotoxin (GABAA receptor antagonist, 0.89 nmol), CGP54626 (GABAB receptor antagonist, 0.047 nmol) instead of B5063. Feed consumption was measured 120 min after the injection. RESULTS A dose-dependent increase in appetite was observed after the injection of adiponectin (20.73, 41.45, and 62.18 nmol) (P<0.05). The injection of B5063 + adiponectin attenuated the hyperphagic effect of adiponectin (P< 0.05). In addition, co-injection of picrotoxin and adiponectin significantly decreased adiponectin-induced hyperphagia (P<0.05). In addition, adiponectin significantly increased the number of steps, jumps, exploratory food, pecks, and standing time, while decreasing sitting time and rest time (P<0.05). CONCLUSION These results suggest that the hyperphagic effects of adiponectin are probably mediated through NPY1 and GABAA receptors in neonatal layer-type chickens.
Collapse
Affiliation(s)
- Sedigheh Madadi
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shahin Hasasnpour
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Morteza Zendehdel
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453, Tehran, Iran
| | - Bita Vazir
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Jahandideh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
31
|
Munhoz AC, Serna JDC, Vilas-Boas EA, Caldeira da Silva CC, Santos TG, Mosele FC, Felisbino SL, Martins VR, Kowaltowski AJ. Adiponectin reverses β-Cell damage and impaired insulin secretion induced by obesity. Aging Cell 2023:e13827. [PMID: 37060190 DOI: 10.1111/acel.13827] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/16/2023] Open
Abstract
Obesity significantly decreases life expectancy and increases the incidence of age-related dysfunctions, including β-cell dysregulation leading to inadequate insulin secretion. Here, we show that diluted plasma from obese human donors acutely impairs β-cell integrity and insulin secretion relative to plasma from lean subjects. Similar results were observed with diluted sera from obese rats fed ad libitum, when compared to sera from lean, calorically restricted, animals. The damaging effects of obese circulating factors on β-cells occurs in the absence of nutrient overload, and mechanistically involves mitochondrial dysfunction, limiting glucose-supported oxidative phosphorylation and ATP production. We demonstrate that increased levels of adiponectin, as found in lean plasma, are the protective characteristic preserving β-cell function; indeed, sera from adiponectin knockout mice limits β-cell metabolic fluxes relative to controls. Furthermore, oxidative phosphorylation and glucose-sensitive insulin secretion, which are completely abrogated in the absence of this hormone, are restored by the presence of adiponectin alone, surprisingly even in the absence of other serological components, for both the insulin-secreting INS1 cell line and primary islets. The addition of adiponectin to cells treated with plasma from obese donors completely restored β-cell functional integrity, indicating the lack of this hormone was causative of the dysfunction. Overall, our results demonstrate that low circulating adiponectin is a key damaging element for β-cells, and suggest strong therapeutic potential for the modulation of the adiponectin signaling pathway in the prevention of age-related β-cell dysfunction.
Collapse
Affiliation(s)
- Ana Cláudia Munhoz
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Julian D C Serna
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Tiago G Santos
- Centro Internacional de Pesquisa (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Francielle C Mosele
- Instituto de Biociências de Botucatu (IBB), Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Sergio L Felisbino
- Instituto de Biociências de Botucatu (IBB), Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Vilma Regina Martins
- Centro Internacional de Pesquisa (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Qin L, Sitticharoon C, Petyim S, Keadkraichaiwat I, Sririwichitchai R, Maikaew P, Churintaraphan M. A Longitudinal Study of the Relationship of Adiponectin with Reproduction in Infertile Women Undergoing IVF/ICSI Treatment, and an Experimental Study in Human Granulosa Cells. Life (Basel) 2023; 13:life13040994. [PMID: 37109523 PMCID: PMC10141627 DOI: 10.3390/life13040994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/17/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
This study investigated the roles of adiponectin in IVF treatment during Phase I (the basal stage before gonadotropin administration), Phase II (approximately 8 days after gonadotropin administration), and Phase III (on the ovum pick-up day), as well as the effects of adiponectin on CYP19A1 and the FSH receptor (FSHR) mRNA expression in a human granulosa-like tumor cell line (KGN). In human subjects (a longitudinal study, n = 30), blood samples were collected in all phases, while follicular fluid (FF) was only collected in Phase III. The participants were classified into successful and unsuccessful groups based on the determination of fetal heartbeats. KGN cells were treated with adiponectin/FSH/IGF-1 (an experimental study, n = 3). There was no difference in the adiponectin levels between successful and unsuccessful pregnancies in the FF (Phase III) and in serum (all phases), as well as among the three phases in both groups. Serum FSH (Phase I) was positively associated with serum adiponectin in the unsuccessful group, but it had a negative association in the successful group (all phases). Serum adiponectin and serum FSH (Phase I) were positively correlated in the unsuccessful group, whereas they were negatively correlated (all phases) in the successful group. The serum adiponectin levels (Phase III) were significantly higher than in the FF in unsuccessful pregnancies, but there was no difference in successful pregnancies. FF adiponectin concentrations were negatively correlated with serum LH in successful subjects. In KGN cells, adiponectin had no influence on CYP19A1 and FSHR mRNA expression. High adiponectin levels in serum compared to FF (Phase III) in unsuccessful subjects might negatively impact IVF treatment.
Collapse
Affiliation(s)
- Lixian Qin
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Chantacha Sitticharoon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Somsin Petyim
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Issarawan Keadkraichaiwat
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Rungnapa Sririwichitchai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Pailin Maikaew
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Malika Churintaraphan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| |
Collapse
|
33
|
Abstract
The recently uncovered key role of the peripheral and central nervous systems in controlling tumorigenesis and metastasis has opened a new area of research to identify innovative approaches against cancer. Although the 'neural addiction' of cancer is only partially understood, in this Perspective we discuss the current knowledge and perspectives on peripheral and central nerve circuitries and brain areas that can support tumorigenesis and metastasis and the possible reciprocal influence that the brain and peripheral tumours exert on one another. Tumours can build up local autonomic and sensory nerve networks and are able to develop a long-distance relationship with the brain through circulating adipokines, inflammatory cytokines, neurotrophic factors or afferent nerve inputs, to promote cancer initiation, growth and dissemination. In turn, the central nervous system can affect tumour development and metastasis through the activation or dysregulation of specific central neural areas or circuits, as well as neuroendocrine, neuroimmune or neurovascular systems. Studying neural circuitries in the brain and tumours, as well as understanding how the brain communicates with the tumour or how intratumour nerves interplay with the tumour microenvironment, can reveal unrecognized mechanisms that promote cancer development and progression and open up opportunities for the development of novel therapeutic strategies. Targeting the dysregulated peripheral and central nervous systems might represent a novel strategy for next-generation cancer treatment that could, in part, be achieved through the repurposing of neuropsychiatric drugs in oncology.
Collapse
Affiliation(s)
- Claire Magnon
- Laboratory of Cancer and Microenvironment-National Institute of Health and Medical Research (INSERM), Institute of Biology François Jacob-Atomic Energy Commission (CEA), University of Paris Cité, University of Paris-Saclay, Paris, France.
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
34
|
Blokhin ME, Kuranov SO, Khvostov MV, Fomenko VV, Luzina OA, Zhukova NA, Elhajjar C, Tolstikova TG, Salakhutdinov NF. Terpene-Containing Analogues of Glitazars as Potential Therapeutic Agents for Metabolic Syndrome. Curr Issues Mol Biol 2023; 45:2230-2247. [PMID: 36975514 PMCID: PMC10047834 DOI: 10.3390/cimb45030144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic syndrome is a complex of abnormalities involving impaired glucose and lipid metabolism, which needs effective pharmacotherapy. One way to reduce lipid and glucose levels associated with this pathology is the simultaneous activation of nuclear PPAR-alpha and gamma. For this purpose, we synthesized a number of potential agonists based on the pharmacophore fragment of glitazars with the inclusion of mono- or diterpenic moiety in the molecular structure. The study of their pharmacological activity in mice with obesity and type 2 diabetes mellitus (C57Bl/6Ay) revealed one substance that was capable of reducing the triglyceride levels in the liver and adipose tissue of mice by enhancing their catabolism and expressing a hypoglycemic effect connected with the sensitization of mice tissue to insulin. It has also been shown to have no toxic effects on the liver.
Collapse
Affiliation(s)
- Mikhail E Blokhin
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Sergey O Kuranov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Mikhail V Khvostov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Vladislav V Fomenko
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Olga A Luzina
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Natalia A Zhukova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Cham Elhajjar
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | - Tatiana G Tolstikova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090 Novosibirsk, Russia
| | | |
Collapse
|
35
|
Mansour A, Motamed S, Hekmatdoost A, Karimi S, Mohajeri-Tehrani MR, Abdollahi M, Jelodar R, Sajjadi-Jazi SM. Factors related to hypermetabolism in individuals with type 2 diabetes mellitus and non-alcoholic fatty liver disease. Sci Rep 2023; 13:3669. [PMID: 36871124 PMCID: PMC9985614 DOI: 10.1038/s41598-023-30945-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/03/2023] [Indexed: 03/06/2023] Open
Abstract
Considering the progressive prevalence and co-occurrence of type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD), as well as the current evidence suggesting the elevated levels of basal metabolic rate (BMR) among these individuals, the present study aimed to identify factors determining hypermetabolism in such subjects. This cross sectional study was conducted in 30 to 53-year-old individuals with concurrent T2DM and NAFLD (controlled attenuation parameter score ≥ 260 dB/m). Resting energy expenditure (REE) was determined by an indirect calorimetry device. Hypermetabolism was defined as an elevated measured REE > 110% of the predicted REE. The multivariate logistic regression test was used for detecting factors associated with hypermetabolism. Between September, 2017, and March, 2018, a total of 95 eligible participants (64.40% male) with both T2DM and NAFLD were included, while 32.63% of them were classified as hypermetabolic. Overall, the mean recruitment age ± standard deviation and median (interquartile range) body mass index were 44.69 ± 5.47 years and 30.20 (27.80-33.30) kg/m2, respectively. Demographic, anthropometric and biochemical variables did not vary significantly across two groups except for total body water, low-density lipoprotein cholesterol and dipeptidyl peptidase 4 (DPP-4) inhibitors (p < 0.05). According to the results of multivariable logistic regression analyses, hypermetabolism had a positive association with adiponectin (odds ratio [OR] 1.167, 95% confidence interval [CI] 1.015-1.342, p = 0.030), physical activity (OR 1.134, 95% CI 1.002-1.284, p = 0.046), alanine transaminase (OR 1.062, 95% CI 1.006-1.122, p = 0.031) and diastolic blood pressure (OR 1.067, 95% CI 1.010-1.127, p = 0.021). However, fat free mass was inversely related to hypermetabolism (OR 0.935, 95% CI 0.883-0.991, p = 0.023). Adiponectin, alanine transaminase, physical activity, diastolic blood pressure and fat free mass were independently associated with hypermetabolism in subjects with NAFLD and T2DM.
Collapse
Affiliation(s)
- Asieh Mansour
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Azita Hekmatdoost
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Karimi
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Mohajeri-Tehrani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reihane Jelodar
- Department of Internal Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahmoud Sajjadi-Jazi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Xu W, Li J, Ji C, Fang D, Yao L, Xu N, Yi W. Activation of POMC neurons to adiponectin participating in EA-mediated improvement of high-fat diet IR mice. Front Neurosci 2023; 17:1145079. [PMID: 37034166 PMCID: PMC10077892 DOI: 10.3389/fnins.2023.1145079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Background Insulin resistance (IR) is one of the common pathological manifestations of metabolic-related diseases, and the prevalence of relevant diseases is high. Acupuncture is beneficial to IR patients, but the central mechanism underlying this treatment remains unclear. This study provides mechanistic insights into how electroacupuncture (EA) improves IR through the response of Pro-opiomelanocortin (POMC) neurons to adiponectin (Adipo). Methods Glucose tolerance tests (GTT), Insulin tolerance tests (ITT) and fasting blood glucose (FBG) were detected by glucometer. Serum insulin, Adipo and skeletal muscle adiponectin receptor 1 (AdipoR1) protein levels were examined by ELISA. Homeostasis model assessment estimated insulin resistance (HOMA-IR) was calculated using the following formula: HOMA-IR = fasting insulin (FINS) (mU/L) × FBG (mmol/L)/22.5. The expression levels of AdipoR1 and Adipo mRNA in skeletal muscle were detected by real-time PCR quantification. The co-marking of c-Fos/AdipoR1 and POMC neurons were investigated using immunofluorescence. Spontaneous excitatory postsynaptic currents (sEPSCs) of POMC neurons and the response of POMC neurons to Adipo were detected via electrophysiology. Results EA significantly ameliorated HFD-induced impairment of GTT, ITT, FBG, and HOMA-IR which was correlated with recovery of the expression level of AdipoR1 and Adipo in skeletal muscle. The improved response of POMC neurons to Adipo in the hypothalamus may be a key factor in correcting abnormal glucose tolerance and improving IR. Conclusion This study demonstrates that EA can ameliorate HFD-induced impaired glucose tolerance through improved response of POMC neurons to Adipo in the hypothalamus, providing insight into the central mechanism of improving IR through EA.
Collapse
Affiliation(s)
- Wanling Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junfeng Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang Ji
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Danwei Fang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lulu Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Yi
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Wei Yi,
| |
Collapse
|
37
|
9-N-n-alkyl Berberine Derivatives: Hypoglycemic Activity Evaluation. Pharmaceutics 2022; 15:pharmaceutics15010044. [PMID: 36678673 PMCID: PMC9865096 DOI: 10.3390/pharmaceutics15010044] [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] [Received: 12/05/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Several novel 9-N-n-alkyl derivatives of berberine (C5, C7, C10, C12) were synthesized. They were analyzed in vitro and in vivo for their hypoglycemic activity. In vitro studies showed that the derivatives with shorter alkyl substitutes at concentrations ranging from 2.5 to 10 μM were able to stimulate glucose consumption by HepG2 cells more prominently than the derivatives with longer substitutes (C10 and C12). All compounds demonstrated a better effect compared to berberine. Their impact on cells' viability also depended on the alkyl substitutes length, but in this case, C10 and C12 derivatives demonstrated the best results. A similar correlation was also found in the OGTT, where the C5 derivative demonstrated a pronounced hypoglycemic effect at a dose of 15 mg/kg and C12 was less effective. This compound was further investigated in C57BL/6Ay mice for four weeks and was administered at a dose of 15 mg/kg. Pronounced effect of C12 on carbohydrate metabolism in mice was discovered: there was a decrease in fasting glucose levels and an increase in glucose tolerance in OGTT on the 14th and 28th days of the experiment. However, at the end of the experiment, signs of hepatosis exacerbation and an increase in the content of hepatic aminotransferases in blood were found.
Collapse
|
38
|
Study of Hypoglycemic Activity of Novel 9-N-alkyltetrahydroberberine Derivatives. Int J Mol Sci 2022; 23:ijms232214186. [PMID: 36430664 PMCID: PMC9698964 DOI: 10.3390/ijms232214186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Novel 9-N-alkyltetrahydroberberine derivatives were synthesized, among which, based on the results of OGTT, one compound containing the longest aliphatic substituent was selected for study in mice C57BL/6Ay, which demonstrate obesity, impaired glucose tolerance, and concomitant liver non-alcoholic fatty disease. Administration of this substance at a dose of 15 mg/kg for four weeks improved the insulin sensitivity of mice, which resulted in a decrease in fasting glucose levels and improved the tolerance of mice to OGTT glucose loading. A decrease in the level of lactate in the blood and a decrease in the amount of glucokinase in the liver were also found. The introduction of compound 3c did not have a toxic effect on animals based on biochemical data, histological analysis, and measurements of general parameters such as body weight and feed intake. Thus, the 9-N-heptyltetrahydroberberine derivative showed prominent hypoglycemic effects, which makes it promising to obtain and study other derivatives with longer substituents.
Collapse
|
39
|
Normand E, Franco A, Alos N, Parent S, Moreau A, Marcil V. Circulatory Adipokines and Incretins in Adolescent Idiopathic Scoliosis: A Pilot Study. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1619. [PMID: 36360347 PMCID: PMC9688531 DOI: 10.3390/children9111619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 08/26/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a three-dimensional malformation of the spine of unknown cause that develops between 10 and 18 years old and affects 2-3% of adolescents, mostly girls. It has been reported that girls with AIS have a taller stature, lower body mass index (BMI), and bone mineral density (BMD) than their peers, but the causes remain unexplained. Energy metabolism discrepancies, including alterations in adipokine and incretin circulatory levels, could influence these parameters and contribute to disease pathophysiology. This pilot study aims to compare the anthropometry, BMD, and metabolic profile of 19 AIS girls to 19 age-matched healthy controls. Collected data include participants' fasting metabolic profile, anthropometry (measurements and DXA scan), nutritional intake, and physical activity level. AIS girls (14.8 ± 1.7 years, Cobb angle 27 ± 10°), compared to controls (14.8 ± 2.1 years), were leaner (BMI-for-age z-score ± SD: -0.59 ± 0.81 vs. 0.09 ± 1.11, p = 0.016; fat percentage: 24.4 ± 5.9 vs. 29.2 ± 7.2%, p = 0.036), had lower BMD (total body without head z-score ± SD: -0.6 ± 0.83 vs. 0.23 ± 0.98, p = 0.038; femoral neck z-score: -0.54 ± 1.20 vs. 0.59 ± 1.59, p = 0.043), but their height was similar. AIS girls had higher adiponectin levels [56 (9-287) vs. 32 (7-74) μg/mL, p = 0.005] and lower leptin/adiponectin ratio [0.042 (0.005-0.320) vs. 0.258 (0.024-1.053), p = 0.005]. AIS participants with a Cobb angle superior to 25° had higher resistin levels compared to controls [98.2 (12.8-287.2) vs. 32.1 (6.6-73.8), p = 0.0013]. This pilot study suggests that adipokines are implicated in AIS development and/or progression, but more work is needed to confirm their role in the disease.
Collapse
Affiliation(s)
- Emilie Normand
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Anita Franco
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Viscogliosi Laboratory in Molecular Genetics and Musculoskeletal Diseases, Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
| | - Nathalie Alos
- Endocrine Service, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Stefan Parent
- Department of Surgery, CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
| | - Alain Moreau
- Viscogliosi Laboratory in Molecular Genetics and Musculoskeletal Diseases, Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montreal, QC H3A 1J4, Canada
| | - Valérie Marcil
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
40
|
Pfabigan DM, Vezzani C, Thorsby PM, Sailer U. Sex difference in human olfactory sensitivity is associated with plasma adiponectin. Horm Behav 2022; 145:105235. [PMID: 35868172 DOI: 10.1016/j.yhbeh.2022.105235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022]
Abstract
Energy deprivation as well as hormones that regulate appetite and eating can influence olfactory function. This study investigated olfactory sensitivity for a food-related and a non-food odour prior to and after a meal, and its relationship to the energy-regulating hormones ghrelin and adiponectin. The olfactory sensitivity for orange and rose (PEA) odour in healthy, normal-weight volunteers (19 women, 45 men, 1 undisclosed individual) was not affected by the consumption of a meal. Olfactory sensitivity was not associated with concentrations of circulating ghrelin. However, olfactory sensitivity was higher for women than for men, indicating better olfactory performance. This difference between women and men was related to concentrations of plasma adiponectin, an adipose-specific hormone. Adiponectin may thus explain why sex differences in olfactory sensitivity emerge, and may also account for some of the inconsistencies in previous findings on sex differences. Our findings add to the limited literature on the impact of stomach and adipose tissue-derived hormones on olfactory sensitivity. Further studies are needed to establish a causal link between circulating adiponectin and a sex difference in olfactory sensitivity.
Collapse
Affiliation(s)
- Daniela M Pfabigan
- Dept. of Behavioural Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Morbid Obesity Centre, Department of Medicine, Vestfold Hospital Trust, Tønsberg, Norway
| | - Cecilia Vezzani
- Dept. of Behavioural Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Per Medbøe Thorsby
- Hormone Laboratory, Dep of Medical Biochemistry and Biochemical Endocrinology and Metabolism Research Group, Oslo University Hospital, Aker, Oslo, Norway
| | - Uta Sailer
- Dept. of Behavioural Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
41
|
Frohlich J, Kovacovicova K, Raffaele M, Virglova T, Cizkova E, Kucera J, Bienertova-Vasku J, Wabitsch M, Peyrou M, Bonomini F, Rezzani R, Chaldakov GN, Tonchev AB, Di Rosa M, Blavet N, Hejret V, Vinciguerra M. GDF11 inhibits adipogenesis and improves mature adipocytes metabolic function via WNT/β-catenin and ALK5/SMAD2/3 pathways. Cell Prolif 2022; 55:e13310. [PMID: 35920128 PMCID: PMC9528760 DOI: 10.1111/cpr.13310] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/11/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022] Open
Abstract
Objective GDF11 is a member of the TGF‐β superfamily that was recently implicated as potential “rejuvenating” factor, which can ameliorate metabolic disorders. The main objective of the presented study was to closely characterize the role of GDF11 signaling in the glucose homeostasis and in the differentiation of white adipose tissue. Methods We performed microscopy imaging, biochemical and transcriptomic analyses of adipose tissues of 9 weeks old ob/ob mice and murine and human pre‐adipocyte cell lines. Results Our in vivo experiments employing GDF11 treatment in ob/ob mice showed improved glucose/insulin homeostasis, decreased weight gain and white adipocyte size. Furthermore, GDF11 treatment inhibited adipogenesis in pre‐adipocytes by ALK5‐SMAD2/3 activation in cooperation with the WNT/β‐catenin pathway, whose inhibition resulted in adipogenic differentiation. Lastly, we observed significantly elevated levels of the adipokine hormone adiponectin and increased glucose uptake by mature adipocytes upon GDF11 exposure. Conclusion We show evidence that link GDF11 to adipogenic differentiation, glucose, and insulin homeostasis, which are pointing towards potential beneficial effects of GDF11‐based “anti‐obesity” therapy.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Kristina Kovacovicova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Psychogenics Inc, Tarrytown, New York, USA
| | - Marco Raffaele
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Tereza Virglova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Eliska Cizkova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jan Kucera
- Research Center for Toxic Compounds in the Environment (RECETOX), Masaryk University, Brno, Czech Republic
| | - Julie Bienertova-Vasku
- Research Center for Toxic Compounds in the Environment (RECETOX), Masaryk University, Brno, Czech Republic.,Faculty of Medicine, Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Marion Peyrou
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red "Fisiopatología de la Obesidad y Nutrición", Madrid, Spain.,Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Francesca Bonomini
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, Brescia, Italy
| | - George N Chaldakov
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.,Department of Anatomy and Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
| | - Anton B Tonchev
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.,Department of Anatomy and Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Nicolas Blavet
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Vaclav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,National Center for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
| |
Collapse
|
42
|
Luo L, Liu M. Adiponectin: friend or foe in obesity and inflammation. MEDICAL REVIEW (2021) 2022; 2:349-362. [PMID: 37724325 PMCID: PMC10388816 DOI: 10.1515/mr-2022-0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 09/20/2023]
Abstract
Adiponectin is an adipokine predominantly produced by fat cells, circulates and exerts insulin-sensitizing, cardioprotective and anti-inflammatory effects. Dysregulation of adiponectin and/or adiponectin signaling is implicated in a number of metabolic diseases such as obesity, insulin resistance, diabetes, and cardiovascular diseases. However, while the insulin-sensitizing and cardioprotective effects of adiponectin have been widely appreciated in the field, the obesogenic and anti-inflammatory effects of adiponectin are still of much debate. Understanding the physiological function of adiponectin is critical for adiponectin-based therapeutics for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Liping Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
43
|
Ramírez-Plascencia OD, Saderi N, Cárdenas-Romero S, García-García F, Peña-Escudero C, Flores-Sandoval O, Azuara-Álvarez L, Báez-Ruiz A, Salgado-Delgado R. Leptin and adiponectin regulate the activity of nuclei involved in sleep-wake cycle in male rats. Front Neurosci 2022; 16:907508. [PMID: 35937866 PMCID: PMC9355486 DOI: 10.3389/fnins.2022.907508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiological and experimental evidence recognize a relationship between sleep-wake cycles and adiposity levels, but the mechanisms that link both are not entirely understood. Adipose tissue secretes adiponectin and leptin hormones, mainly involved as indicators of adiposity levels and recently associated to sleep. To understand how two of the main adipose tissue hormones could influence sleep-wake regulation, we evaluated in male rats, the effect of direct administration of adiponectin or leptin in the ventrolateral preoptic nuclei (VLPO), a major area for sleep promotion. The presence of adiponectin (AdipoR1 and AdipoR2) and leptin receptors in VLPO were confirmed by immunohistochemistry. Adiponectin administration increased wakefulness during the rest phase, reduced delta power, and activated wake-promoting neurons, such as the locus coeruleus (LC), tuberomammillary nucleus (TMN) and hypocretin/orexin neurons (OX) within the lateral hypothalamus (LH) and perifornical area (PeF). Conversely, leptin promoted REM and NREM sleep, including increase of delta power during NREM sleep, and induced c-Fos expression in VLPO and melanin concentrating hormone expressing neurons (MCH). In addition, a reduction in wake-promoting neurons activity was found in the TMN, lateral hypothalamus (LH) and perifornical area (PeF), including in the OX neurons. Moreover, leptin administration reduced tyrosine hydroxylase (TH) immunoreactivity in the LC. Our data suggest that adiponectin and leptin act as hormonal mediators between the status of body energy and the regulation of the sleep-wake cycle.
Collapse
Affiliation(s)
- Oscar Daniel Ramírez-Plascencia
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Nadia Saderi
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Skarleth Cárdenas-Romero
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Fabio García-García
- Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| | - Carolina Peña-Escudero
- Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| | - Omar Flores-Sandoval
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Lucia Azuara-Álvarez
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Adrián Báez-Ruiz
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Roberto Salgado-Delgado
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- *Correspondence: Roberto Salgado-Delgado,
| |
Collapse
|
44
|
Vermeiren E, Van Eyck A, Van De Maele K, Ysebaert M, Makhout S, De Guchtenaere A, Van Helvoirt M, Tanghe A, Naets T, Vervoort L, Braet C, Bruyndonckx L, De Winter B, Verhulst S, Van Hoorenbeeck K. The Predictive Value of Adipokines and Metabolic Risk Factors for Dropouts and Treatment Outcomes in Children With Obesity Treated in a Pediatric Rehabilitation Center. Front Endocrinol (Lausanne) 2022; 13:822962. [PMID: 35769076 PMCID: PMC9234213 DOI: 10.3389/fendo.2022.822962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Background Inpatient pediatric obesity treatments are highly effective, although dropouts and weight regain threaten long-term results. Preliminary data indicate that leptin, adiponectin, and cardiometabolic comorbidities might predict treatment outcomes. Previous studies have mainly focused on the individual role of adipokines and comorbidities, which is counterintuitive, as these risk factors tend to cluster. This study aimed to predict the dropouts and treatment outcomes by pre-treatment patient characteristics extended with cardiometabolic comorbidities (individually and in total), leptin, and adiponectin. Methods Children aged 8-18 years were assessed before, immediately after and 6 months after a 12-month inpatient obesity treatment. Anthropometric data were collected at each visit. Pre-treatment lipid profiles; glucose, insulin, leptin, and adiponectin levels; and blood pressure were measured. The treatment outcome was evaluated by the change in body mass index (BMI) standard deviation score (SDS) corrected for age and sex. Results We recruited 144 children with a mean age of 14.3 ± 2.2 years and a mean BMI of 36.7 ± 6.2 kg/m2 corresponding to 2.7 ± 0.4 BMI SDS. The 57 patients who dropped out during treatment and the 44 patients who dropped out during aftercare had a higher pre-treatment BMI compared to the patients who completed the treatment (mean BMI, 38.3 ± 6.8 kg/m2 vs 35.7 ± 5.5 kg/m2) and those who completed aftercare (mean BMI, 34.6 ± 5.3 kg/m2 vs 37.7 ± 6.3 kg/m2) (all p<0.05). Additionally, aftercare attenders were younger than non-attenders (mean age, 13.4 ± 2.3 years vs 14.9 ± 2.0, p<0.05).Patients lost on average 1.0 ± 0.4 SDS during treatment and regained 0.4 ± 0.3 SDS post-treatment corresponding to regain of 43 ± 27% (calculated as the increase in BMI SDS post-treatment over the BMI SDS lost during treatment). A higher BMI and more comorbidities inversely predicted BMI SDS reduction in linear regression (all p<0.05).The absolute BMI SDS increase after returning home was predicted by pre-treatment leptin and systolic blood pressure, whereas the post-treatment BMI SDS regain was predicted by pre-treatment age, leptin, and adiponectin levels (all p<0.05) in multivariate linear regressions. Conclusion Patients who need treatment the most are at increased risk for dropouts and weight regain, emphasizing the urgent need for interventions to reduce dropout and support inpatients after discharge. Furthermore, this study is the first to report that pre-treatment leptin and adiponectin levels predict post-treatment BMI SDS regain, requiring further research.
Collapse
Affiliation(s)
- Eline Vermeiren
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Annelies Van Eyck
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| | | | - Marijke Ysebaert
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| | - Sanae Makhout
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | - Tiffany Naets
- Department of Developmental, Personality and Social Psychology, Ghent University, Ghent, Belgium
| | - Leentje Vervoort
- Department of Developmental, Personality and Social Psychology, Ghent University, Ghent, Belgium
- Department of Developmental Psychology, Radboud University, Nijmegen, Netherlands
| | - Caroline Braet
- Department of Developmental, Personality and Social Psychology, Ghent University, Ghent, Belgium
| | - Luc Bruyndonckx
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
- Department of Pediatric Cardiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Benedicte De Winter
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Stijn Verhulst
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| | - Kim Van Hoorenbeeck
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
45
|
Dai W, Liu J, Qiu Y, Teng Z, Li S, Huang J, Xiang H, Tang H, Wang B, Chen J, Wu H. Shared postulations between bipolar disorder and polycystic ovary syndrome pathologies. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110498. [PMID: 34929323 DOI: 10.1016/j.pnpbp.2021.110498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 12/06/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Women with bipolar disorder (BD) present a high prevalence of polycystic ovary syndrome (PCOS) and other reproductive disorders even before diagnosis or treatment of the disease. Postulations on the potential molecular mechanisms of comorbid PCOS in women with BD remain limited to influence of medications and need further extension. OBJECTIVES This review focuses on evidence suggesting that common metabolic and immune disorders may play an important role in the development of BD and PCOS. RESULTS The literature covered in this review suggests that metabolic and immune disorders, including the dysfunction of the hypothalamic-pituitary-adrenal axis, chronic inflammatory state, gut microbial alterations, adipokine alterations and circadian rhythm disturbance, are observed in patients with BD and PCOS. Such disorders may be responsible for the increased prevalence of PCOS in the BD population and indicate a susceptibility gene overlap between the two diseases. Current evidence supports postulations of common metabolic and immune disorders as endophenotype in BD as well as in PCOS. CONCLUSIONS Metabolic and immune disorders may be responsible for the comorbid PCOS in the BD population. The identification of hallmark metabolic and immune features common to these two diseases will contribute to the clarification of the effect of BD on the reproductive endocrine function and development of symptomatic treatments targeting the biomarkers of the two diseases.
Collapse
Affiliation(s)
- Wenyu Dai
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jieyu Liu
- Department of Ultrasound Diagnostic, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yan Qiu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Ziwei Teng
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Sujuan Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jing Huang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hui Xiang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hui Tang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Bolun Wang
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jindong Chen
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| | - Haishan Wu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, China National Technology Institute on Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
46
|
Araújo MC, Soczek SHS, Pontes JP, Marques LAC, Santos GS, Simão G, Bueno LR, Maria-Ferreira D, Muscará MN, Fernandes ES. An Overview of the TRP-Oxidative Stress Axis in Metabolic Syndrome: Insights for Novel Therapeutic Approaches. Cells 2022; 11:cells11081292. [PMID: 35455971 PMCID: PMC9030853 DOI: 10.3390/cells11081292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is a complex pathology characterized by visceral adiposity, insulin resistance, arterial hypertension, and dyslipidaemia. It has become a global epidemic associated with increased consumption of high-calorie, low-fibre food and sedentary habits. Some of its underlying mechanisms have been identified, with hypoadiponectinemia, inflammation and oxidative stress as important factors for MS establishment and progression. Alterations in adipokine levels may favour glucotoxicity and lipotoxicity which, in turn, contribute to inflammation and cellular stress responses within the adipose, pancreatic and liver tissues, in addition to hepatic steatosis. The multiple mechanisms of MS make its clinical management difficult, involving both non-pharmacological and pharmacological interventions. Transient receptor potential (TRP) channels are non-selective calcium channels involved in a plethora of physiological events, including energy balance, inflammation and oxidative stress. Evidence from animal models of disease has contributed to identify their specific contributions to MS and may help to tailor clinical trials for the disease. In this context, the oxidative stress sensors TRPV1, TRPA1 and TRPC5, play major roles in regulating inflammatory responses, thermogenesis and energy expenditure. Here, the interplay between these TRP channels and oxidative stress in MS is discussed in the light of novel therapies to treat this syndrome.
Collapse
Affiliation(s)
- Mizael C. Araújo
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Suzany H. S. Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Jaqueline P. Pontes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 565085-080, MA, Brazil;
| | - Leonardo A. C. Marques
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Gabriela S. Santos
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Laryssa R. Bueno
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Elizabeth S. Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
- Correspondence:
| |
Collapse
|
47
|
Santos RPM, Ribeiro R, Ferreira-Vieira TH, Aires RD, de Souza JM, Oliveira BS, Lima ALD, de Oliveira ACP, Reis HJ, de Miranda AS, Vieira EML, Ribeiro FM, Vieira LB. Metabotropic glutamate receptor 5 knockout rescues obesity phenotype in a mouse model of Huntington's disease. Sci Rep 2022; 12:5621. [PMID: 35379852 PMCID: PMC8980063 DOI: 10.1038/s41598-022-08924-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity represents a global health problem and is characterized by metabolic dysfunctions and a low-grade chronic inflammatory state, which can increase the risk of comorbidities, such as atherosclerosis, diabetes and insulin resistance. Here we tested the hypothesis that the genetic deletion of metabotropic glutamate receptor 5 (mGluR5) may rescue metabolic and inflammatory features present in BACHD mice, a mouse model of Huntington's disease (HD) with an obese phenotype. For that, we crossed BACHD and mGluR5 knockout mice (mGluR5-/-) in order to obtain the following groups: Wild type (WT), mGluR5-/-, BACHD and BACHD/mGluR5-/- (double mutant mice). Our results showed that the double mutant mice present decreased body weight as compared to BACHD mice in all tested ages and reduced visceral adiposity as compared to BACHD at 6 months of age. Additionally, 12-month-old double mutant mice present increased adipose tissue levels of adiponectin, decreased leptin levels, and increased IL-10/TNF ratio as compared to BACHD mice. Taken together, our preliminary data propose that the absence of mGluR5 reduce weight gain and visceral adiposity in BACHD mice, along with a decrease in the inflammatory state in the visceral adipose tissue (VAT), which may indicate that mGluR5 may play a role in adiposity modulation.
Collapse
Affiliation(s)
- Rebeca P M Santos
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Roberta Ribeiro
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Talita H Ferreira-Vieira
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
- Faculdade Sete Lagoas, Sete Lagoas, Brazil
| | - Rosaria D Aires
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
- Faculdade Sete Lagoas, Sete Lagoas, Brazil
| | - Jessica M de Souza
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Bruna S Oliveira
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anna Luiza D Lima
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Antônio Carlos P de Oliveira
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Helton J Reis
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Aline S de Miranda
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erica M L Vieira
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - Fabiola M Ribeiro
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil.
| | - Luciene B Vieira
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Ave. Antonio Carlos 6627, Belo Horizonte, MG, CEP 31270-901, Brazil.
| |
Collapse
|
48
|
Green tea extract increases adiponectin and PPARα levels to improve hepatic steatosis. J Nutr Biochem 2022; 103:108957. [DOI: 10.1016/j.jnutbio.2022.108957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 11/18/2021] [Accepted: 01/05/2022] [Indexed: 01/08/2023]
|
49
|
Park JS, Saeed K, Jo MH, Kim MW, Lee HJ, Park CB, Lee G, Kim MO. LDHB Deficiency Promotes Mitochondrial Dysfunction Mediated Oxidative Stress and Neurodegeneration in Adult Mouse Brain. Antioxidants (Basel) 2022; 11:antiox11020261. [PMID: 35204143 PMCID: PMC8868245 DOI: 10.3390/antiox11020261] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Age-related decline in mitochondrial function and oxidative stress plays a critical role in neurodegeneration. Lactate dehydrogenase-B (LDHB) is a glycolytic enzyme that catalyzes the conversion of lactate, an important brain energy substrate, into pyruvate. It has been reported that the LDHB pattern changes in the brain during ageing. Yet very little is known about the effect of LDHB deficiency on brain pathology. Here, we have used Ldhb knockout (Ldhb−/−) mice to test the hypothesis that LDHB deficiency plays an important role in oxidative stress-mediated neuroinflammation and neurodegeneration. LDHB knockout (Ldhb−/−) mice were generated by the ablation of the Ldhb gene using the Cre/loxP-recombination system in the C57BL/6 genetic background. The Ldhb−/− mice were treated with either osmotin (15 μg/g of the body; intraperitoneally) or vehicle twice a week for 5-weeks. After behavior assessments, the mice were sacrificed, and the cortical and hippocampal brain regions were analyzed through biochemical and morphological analysis. Ldhb−/− mice displayed enhanced reactive oxygen species (ROS) and lipid peroxidation (LPO) production, and they revealed depleted stores of cellular ATP, GSH:GSSG enzyme ratio, and downregulated expression of Nrf2 and HO-1 proteins, when compared to WT littermates. Importantly, the Ldhb−/− mice showed upregulated expression of apoptosis mediators (Bax, Cytochrome C, and caspase-3), and revealed impaired p-AMPK/SIRT1/PGC-1alpha signaling. Moreover, LDHB deficiency-induced gliosis increased the production of inflammatory mediators (TNF-α, Nf-ĸB, and NOS2), and revealed cognitive deficits. Treatment with osmotin, an adipoR1 natural agonist, significantly increased cellular ATP production by increasing mitochondrial function and attenuated oxidative stress, neuroinflammation, and neuronal apoptosis, probably, by upregulating p-AMPK/SIRT1/PGC-1alpha signaling in Ldhb−/− mice. In brief, LDHB deficiency may lead to brain oxidative stress-mediated progression of neurodegeneration via regulating p-AMPK/SIRT1/PGC-1alpha signaling, while osmotin could improve mitochondrial functions, abrogate oxidative stress and alleviate neuroinflammation and neurodegeneration in adult Ldhb−/− mice.
Collapse
Affiliation(s)
- Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
| | - Kamran Saeed
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
| | - Myeung Hoon Jo
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
| | - Min Woo Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
| | - Hyeon Jin Lee
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
| | - Chan-Bae Park
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea; or
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.S.P.); (K.S.); (M.H.J.); (M.W.K.); (H.J.L.)
- Alz-Dementia Korea Co., Jinju 52828, Korea
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
50
|
APOE Polymorphism and Endocrine Functions in Subjects with Morbid Obesity Undergoing Bariatric Surgery. Genes (Basel) 2022; 13:genes13020222. [PMID: 35205269 PMCID: PMC8871864 DOI: 10.3390/genes13020222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/18/2022] [Accepted: 01/23/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Obesity is an interplay between genes and the environment, including lifestyle. The genetics of obesity is insufficiently understood. Apolipoprotein E (APOE) genetic polymorphism has been associated with a wide range of disorders. Knowing that some APOE alleles are associated with obesity and endocrine disorders that are common in obesity, the present study aimed at exploring associations between APOE polymorphisms and endocrine functions in subjects with obesity undergoing bariatric surgery. METHODS Analyses of hormones in blood collected before and one year after bariatric surgery were examined. The APOE alleles were grouped as follows: E2 = ε2ε2 + ε2ε3; E3 = ε3ε3 + ε2ε4; E4 = ε3ε4 + ε4ε4. The APOE groups were analysed as nominal and ordered groups (E2-E3-E4) with a linear mixed model to predict the hormonal effects of the groups. RESULTS Forty-nine women (79%) and thirteen (21%) men with a mean age of 47.7 (SD 8.5) years were included in the study. The adiponectin level was significantly lower (p < 0.05) in the E2 group compared with the E4 group. Adiponectin and cortisol were positively and negatively associated, respectively, with the ordered APOE groups. CONCLUSIONS The ordered APOE groups E2-E3-E4 were significantly associated with high and low levels of adiponectin and cortisol, respectively. The findings indicate APOE-mediated effects on body weight and metabolic functions in subjects with morbid obesity.
Collapse
|