1
|
Guo J, Xue H, Chen Y, Li X, Chen Y, Zhang X, An Y, Zhang H, Yang Y, Cai L, Zhang W, Xiao Y. Infrared thermography-based radiomics for early detection of metabolic syndrome. Sci Rep 2025; 15:13984. [PMID: 40263480 PMCID: PMC12015423 DOI: 10.1038/s41598-025-98831-1] [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: 10/22/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Radiomics is increasingly utilized in medical image analysis. This study evaluates the use of infrared thermography, a technique well-suited for radiomic analysis, in diagnosing metabolic syndrome (MS). Facial and palmar thermographs from 200 males (100 healthy controls and 100 MS patients) were analyzed. The dataset was split into a training cohort (n = 140) and a validation cohort (n = 60). All participants underwent laboratory testing on the same day as infrared thermography imaging. A total of 1656 radiomic features were extracted from each participant's thermographs and refined using Pearson correlation coefficients, two-sample t-tests, and LASSO regression. A binary random forest (RF) classification model was then constructed and evaluated based on its calibration, discrimination, and clinical utility. The RF model demonstrated strong diagnostic performance, achieving an AUC of 0.91 in the validation cohort. Calibration and decision curve analyses confirmed the model's clinical applicability. Infrared thermography-based radiomics offers a promising, non-invasive method for early screening of MS, highlighting its potential clinical utility.
Collapse
Affiliation(s)
- Jiayang Guo
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Huizhong Xue
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoran Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yiyun Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xianhui Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanhong An
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Hua Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yimeng Yang
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Luqi Cai
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzheng Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Yonghua Xiao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Mravec B, Szantova M. Liver Neurobiology: Regulation of Liver Functions by the Nervous System. Semin Liver Dis 2025. [PMID: 40239709 DOI: 10.1055/a-2562-2000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The nervous system plays an important role in the regulation of liver functions during physiological as well as pathological conditions. This regulatory effect is based on the processing of signals transmitted to the brain by sensory nerves innervating the liver tissue and other visceral organs and by humoral pathways transmitting signals from peripheral tissues and organs. Based on these signals, the brain modulates metabolism, detoxification, regeneration, repair, inflammation, and other processes occurring in the liver. The nervous system thus determines the functional and morphological characteristics of the liver. Liver innervation also mediates the influence of psychosocial factors on liver functions. The aim of this review is to describe complexity of bidirectional interactions between the brain and liver and to characterize the mechanisms and pathways through which the nervous system influences liver function during physiological conditions and maintains liver and systemic homeostasis.
Collapse
Affiliation(s)
- Boris Mravec
- Department of Physiology Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
3
|
de Almeida LV, Santos-de-Araújo AD, da Silva LCN, Santos PM, Maia MC, Frutuoso VP, Rocha DS, Rêgo AS, Bassi-Dibai D. Cholesterol, triglycerides, HDL, and nitric oxide as determinants of resting heart rate variability in non-hospitalized mild post-COVID individuals: a cross-sectional study. BMC Cardiovasc Disord 2025; 25:69. [PMID: 39891044 PMCID: PMC11783953 DOI: 10.1186/s12872-025-04523-z] [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: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The association between plasma lipids, nitric oxide (NO) and cardiovascular risk has been well documented in the literature, however, the association between these outcomes and heart rate variability (HRV) in COVID-19 remains incipient as there is no scientific evidence that has investigated this outcome. OBJECTIVE Investigate whether metabolic outcomes may be associated with cardiac autonomic behavior arising from short-term HRV variables in non-hospitalized mild post-COVID individuals. METHODS This is a cross-sectional study. Individuals of both sexes, aged ≥ 18 years, who tested positive for SARS-CoV-2 according to the RT-PCR test, without the need for hospitalization, were included. The HRV was collected in the supine position for at least 10 min for later analysis in the Kubios software. Metabolic outcomes [high density lipoprotein (HDL) (mg/dL), cholesterol (mg/dL), triglycerides (mg/dL) and NO (µmol/L)] were collected through a blood sample. RESULTS Seventy-three individuals were included (post-COVID = 32; control = 41). HRV was worse in the post-COVID group when compared to the control group (p < 0.05). Cholesterol, HDL, triglycerides and NO showed significant correlations with HRV indices. Regression models indicated that cholesterol and triglycerides, as well as NO, explain up to 30.3% of the variations in certain HRV indices, suggesting an impact of metabolic outcomes on autonomic modulation. CONCLUSION There is a relationship between plasma lipids, NO and HRV in non-hospitalized individuals with mild COVID-19. Metabolic outcomes are associated and explain between 16.6% and 30.30% of certain variables of resting HRV in post-COVID individuals. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Lucivalda Viegas de Almeida
- Postgraduate Program in Programs Management and Health Services, Universidade Ceuma, Josué Montello, number 1, São Luís, 65075-120, MA, Brazil
| | - Aldair Darlan Santos-de-Araújo
- Postgraduate Program in Physical Therapy, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | | | | | | | | | - Daniel Santos Rocha
- Postgraduate program in Physical Education, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Adriana Sousa Rêgo
- Postgraduate Program in Environment, Universidade Ceuma, São Luís, MA, Brazil
| | - Daniela Bassi-Dibai
- Postgraduate Program in Programs Management and Health Services, Universidade Ceuma, Josué Montello, number 1, São Luís, 65075-120, MA, Brazil.
- Department of Physical Therapy, Universidade CEUMA, São Luís, MA, Brazil.
- Postgraduate program in Dentistry, Universidade Ceuma, São Luís, MA, Brazil.
- Healthy Living for Pandemic Event Protection (HL - PIVOT) Network, Chicago, IL, USA.
| |
Collapse
|
4
|
Wang JK, Zhang D, Wang JF, Lu WL, Wang JY, Liang SF, Liu R, Jiang JX, Li HT, Yang X. Clinical study on the effect of jejunoileal side-to-side anastomosis on metabolic parameters in patients with type 2 diabetes. World J Diabetes 2025; 16:99526. [PMID: 39817226 PMCID: PMC11718457 DOI: 10.4239/wjd.v16.i1.99526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/04/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND At present, the existing internal medicine drug treatment can alleviate the high glucose toxicity of patients to a certain extent, to explore the efficacy of laparoscopic jejunoileal side to side anastomosis in the treatment of type 2 diabetes, the report is as follows. AIM To investigate the effect of jejunoileal side-to-side anastomosis on metabolic parameters in patients with type 2 diabetes mellitus (T2DM). METHODS We retrospectively analyzed the clinical data of 78 patients with T2DM who were treated via jejunoileal lateral anastomosis. Metabolic indicators were collected preoperatively, as well as at 3 and 6 months postoperative. The metabolic indicators analyzed included body mass index (BMI), systolic blood pressure (SBP), diastolic blood pressure (DBP), fasting blood glucose (FBG), 2-hour blood glucose (PBG), glycated hemoglobin (HbA1c), fasting C-peptide, 2-hour C-peptide (PCP), fasting insulin (Fins), 2-hour insulin (Pins), insulin resistance index (HOMA-IR), β Cellular function index (HOMA-β), alanine aminotransferase, aspartate aminotransferase, serum total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), triglycerides (TG), high-density lipoprotein, and uric acid (UA) levels. RESULTS SBP, DBP, PBG, HbA1c, LDL-C, and TG were all significantly lower 3 months postoperative vs preoperative values; body weight, BMI, SBP, DBP, FBG, PBG, HbA1c, TC, TG, UA, and HOMA-IR values were all significantly lower 6 months postoperative vs at 3 months; and PCP, Fins, Pins, and HOMA-β were all significantly higher 6 months postoperative vs at 3 months (all P < 0.05). CONCLUSION Side-to-side anastomosis of the jejunum and ileum can effectively treat T2DM and improve the metabolic index levels associated with it.
Collapse
Affiliation(s)
- Ji-Kui Wang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Di Zhang
- Department of Health Management Center, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Jin-Feng Wang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Wan-Lin Lu
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Jing-Yuan Wang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Shi-Feng Liang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Ran Liu
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Jing-Xin Jiang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Hong-Tao Li
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| | - Xuan Yang
- Department of General Thoracic Surgery, Liaoning Electric Power Center Hospital, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
5
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [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: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
6
|
Mravec B, Szantova M. The role of the nervous system in liver diseases. Hepatol Res 2024; 54:970-980. [PMID: 39392763 DOI: 10.1111/hepr.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
The nervous system significantly participates in maintaining homeostasis, and modulating repair and regeneration processes in the liver. Moreover, the nervous system also plays an important role in the processes associated with the development and progression of liver disease, and can either potentiate or inhibit these processes. The aim of this review is to describe the mechanisms and pathways through which the nervous system influences the development and progression of liver diseases, such as alcohol-associated liver disease, nonalcoholic fatty liver disease, cholestatic liver disease, hepatitis, cirrhosis, and hepatocellular carcinoma. Possible therapeutic implications based on modulation of signals transduction between the nervous system and the liver are also discussed.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
7
|
Goulet N, Marcoux C, Bourgon V, Morin R, Mauger JF, Amaratunga R, Imbeault P. Biological sex-related differences in the postprandial triglyceride response to intermittent hypoxaemia in young adults: a randomized crossover trial. J Physiol 2024; 602:5817-5834. [PMID: 38285004 DOI: 10.1113/jp285430] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Abstract
Obstructive sleep apnoea is characterized by chronic intermittent hypoxaemia and is independently associated with an increased risk of metabolic comorbidities (e.g. type II diabetes and ischaemic heart disease). These comorbidities could be attributable to hypoxaemia-induced alterations in blood lipid profiles. However, it remains unclear whether intermittent hypoxaemia alters triglyceridaemia differently between biological sexes. Therefore, we used a randomized crossover design to examine whether 6 h of moderate intermittent hypoxaemia (15 hypoxaemic cycles/h, 85% oxyhaemoglobin saturation) alters plasma triglyceride levels differently between men and women after a high-fat meal. Relative to men, women displayed lower levels of total triglycerides, in addition to denser triglyceride-rich lipoprotein triglycerides (TRL-TG; mainly very low-density lipoprotein triglycerides and chylomicron remnant triglycerides) and buoyant TRL-TG (mainly chylomicron triglycerides) during normoxia (ambient air) and intermittent hypoxaemia (sex × time: all P ≤ 0.008). Intermittent hypoxaemia led to higher triglyceride levels (condition: all P ≤ 0.016); however, this effect was observed only in men (sex × condition: all P ≤ 0.002). Compared with normoxia, glucose levels were higher in men and lower in women during intermittent hypoxaemia (sex × condition: P < 0.001). The different postprandial responses between biological sexes occurred despite similar reductions in mean oxyhaemoglobin saturation and similar elevations in insulin levels, non-esterified fatty acid levels and mean heart rate (sex × condition: all P ≥ 0.185). These results support growing evidence showing that intermittent hypoxaemia impacts men and women differently, and they might help to explain biological sex-related discrepancies in the rate of certain comorbidities associated with intermittent hypoxaemia. KEY POINTS: Intermittent hypoxaemia is a key characteristic of obstructive sleep apnoea and alters lipid metabolism in multiple tissues, resulting in increased circulating triglyceride levels, an important risk factor for cardiometabolic diseases. Circulating triglyceride levels are regulated differently between biological sexes, with women typically displaying much lower fasting and postprandial triglyceride levels than men, partly explaining why women of all ages experience lower mortality rates from cardiometabolic diseases. In this study, healthy young men and women consumed a high-fat meal and were then exposed to 6 h of intermittent hypoxaemia or ambient air. We show that postprandial triglyceride levels are significantly lower in women compared with men and that intermittent hypoxaemia leads to higher postprandial triglyceride levels in men only. These results might help us to understand better why women living with obstructive sleep apnoea experience lower rates of cardiometabolic diseases (e.g. type II diabetes and ischaemic heart disease) than men living with obstructive sleep apnoea.
Collapse
Affiliation(s)
- Nicholas Goulet
- Behavioural and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Caroline Marcoux
- Behavioural and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Vincent Bourgon
- Laboratoire du Sommeil, Département de psychoéducation et de psychologie, Université du Québec en Outaouais, Gatineau, QC, Canada
| | - Renée Morin
- Behavioural and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Mauger
- Behavioural and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Ruwan Amaratunga
- Institut du Savoir Montfort, Montfort Hospital, Ottawa, ON, Canada
| | - Pascal Imbeault
- Behavioural and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort, Montfort Hospital, Ottawa, ON, Canada
| |
Collapse
|
8
|
Dutil C, Podinic I, Featherstone RB, Eaton A, Sadler CM, Goldfield GS, Hadjiyannakis S, Gruber R, Tremblay MS, Prud’homme D, Chaput JP. Sleep and insulin sensitivity in adolescents at risk of type 2 diabetes: the Sleep Manipulation in Adolescents at Risk of Type 2 Diabetes randomized crossover study. Sleep 2024; 47:zsad313. [PMID: 38070132 PMCID: PMC11082473 DOI: 10.1093/sleep/zsad313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/01/2023] [Indexed: 01/21/2024] Open
Abstract
STUDY OBJECTIVES To investigate the effect of increasing sleep duration for 1 week, compared to a week of habitual and decreased sleep, on insulin sensitivity (IS) in adolescents at risk for type 2 diabetes (T2D). METHODS Adolescents, 13-18 years old, at risk for T2D, with obesity and other risk factors, were recruited for a randomized (1:1), open-label, sex-stratified crossover study, that manipulated time-in-bed to modify sleep duration (measured by actigraphy). Following a week of habitual (HB) sleep, time-in-bed was increased (IN) and decreased (DE) by 1 hour 30 min/night for 1 week, counterbalanced across participants (HBINDE or HBDEIN), and separated by a week of washout sleep. The main outcome measure was IS, obtained via 2-hour oral-glucose-tolerance-test conducted after each sleep week. RESULTS Of the 43 participants recruited, 36 (84%) completed all sleep interventions (52.8% female, age = 15.1 years, body mass index = 99.9th percentile, order: HBINDE = 18 and HBDEIN = 18). On average, during the HB week, participants slept 7 hours 31 min/night; sleep duration was 1 hour 02 min/night higher during the IN week and 1 hour 19 min/night lower during the DE week. We found a significant effect of sleep week on IS with a large effect size. Following the IN sleep week, IS was 20% higher compared to after the HB and DE sleep weeks, but there was no significant difference in IS following HB versus DE sleep weeks. CONCLUSIONS Whenever possible, clinicians should empower youth at risk of T2D to improve their sleep duration, since even a modest increase in sleep duration of 1 h/night for 1 week can have a positive impact on IS in this population. CLINICAL TRIALS Sleep Extension and IS in Adolescents, https://clinicaltrials.gov/study/NCT03754036, November 23rd, 2018. TRIAL REGISTRATION ClinicalTrials.gov (ID:NCT03754036).
Collapse
Affiliation(s)
- Caroline Dutil
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Irina Podinic
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Ryan B Featherstone
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
- Centre for Healthy Active Living (CHAL), Children’s Hospital of Eastern Ontario (CHEO), Ottawa, ON, Canada
| | - Amelia Eaton
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Christin M Sadler
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Gary S Goldfield
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Stasia Hadjiyannakis
- Centre for Healthy Active Living (CHAL), Children’s Hospital of Eastern Ontario (CHEO), Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Reut Gruber
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Attention, Behaviour and Sleep Lab, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Mark S Tremblay
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Denis Prud’homme
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
- Université de Moncton, Moncton, NB, Canada
| | - Jean-Philippe Chaput
- Healthy Active Living and Obesity Research Group, Children’s Hospital of Eastern Ontario (CHEO) Research Institute, Ottawa, ON, Canada
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
包 汉, 王 苏, 吕 穆, 王 永, 姜 萍, 李 晓. [Activation of α7 nAChR improves white fat homeostasis and promotes beige adipogenesis and thermogenesis in obese mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:499-506. [PMID: 38597441 PMCID: PMC11006706 DOI: 10.12122/j.issn.1673-4254.2024.03.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 04/11/2024]
Abstract
OBJECTIVE To investigate the effects of α7 nicotinic acetylcholine receptor (nAChR) agonist on β3-adrenoceptor agonist-induced impairment of white fat homeostasis and beige adipose formation and heat production in obese mice. METHODS Forty obese C57BL/6J mice were randomized into high-fat feeding group, β3-adrenoceptor agonist-treated model group, α7 nAChR agonist group, and α7 nAChR inhibitor group (n=10), with another 10 mice with normal feeding as the blank control group. White adipose tissue from the epididymis of the mice were sampled for HE staining of the adipocytes. The expression levels of TNF-α, IL-1β, IL-10 and TGF-β in the white adipose tissue were determined by ELISA, and the mRNA levels of iNOS, Arg1, UCP-1, PRDM-16 and PGC-1α were detected using RT-qPCR. Western blotting was performed to detect the expression levels of NF-κB P65, p-JAK2, p-STAT3 in the white adipose tissue. RESULTS Compared with those in the blank control group, the mice with high-fat feeding showed significantly increased body weight, more fat vacuoles in the white adipose tissue, increased volume of lipid droplets in the adipocytes, upregulated iNOS mRNA expression and protein expression of TNF-α and IL-1β, and lowered expression of Arg-1 mRNA and IL-10 and TGF-β proteins (P < 0.01). Treatment with α7 nAChR significantly reduced mRNA levels of PRDM-16, PGC-1α and UCP-1, lowered TNF-α and IL-1β expressions, increased IL-10 and TGF-β expressions, and reduced M1/M2 macrophage ratio in the white adipose tissues (P < 0.05 or 0.01). CONCLUSION Activation of α7 nAchR improves white adipose tissue homeostasis impairment induced by β3 agonist, promotes transformation of M1 to M2 macrophages, reduces inflammatory response in white adipose tissue, and promote beige adipogenesis and thermogenesis in obese mice.
Collapse
Affiliation(s)
- 汉生 包
- 山东中医药大学,山东 济南 250355Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - 苏童 王
- 山东中医药大学,山东 济南 250355Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - 穆杰 吕
- 山东中医药大学,山东 济南 250355Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - 永成 王
- 山东中医药大学附属医院,山东 济南 250014Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - 萍 姜
- 山东中医药大学,山东 济南 250355Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - 晓 李
- 山东中医药大学附属医院,山东 济南 250014Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
10
|
Shen C, Chen Y, Wang Q, Sun Y, Lin H, Ni M, Chen Y, Zhang L, Jin J, Ying X, Zou Y, Wang X, Ye Y, Yuan M, Yin N, Cao Z, Zhang Y, Gu W, Wang W, Ning G, Wang J, Zhao S, Hong J, Liu R. Fecal short chain fatty acids modify therapeutic effects of sleeve gastrectomy. Front Endocrinol (Lausanne) 2023; 14:1277035. [PMID: 38027127 PMCID: PMC10663943 DOI: 10.3389/fendo.2023.1277035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Aims We aimed to investigate changes of fecal short chain fatty acids (SCFAs) and their association with metabolic benefits after sleeve gastrectomy (SG). Specifically, whether pre-surgery SCFAs modify surgical therapeutic effects was determined. Methods 62 participants with measurements of fecal SCFAs and metabolic indices before and 1, 3, 6 months after SG were included. Changes of fecal SCFAs and their association with post-surgery metabolic benefits were calculated. Then, participants were stratified by medians of pre-surgery fecal SCFAs and modification effects of pre-surgery fecal SCFAs on surgical therapeutic effects were investigated, through calculating interaction of group by surgery. Results Fecal SCFAs were markedly changed by SG. Changes of propionate and acetate were positively correlated with serum triglycerides and total cholesterol, respectively. Notably, high pre-surgery fecal hexanoate group showed a better effect of SG treatment on lowering body weight (P=0.01), BMI (P=0.041) and serum triglycerides (P=0.031), and low pre-surgery fecal butyrate had a better effect of SG on lowering ALT (P=0.003) and AST (P=0.019). Conclusion Fecal SCFAs were changed and correlated with lipid profiles improvement after SG. Pre-surgery fecal hexanoate and butyrate were potential modifiers impacting metabolic benefits of SG.
Collapse
Affiliation(s)
- Chongrong Shen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yanru Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Qiaoling Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Mengshan Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Ling Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiayang Ying
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyao Zou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Miaomiao Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Nan Yin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| |
Collapse
|
11
|
Oruganti L, Reddy Sankaran K, Dinnupati HG, Kotakadi VS, Meriga B. Anti-adipogenic and lipid-lowering activity of piperine and epigallocatechin gallate in 3T3-L1 adipocytes. Arch Physiol Biochem 2023; 129:1152-1159. [PMID: 33836628 DOI: 10.1080/13813455.2021.1908366] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 12/26/2022]
Abstract
The present study was aimed to evaluate the anti-adipogenic activity of piperine (PIP) and epigallocatechin gallate (EGCG) in 3T3-L1 cells. In cytotoxicity studies, PIP and EGCG showed IC50 values of 260 and 218 µM respectively and in combination (20 µM each) did not show cytotoxicity. Treatment with PIP and EGCG (20 µM each) significantly (p<.01) inhibited cell differentiation, lipid droplets deposition and enhanced glycerol release in 3T3-L1 cells. The secreted level of leptin was decreased but adiponectin level was increased in treated 3T3-L1 cells than untreated cells. In molecular expression studies, key adipogenic genes PPAR-γ, SREBP-1c, FAS, Fab-4, C/EBP-α and HMG-CoA reductase were markedly down-regulated but UCP-1 was up-regulated intreated 3T3-L1 cells and the same trend was observed in expression levels of selected proteins. In conclusion, our results demonstrated a combination of PIP and EGCG exhibited strong anti-adipogenic and lipid lowering effect than individual treatments due to synergism.
Collapse
Affiliation(s)
- Lokanatha Oruganti
- Division of Cell Culture and Molecular Biology, Department of Biochemistry, Sri Venkateswara University, Tirupati, India
| | - Karunakaran Reddy Sankaran
- Division of Cell Culture and Molecular Biology, Department of Biochemistry, Sri Venkateswara University, Tirupati, India
| | | | | | - Balaji Meriga
- Division of Cell Culture and Molecular Biology, Department of Biochemistry, Sri Venkateswara University, Tirupati, India
| |
Collapse
|
12
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
13
|
Badmus OO, Hinds TD, Stec DE. Mechanisms Linking Metabolic-Associated Fatty Liver Disease (MAFLD) to Cardiovascular Disease. Curr Hypertens Rep 2023; 25:151-162. [PMID: 37191842 PMCID: PMC10839567 DOI: 10.1007/s11906-023-01242-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE OF REVIEW Metabolic-associated fatty liver disease (MAFLD) is a condition of fat accumulation in the liver that occurs in the majority of patients in combination with metabolic dysfunction in the form of overweight or obesity. In this review, we highlight the cardiovascular complications in MAFLD patients as well as some potential mechanisms linking MAFLD to the development of cardiovascular disease and highlight potential therapeutic approaches to treating cardiovascular diseases in patients with MAFLD. RECENT FINDINGS MAFLD is associated with an increased risk of cardiovascular diseases (CVD), including hypertension, atherosclerosis, cardiomyopathies, and chronic kidney disease. While clinical data have demonstrated the link between MAFLD and the increased risk of CVD development, the mechanisms responsible for this increased risk remain unknown. MAFLD can contribute to CVD through several mechanisms including its association with obesity and diabetes, increased levels of inflammation, and oxidative stress, as well as alterations in hepatic metabolites and hepatokines. Therapies to potentially treat MAFLD-induced include statins and lipid-lowering drugs, glucose-lowering agents, antihypertensive drugs, and antioxidant therapy.
Collapse
Affiliation(s)
- Olufunto O Badmus
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, Barnstable Brown Diabetes Center, Markey Cancer Center, University of Kentucky, Lexington, KY, 40508, USA
| | - David E Stec
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
14
|
Carolina Costa Veras A, da Silva Bruzasco L, Beatriz Profiro Lopes A, da Silva Franco B, Spencer de Souza Holanda A, Maculano Esteves A, Milanski M, Souza Torsoni A, Martins Ignacio-Souza L, Alberto Torsoni M. Supplementation with CO induces lipogenesis in adipose tissue, leptin and insulin resistance in healthy Swiss mice. J Funct Foods 2023; 106:105600. [DOI: 10.1016/j.jff.2023.105600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
15
|
Cimas FJ, De la Cruz-Morcillo MÁ, Cifuentes C, Moratalla-López N, Alonso GL, Nava E, Llorens S. Effect of Crocetin on Basal Lipolysis in 3T3-L1 Adipocytes. Antioxidants (Basel) 2023; 12:1254. [PMID: 37371984 DOI: 10.3390/antiox12061254] [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: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Crocetin (CCT) is a natural saffron-derived apocarotenoid that possesses healthy properties such as anti-adipogenic, anti-inflammatory, and antioxidant activities. Lipolysis is enhanced in obesity and correlates with a pro-inflammatory, pro-oxidant state. In this context, we aimed to investigate whether CCT affects lipolysis. To evaluate CCT's possible lipolytic effect, 3T3-L1 adipocytes were treated with CCT10μM at day 5 post-differentiation. Glycerol content and antioxidant activity were assessed using colorimetric assays. Gene expression was measured using qRT-PCR to evaluate the effect of CCT on key lipolytic enzymes and on nitric oxide synthase (NOS) expression. Total lipid accumulation was assessed using Oil Red O staining. CCT10μM decreased glycerol release from 3T3-L1 adipocytes and downregulated adipose tissue triglyceride lipase (ATGL) and perilipin-1, but not hormone-sensitive lipase (HSL), suggesting an anti-lipolytic effect. CCT increased catalase (CAT) and superoxide dismutase (SOD) activity, thus showing an antioxidant effect. In addition, CCT exhibited an anti-inflammatory profile, i.e., diminished inducible NOS (NOS2) and resistin expression, while enhanced the expression of adiponectin. CCT10μM also decreased intracellular fat and C/EBPα expression (a transcription factor involved in adipogenesis), thus revealing an anti-adipogenic effect. These findings point to CCT as a promising biocompound for improving lipid mobilisation in obesity.
Collapse
Affiliation(s)
- Francisco J Cimas
- Mecenazgo COVID-19, Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Miguel Ángel De la Cruz-Morcillo
- Food Quality Research Group, Institute for Regional Development (IDR), Campus Universitario s/n, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain
| | - Carmen Cifuentes
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Natalia Moratalla-López
- Cátedra de Química Agrícola, Higher Technical School of Agronomic and Forestry Engineering and Biotechnology (ETSIAMB), University of Castilla-La Mancha (UCLM), Campus Universitario, 02006 Albacete, Spain
| | - Gonzalo L Alonso
- Cátedra de Química Agrícola, Higher Technical School of Agronomic and Forestry Engineering and Biotechnology (ETSIAMB), University of Castilla-La Mancha (UCLM), Campus Universitario, 02006 Albacete, Spain
| | - Eduardo Nava
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Sílvia Llorens
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| |
Collapse
|
16
|
Kuo HCN, LaRussa Z, Xu FM, West K, Consitt L, Davidson WS, Liu M, Coschigano KT, Shi H, Lo CC. Apolipoprotein A4 Elevates Sympathetic Activity and Thermogenesis in Male Mice. Nutrients 2023; 15:2486. [PMID: 37299447 PMCID: PMC10255745 DOI: 10.3390/nu15112486] [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: 04/21/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Long-chain fatty acids induce apolipoprotein A4 (APOA4) production in the small intestine and activate brown adipose tissue (BAT) thermogenesis. The increase in BAT thermogenesis enhances triglyceride clearance and insulin sensitivity. Acute administration of recombinant APOA4 protein elevates BAT thermogenesis in chow-fed mice. However, the physiological role of continuous infusion of recombinant APOA4 protein in regulating sympathetic activity, thermogenesis, and lipid and glucose metabolism in low-fat-diet (LFD)-fed mice remained elusive. The hypothesis of this study was that continuous infusion of mouse APOA4 protein would increase sympathetic activity and thermogenesis in BAT and subcutaneous inguinal white adipose tissue (IWAT), attenuate plasma lipid levels, and improve glucose tolerance. To test this hypothesis, sympathetic activity, BAT temperature, energy expenditure, body weight, fat mass, caloric intake, glucose tolerance, and levels of BAT and IWAT thermogenic and lipolytic proteins, plasma lipids, and markers of fatty acid oxidation in the liver in mice with APOA4 or saline treatment were measured. Plasma APOA4 levels were elevated, BAT temperature and thermogenesis were upregulated, and plasma triglyceride (TG) levels were reduced, while body weight, fat mass, caloric intake, energy expenditure, and plasma cholesterol and leptin levels were comparable between APOA4- and saline-treated mice. Additionally, APOA4 infusion stimulated sympathetic activity in BAT and liver but not in IWAT. APOA4-treated mice had greater fatty acid oxidation but less TG content in the liver than saline-treated mice had. Plasma insulin in APOA4-treated mice was lower than that in saline-treated mice after a glucose challenge. In conclusion, continuous infusion of mouse APOA4 protein stimulated sympathetic activity in BAT and the liver, elevated BAT thermogenesis and hepatic fatty acid oxidation, and consequently attenuated levels of plasma and hepatic TG and plasma insulin without altering caloric intake, body weight gain and fat mass.
Collapse
Affiliation(s)
- Hsuan-Chih N. Kuo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Zachary LaRussa
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Flora Mengyang Xu
- Department of Biology, Miami University, Oxford, OH 45056, USA; (F.M.X.); (H.S.)
| | - Kathryn West
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - Leslie Consitt
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - William Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA; (W.S.D.); (M.L.)
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA; (W.S.D.); (M.L.)
| | - Karen T. Coschigano
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA; (F.M.X.); (H.S.)
| | - Chunmin C. Lo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| |
Collapse
|
17
|
Kaur J, Singh DP, Kumar V, Kaur S, Bhunia RK, Kondepudi KK, Kuhad A, Bishnoi M. Transient Receptor Potential (TRP) based polypharmacological combination stimulates energy expending phenotype to reverse HFD-induced obesity in mice. Life Sci 2023; 324:121704. [PMID: 37075945 DOI: 10.1016/j.lfs.2023.121704] [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: 03/09/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND & AIM Obesity is a worldwide epidemic leading to decreased quality of life, higher medical expenses and significant morbidity. Enhancing energy expenditure and substrate utilization in adipose tissues through dietary constituents and polypharmacological approaches is gaining importance for the prevention and therapeutics of obesity. An important factor in this regard is Transient Receptor Potential (TRP) channel modulation and resultant activation of "brite" phenotype. Various dietary TRP channel agonists like capsaicin (TRPV1), cinnamaldehyde (TRPA1), and menthol (TRPM8) have shown anti-obesity effects, individually and in combination. We aimed to determine the therapeutic potential of such combination of sub-effective doses of these agents against diet-induced obesity, and explore the involved cellular processes. KEY FINDINGS The combination of sub-effective doses of capsaicin, cinnamaldehyde and menthol induced "brite" phenotype in differentiating 3T3-L1 cells and subcutaneous white adipose tissue of HFD-fed obese mice. The intervention prevented adipose tissue hypertrophy and weight gain, enhanced the thermogenic potential, mitochondrial biogenesis and overall activation of brown adipose tissue. These changes observed in vitro as well as in vivo, were linked to increased phosphorylation of kinases, AMPK and ERK. In the liver, the combination treatment enhanced insulin sensitivity, improved gluconeogenic potential and lipolysis, prevented fatty acid accumulation and enhanced glucose utilization. SIGNIFICANCE We report on the discovery of therapeutic potential of TRP-based dietary triagonist combination against HFD-induced abnormalities in metabolic tissues. Our findings indicate that a common central mechanism may affect multiple peripheral tissues. This study opens up avenues of development of therapeutic functional foods for obesity.
Collapse
Affiliation(s)
- Jasleen Kaur
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India; University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Dhirendra Pratap Singh
- Neurotoxicology and Immunotoxicology Laboratory, Division of Biological Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat 380016, India
| | - Vijay Kumar
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India
| | - Simranjit Kaur
- Department of Agricultural Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India
| | - Rupam Kumar Bhunia
- Department of Agricultural Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India
| | - Kanthi Kiran Kondepudi
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India
| | - Anurag Kuhad
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India.
| | - Mahendra Bishnoi
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
18
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
19
|
Eimonte M, Eimantas N, Baranauskiene N, Solianik R, Brazaitis M. Kinetics of lipid indicators in response to short- and long-duration whole-body, cold-water immersion. Cryobiology 2022; 109:62-71. [PMID: 36150503 DOI: 10.1016/j.cryobiol.2022.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 01/16/2023]
Abstract
Cold exposure-induced secretion of stress hormones activates cold-defense responses and mobilizes substrates for increased energy demands to fuel thermogenesis. However, it is unclear whether acute cold exposure-induced stress hormone response kinetics affect circulating lipid parameter kinetics. Therefore, we aimed to investigate the 2-day kinetics of stress hormones (i.e., cortisol, epinephrine, and norepinephrine) and the lipid profile (i.e., total cholesterol [TC], high-density lipoprotein [HDL] cholesterol, low-density lipoprotein [LDL] cholesterol, and triglycerides) in response to whole-body long- (intermittent 170 min; 170-CWI) or short-duration (10 min; 10-CWI) cold-water immersion (CWI; 14 °C water) in 17 healthy, young, adult men. Both CWI trials induced a marked release of the stress hormones, epinephrine, and norepinephrine, with higher concentrations detected after 170-CWI (p < 0.05) and a disrupted diurnal peak of cortisol lasting for a few hours. 170-CWI increased triglyceride levels from immediately after until 2 h after CWI, thereafter the concentration decreased at 4 h, 6 h, 1 day and 2 days after CWI (p < 0.05). Furthermore, the HDL-cholesterol level increased immediately after and at 6 h after 170-CWI (p < 0.05), while TC and LDL-cholesterol levels were not altered within 2 days. Lipid parameters were not affected within the 2 days after 10-CWI. Although both CWIs decreased deep body temperature and increased stress hormone levels for a few hours, only long-duration CWI induced changes in the circulating lipid profile within 2 days after CWI. This should be considered when discussing therapeutic protocols to improve circulating lipid profiles and ameliorate diseases associated with such profiles.
Collapse
Affiliation(s)
- Milda Eimonte
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.
| | - Nerijus Eimantas
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Neringa Baranauskiene
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Rima Solianik
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Marius Brazaitis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.
| |
Collapse
|
20
|
Temperature, cardiovascular mortality, and the role of hypertension and renin-angiotensin-aldosterone axis in seasonal adversity: a narrative review. J Hum Hypertens 2022; 36:1035-1047. [PMID: 35618875 DOI: 10.1038/s41371-022-00707-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Environmental temperature is now well known to have a U-shaped relationship with cardiovascular (CV) and all-cause mortality. Both heat and cold above and below an optimum temperature, respectively, are associated with adverse outcomes. However, cold in general and moderate cold specifically is predominantly responsible for much of temperature-attributable adversity. Importantly, hypertension-the most important CV risk factor-has seasonal variation such that BP is significantly higher in winter. Besides worsening BP control in established hypertensives, cold-induced BP increase also contributes to long-term BP variability among normotensive and pre-hypertensive patients, also a known CV risk factor. Disappointingly, despite the now well-stablished impact of temperature on BP and on CV mortality separately, direct linkage between seasonal BP change and CV outcomes remains preliminary. Proving or disproving this link is of immense clinical and public health importance because if seasonal BP variation contributes to seasonal adversity, this should be a modifiable risk. Mechanistically, existing evidence strongly suggests a central role of the sympathetic nervous system (SNS), and secondarily, the renin-angiotensin-aldosterone axis (RAAS) in mediating cold-induced BP increase. Though numerous other inflammatory, metabolic, and vascular perturbations likely also contribute, these may also well be secondary to cold-induced SNS/RAAS activation. This review aims to summarize the current evidence linking temperature, BP and CV outcomes. We also examine underlying mechanisms especially in regard to the SNS/RAAS axis, and highlight possible mitigation measures for clinicians.
Collapse
|
21
|
Soares MJ, Zhao Y, Calton EK, Pathak K. Triglycerides and systolic blood pressure negatively mediate the direct relationship of vitamin D status to resting energy expenditure: A cross sectional analysis. Diabetes Metab Syndr 2022; 16:102664. [PMID: 36402072 DOI: 10.1016/j.dsx.2022.102664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND AIMS We determined whether individual components of metabolic syndrome (MetS) mediated the direct association of vitamin D status (25OHD) on resting energy expenditure (REE). METHODS Multiple linear regression determined predictors of REE from data on 180 men and women from two ethnic groups. We then modelled a mediation pathway through components of MetS on the direct association between 25OHD and REE. The mediation modelling used the PROCESS SPSS Macro (version 4.0) based on 5000 bootstrapped samples, with the adjustment for different sets of covariates. RESULTS REE was significantly predicted by age, fat mass (FM), fat free mass (FFM), ethnicity, inverse ln insulin, 25OHD, triglycerides (TG), systolic blood pressure (SBP) and, to some extent, by time of REE measurements (p < 0.094). Adjustment for all these covariates, resulted in a negative indirect mediation effect of TG [β coefficient (bootstrapped SE): 0.95 (0.519); bootstrapped 95% CI: 2.172, -0.165; p < 0.05] and a concurrent negative mediation of SBP [β coefficient (bootstrapped SE): 0.72(0.484); bootstrapped 95% CI: 1.851, -0.011; p < 0.05]. There remained a positive direct pathway from 25OHD to REE [β coefficient (S.E): 4.715 (2.129); p = 0.028], however the total effect of 25OHD was dampened [β coefficient (S.E): 3.04 (2.126); p = 0.154]. CONCLUSIONS Independent of insulin sensitivity, a negative mediation by TG and SBP dampened the overall effect of 25OHD on REE.
Collapse
Affiliation(s)
- Mario J Soares
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley Campus, Perth, Western Australia, 6102, Australia.
| | - Yun Zhao
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley Campus, Perth, Western Australia, 6102, Australia
| | - Emily K Calton
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley Campus, Perth, Western Australia, 6102, Australia
| | - Kaveri Pathak
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley Campus, Perth, Western Australia, 6102, Australia
| |
Collapse
|
22
|
Straat ME, Jurado-Fasoli L, Ying Z, Nahon KJ, Janssen LG, Boon MR, Grabner GF, Kooijman S, Zimmermann R, Giera M, Rensen PC, Martinez-Tellez B. Cold exposure induces dynamic changes in circulating triacylglycerol species, which is dependent on intracellular lipolysis: A randomized cross-over trial. EBioMedicine 2022; 86:104349. [PMID: 36371986 PMCID: PMC9663865 DOI: 10.1016/j.ebiom.2022.104349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The application of cold exposure has emerged as an approach to enhance whole-body lipid catabolism. The global effect of cold exposure on the lipidome in humans has been reported with mixed results depending on intensity and duration of cold. METHODS This secondary study was based on data from a previous randomized cross-over trial (ClinicalTrials.gov ID: NCT03012113). We performed sequential lipidomic profiling in serum during 120 min cold exposure of human volunteers. Next, the intracellular lipolysis was blocked in mice (eighteen 10-week-old male wild-type mice C57BL/6J) using a small-molecule inhibitor of adipose triglyceride lipase (ATGL; Atglistatin), and mice were exposed to cold for a similar duration. The quantitative lipidomic profiling was assessed in-depth using the Lipidyzer platform. FINDINGS In humans, cold exposure gradually increased circulating free fatty acids reaching a maximum at 60 min, and transiently decreased total triacylglycerols (TAGs) only at 30 min. A broad range of TAG species was initially decreased, in particular unsaturated and polyunsaturated TAG species with ≤5 double bonds, while after 120 min a significant increase was observed for polyunsaturated TAG species with ≥6 double bonds in humans. The mechanistic study in mice revealed that the cold-induced increase in polyunsaturated TAGs was largely prevented by blocking adipose triglyceride lipase. INTERPRETATION We interpret these findings as that cold exposure feeds thermogenic tissues with TAG-derived fatty acids for combustion, resulting in a decrease of circulating TAG species, followed by increased hepatic production of polyunsaturated TAG species induced by liberation of free fatty acids stemming from adipose tissue. FUNDING This work was supported by the Netherlands CardioVascular Research Initiative: 'the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organisation for Health Research and Development and the Royal Netherlands Academy of Sciences' [CVON2017-20 GENIUS-II] to Patrick C.N. Rensen. Borja Martinez-Tellez is supported by individual postdoctoral grant from the Fundación Alfonso Martin Escudero and by a Maria Zambrano fellowship by the Ministerio de Universidades y la Unión Europea - NextGenerationEU (RR_C_2021_04). Lucas Jurado-Fasoli was supported by an individual pre-doctoral grant from the Spanish Ministry of Education (FPU19/01609) and with an Albert Renold Travel Fellowship from the European Foundation for the Study of Diabetes (EFSD). Martin Giera was partially supported by NWO XOmics project #184.034.019.
Collapse
Affiliation(s)
- Maaike E. Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Lucas Jurado-Fasoli
- PROmoting FITness and Health Through Physical Activity Research Group (PROFITH), Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Zhixiong Ying
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Kimberly J. Nahon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura G.M. Janssen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Gernot F. Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C.N. Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands,Corresponding author. Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| | - Borja Martinez-Tellez
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Chumasov EI, Petrova ES, Korzhevskii DE. Morphological Peculiarities of Innervation of Rat Epicardial Adipose Tissue in Early Postnatal Ontogenesis. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
24
|
Herrera Moro Chao D, Kirchner MK, Pham C, Foppen E, Denis RGP, Castel J, Morel C, Montalban E, Hassouna R, Bui LC, Renault J, Mouffle C, García-Cáceres C, Tschöp MH, Li D, Martin C, Stern JE, Luquet SH. Hypothalamic astrocytes control systemic glucose metabolism and energy balance. Cell Metab 2022; 34:1532-1547.e6. [PMID: 36198294 PMCID: PMC9615252 DOI: 10.1016/j.cmet.2022.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/23/2022] [Accepted: 09/02/2022] [Indexed: 01/29/2023]
Abstract
The hypothalamus is key in the control of energy balance. However, strategies targeting hypothalamic neurons have failed to provide viable options to treat most metabolic diseases. Conversely, the role of astrocytes in systemic metabolic control has remained largely unexplored. Here, we show that obesity promotes anatomically restricted remodeling of hypothalamic astrocyte activity. In the paraventricular nucleus (PVN) of the hypothalamus, chemogenetic manipulation of astrocytes results in bidirectional control of neighboring neuron activity, autonomic outflow, glucose metabolism, and energy balance. This process recruits a mechanism involving the astrocytic control of ambient glutamate levels, which becomes defective in obesity. Positive or negative chemogenetic manipulation of PVN astrocyte Ca2+ signals, respectively, worsens or improves metabolic status of diet-induced obese mice. Collectively, these findings highlight a yet unappreciated role for astrocytes in the direct control of systemic metabolism and suggest potential targets for anti-obesity strategy.
Collapse
Affiliation(s)
| | - Matthew K Kirchner
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Cuong Pham
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Ewout Foppen
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphael G P Denis
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France; Institut Cochin, Université Paris Cité, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Chloe Morel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Enrica Montalban
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Rim Hassouna
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Linh-Chi Bui
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Justine Renault
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Christine Mouffle
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Cristina García-Cáceres
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany; Medizinische Klinik and Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias H Tschöp
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany
| | - Dongdong Li
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Serge H Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France.
| |
Collapse
|
25
|
Chumasov EI, Petrova ES, Korzhevskii DE. Peculiarities of the Innervation of Epicardial Adipose Tissue in a Rat with Aging (Immunohistochemical Study). ADVANCES IN GERONTOLOGY 2022. [DOI: 10.1134/s2079057022030055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Nabi S, Bhandari U, Haque SE. Saroglitazar ameliorates monosodium glutamate-induced obesity and associated inflammation in Wistar rats: Plausible role of NLRP3 inflammasome and NF- κB. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:827-841. [PMID: 36033946 PMCID: PMC9392566 DOI: 10.22038/ijbms.2022.64041.14102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Inflammation is the major progenitor of obesity and associated metabolic disorders. The current study investigated the modulatory role of saroglitazar on adipocyte dysfunction and associated inflammation in monosodium glutamate (MSG) obese Wistar rats. MATERIALS AND METHODS The molecular docking simulation studies of saroglitazar and fenofibrate were performed on the ligand-binding domain of NLRP3 and NF- κB. Under in vivo study, neonatal pups received normal saline or MSG (4 g/kg, SC) for 7 alternate days after birth. After keeping for 42 days as such, animals were divided into seven groups: Normal control; MSG control; MSG + saroglitazar (2 mg/kg); MSG + saroglitazar (4 mg/kg); saroglitazar (4 mg/kg) per se; MSG + fenofibrate (100 mg/kg); fenofibrate (100 mg/kg) per se. Drug treatments were given orally, from the 42nd to 70th day. On day 71, blood was collected and animals were sacrificed for isolation of liver and fat pads. RESULTS In silico study showed significant binding of saroglitazar and fenofibrate against NLRP3 and NF- κB. Saroglitazar significantly reduced body weight, body mass index, Lee's index, fat pad weights, adiposity index, decreased serum lipids, interleukin-1β (IL-1β), tumor necrosis factor-α(TNF-α), interleukin-6 (IL-6), leptin, insulin, blood glucose, HOMA-IR values, oxidative stress in the liver and increased hepatic low-density lipoprotein receptor levels. Histopathological analysis of the liver showed decreased inflammation and vacuolization, and reduced adipocyte cell size. Immunohistochemical analysis showed suppression of NLRP3 in epididymal adipocytes and NF- κB expression in the liver. CONCLUSION Saroglitazar ameliorated obesity and associated inflammation via modulation of NLRP3 inflammasome and NF- κB in MSG obese Wistar rats.
Collapse
Affiliation(s)
- Sayima Nabi
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, (UGC approved deemed to be University, Govt. of India), New Delhi–110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, (UGC approved deemed to be University, Govt. of India), New Delhi–110062, India,Corresponding author: Uma Bhandari. Department of Pharmacology SPER, Jamia Hamdard New Delhi, India – 110062. uma_bora @hotmail.com
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, (UGC approved deemed to be University, Govt. of India), New Delhi–110062, India
| |
Collapse
|
27
|
Straat ME, Martinez-Tellez B, Sardjoe Mishre A, Verkleij MMA, Kemmeren M, Pelsma ICM, Alcantara JMA, Mendez-Gutierrez A, Kooijman S, Boon MR, Rensen PCN. Cold-Induced Thermogenesis Shows a Diurnal Variation That Unfolds Differently in Males and Females. J Clin Endocrinol Metab 2022; 107:1626-1635. [PMID: 35176767 PMCID: PMC9113803 DOI: 10.1210/clinem/dgac094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Indexed: 11/21/2022]
Abstract
CONTEXT Cold exposure mobilizes lipids to feed thermogenic processes in organs, including brown adipose tissue (BAT). In rodents, BAT metabolic activity exhibits a diurnal rhythm, which is highest at the start of the wakeful period. OBJECTIVE We investigated whether cold-induced thermogenesis displays diurnal variation in humans and differs between the sexes. METHODS This randomized crossover study included 24 young and lean male (n = 12) and female (n = 12) participants who underwent 2.5-hour personalized cooling using water-perfused mattresses in the morning (7:45 am) and evening (7:45 pm), with 1 day in between. We measured energy expenditure (EE) and supraclavicular skin temperature in response to cold exposure. RESULTS In males, cold-induced EE was higher in the morning than in the evening (+54% ± 10% vs +30% ± 7%; P = 0.05) but did not differ between morning and evening in females (+37% ± 9% vs +30% ± 10%; P = 0.42). Only in males, supraclavicular skin temperature upon cold increased more in morning than evening (+0.2 ± 0.1 °C vs -0.2 ± 0.2 °C; P = 0.05). In males, circulating free fatty acid (FFA) levels were increased after morning cold exposure, but not evening (+90% ± 18% vs +9% ± 8%; P < 0.001). In females, circulating FFA (+94% ± 21% vs +20% ± 5%; P = 0.006), but also triglycerides (+42% ± 5% vs +29% ± 4%, P = 0.01) and cholesterol levels (+17% ± 2% vs 11% ± 2%; P = 0.05) were more increased after cold exposure in morning than in evening. CONCLUSION Cold-induced thermogenesis is higher in morning than evening in males; however, lipid metabolism is more modulated in the morning than the evening in females.
Collapse
Affiliation(s)
- Maaike E Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Borja Martinez-Tellez
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Aashley Sardjoe Mishre
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Magdalena M A Verkleij
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Mirjam Kemmeren
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Iris C M Pelsma
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Juan M A Alcantara
- PROFITH “PROmoting FITness and Health Through Physical Activity” Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Andrea Mendez-Gutierrez
- Department of Biochemistry and Molecular Biology II, “José Mataix Verdú” Institute of Nutrition and Food Technology, Center of Biomedical Research, University of Granada, Granada, Spain
- Biohealth Research Institute in Granada (ibs.GRANADA), Granada, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| |
Collapse
|
28
|
Hachiya R, Tanaka M, Itoh M, Suganami T. Molecular mechanism of crosstalk between immune and metabolic systems in metabolic syndrome. Inflamm Regen 2022; 42:13. [PMID: 35490239 PMCID: PMC9057063 DOI: 10.1186/s41232-022-00198-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is currently considered as a molecular basis of metabolic syndrome. Particularly, obesity-induced inflammation in adipose tissue is the origin of chronic inflammation of metabolic syndrome. Adipose tissue contains not only mature adipocytes with large lipid droplets, but also a variety of stromal cells including adipocyte precursors, vascular component cells, immune cells, and fibroblasts. However, crosstalk between those various cell types in adipose tissue in obesity still remains to be fully understood. We focus on two innate immune receptors, Toll-like receptor 4 (TLR4) and macrophage-inducible C-type lectin (Mincle). We provided evidence that adipocyte-derived saturated fatty acids (SFAs) activate macrophage TLR4 signaling pathway, thereby forming a vicious cycle of inflammatory responses during the development of obesity. Intriguingly, the TLR4 signaling pathway is modulated metabolically and epigenetically: SFAs augment TLR4 signaling through the integrated stress response and chromatin remodeling, such as histone methylation, regulates dynamic transcription patterns downstream of TLR4 signaling. Another innate immune receptor Mincle senses cell death, which is a trigger of chronic inflammatory diseases including obesity. Macrophages form a histological structure termed “crown-like structure (CLS)”, in which macrophages surround dead adipocytes to engulf cell debris and residual lipids. Mincle is exclusively expressed in macrophages forming the CLS in obese adipose tissue and regulates adipocyte death-triggered adipose tissue fibrosis. In addition to adipose tissue, we found a structure similar to CLS in the liver of nonalcoholic steatohepatitis (NASH) and the kidney after acute kidney injury. This review article highlights the recent progress of the crosstalk between immune and metabolic systems in metabolic syndrome, with a focus on innate immune receptors.
Collapse
Affiliation(s)
- Rumi Hachiya
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Department of Pediatrics, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michiko Itoh
- Department of Metabolic Syndrome and Nutritional Science, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan. .,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
29
|
Lelou E, Corlu A, Nesseler N, Rauch C, Mallédant Y, Seguin P, Aninat C. The Role of Catecholamines in Pathophysiological Liver Processes. Cells 2022; 11:cells11061021. [PMID: 35326472 PMCID: PMC8947265 DOI: 10.3390/cells11061021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last few years, the number of research publications about the role of catecholamines (epinephrine, norepinephrine, and dopamine) in the development of liver diseases such as liver fibrosis, fatty liver diseases, or liver cancers is constantly increasing. However, the mechanisms involved in these effects are not well understood. In this review, we first recapitulate the way the liver is in contact with catecholamines and consider liver implications in their metabolism. A focus on the expression of the adrenergic and dopaminergic receptors by the liver cells is also discussed. Involvement of catecholamines in physiological (glucose metabolism, lipids metabolism, and liver regeneration) and pathophysiological (impact on drug-metabolizing enzymes expression, liver dysfunction during sepsis, fibrosis development, or liver fatty diseases and liver cancers) processes are then discussed. This review highlights the importance of understanding the mechanisms through which catecholamines influence liver functions in order to draw benefit from the adrenergic and dopaminergic antagonists currently marketed. Indeed, as these molecules are well-known drugs, their use as therapies or adjuvant treatments in several liver diseases could be facilitated.
Collapse
Affiliation(s)
- Elise Lelou
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
| | - Anne Corlu
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
| | - Nicolas Nesseler
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
- CHU Rennes, Department of Anesthesia and Critical Care, F-35000 Rennes, France
| | - Claudine Rauch
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
| | - Yannick Mallédant
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
- CHU Rennes, Department of Anesthesia and Critical Care, F-35000 Rennes, France
| | - Philippe Seguin
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
- CHU Rennes, Department of Anesthesia and Critical Care, F-35000 Rennes, France
| | - Caroline Aninat
- INSERM, Université Rennes, INRAE, Institut NuMeCan, Nutrition, Metabolisms and Cancer, F-35000 Rennes, France; (E.L.); (A.C.); (N.N.); (C.R.); (Y.M.); (P.S.)
- Correspondence: ; Tel.: +33-2-23-23-48-68
| |
Collapse
|
30
|
Bharti V, Bhardwaj A, Elias DA, Metcalfe AWS, Kim JS. A Systematic Review and Meta-Analysis of Lipid Signatures in Post-traumatic Stress Disorder. Front Psychiatry 2022; 13:847310. [PMID: 35599759 PMCID: PMC9120430 DOI: 10.3389/fpsyt.2022.847310] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/12/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Research assessing lipid levels in individuals diagnosed with post-traumatic stress disorder (PTSD) has yielded mixed results. This study aimed to employ meta-analytic techniques to characterize the relationship between the levels of lipid profiles and PTSD. METHODS We performed meta-analyses of studies comparing profiles and levels of lipids between PTSD patients and healthy individuals by searching Embase, Ovid Medline, Scopus, PsycINFO, and Cochrane databases for the studies until March 2021. Meta-analyses were performed using random-effects models with the restricted maximum-likelihood estimator to synthesize the effect size assessed by standardized mean difference (SMD) across studies. FINDINGS A total of 8,657 abstracts were identified, and 17 studies were included. Levels of total cholesterol (TC) (SMD = 0.57 95% CI, 0.27-0.87, p = 0.003), low-density lipoprotein (LDL) (SMD = 0.48, 95% CI, 0.19-0.76, p = 0.004), and triglyceride (TG) (SMD = 0.46, 95% CI, 0.22-0.70, p = 0.001) were found to be higher, while levels of high-density lipoprotein (HDL) (SMD = -0.47, -0.88 to -0.07, p = 0.026) were found to be lower in PTSD patients compared to healthy controls. Subgroup analysis showed that TG levels were higher in PTSD patients who were on or off of psychotropic medications, both < 40 and ≥ 40 years of age, and having body mass index of < 30 and ≥ 30 compared to healthy controls. INTERPRETATION This work suggested dysregulation of lipids in PTSD that may serve as biomarker to predict the risk. The study will be useful for physicians considering lipid profiles in PTSD patients to reduce cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Veni Bharti
- Department of Community Health and Epidemiology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.,Health and Environments Research Centre (HERC) Laboratory, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Aseem Bhardwaj
- Health and Environments Research Centre (HERC) Laboratory, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - David A Elias
- Canadian Health Solutions Inc., Saint John, NB, Canada.,Dalhousie Medicine New Brunswick, Dalhousie University, Halifax, NS, Canada
| | - Arron W S Metcalfe
- Canadian Health Solutions Inc., Saint John, NB, Canada.,Canadian Imaging Research Centre, Saint John, NB, Canada
| | - Jong Sung Kim
- Department of Community Health and Epidemiology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.,Health and Environments Research Centre (HERC) Laboratory, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
31
|
Ito T, Yamamoto Y, Yamagishi N, Kanai Y. Stomach secretes estrogen in response to the blood triglyceride levels. Commun Biol 2021; 4:1364. [PMID: 34876651 PMCID: PMC8651635 DOI: 10.1038/s42003-021-02901-9] [Citation(s) in RCA: 4] [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: 03/16/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022] Open
Abstract
Mammals receive body energy information to maintain energy homeostasis. Ghrelin, insulin, leptin and vagal afferents transmit the status of fasting, blood glucose, body fat, and food intake, respectively. Estrogen also inhibits feeding behavior and lipogenesis, but increases body fat mass. However, how blood triglyceride levels are monitored and the physiological roles of estrogen from the perspective of lipid homeostasis remain unsettled. Here, we show that stomach secretes estrogen in response to the blood triglyceride levels. Estrogen-secreting gastric parietal cells predominantly use fatty acids as an energy source. Blood estrogen levels increase as blood triglyceride levels rise in a stomach-dependent manner. Estrogen levels in stomach tissues increase as blood triglyceride levels rise, and isolated gastric gland epithelium produces estrogen in a fatty acid-dependent manner. We therefore propose that stomach monitors and controls blood triglyceride levels using estrogen, which inhibits feeding behavior and lipogenesis, and promotes triglyceride uptake by adipocytes.
Collapse
Affiliation(s)
- Takao Ito
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuta Yamamoto
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoko Yamagishi
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
32
|
Xu L, Füredi N, Lutter C, Geenen B, Pétervári E, Balaskó M, Dénes Á, Kovács KJ, Gaszner B, Kozicz T. Leptin coordinates efferent sympathetic outflow to the white adipose tissue through the midbrain centrally-projecting Edinger-Westphal nucleus in male rats. Neuropharmacology 2021; 205:108898. [PMID: 34861283 DOI: 10.1016/j.neuropharm.2021.108898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/29/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022]
Abstract
The centrally-projecting Edinger-Westphal nucleus (EWcp) hosts a large population of neurons expressing urocortin 1 (Ucn1) and about half of these neurons also express the leptin receptor (LepRb). Previously, we have shown that the peripheral adiposity hormone leptin signaling energy surfeit modulates EWcp neurons' activity. Here, we hypothesized that Ucn1/LepRb neurons in the EWcp would act as a crucial neuronal node in the brain-white adipose tissue (WAT) axis modulating efferent sympathetic outflow to the WAT. We showed that leptin bound to neurons of the EWcp stimulated STAT3 phosphorylation, and increased Ucn1-production in a time-dependent manner. Besides, retrograde transneuronal tract-tracing using pseudorabies virus (PRV) identified EWcp Ucn1 neurons connected to WAT. Interestingly, reducing EWcp Ucn1 contents by ablating EWcp LepRb-positive neurons with leptin-saporin, did not affect food intake and body weight gain, but substantially (+26%) increased WAT weight accompanied by a higher plasma leptin level and changed plasma lipid profile. We also found that ablation of EWcp Ucn1/LepRb neurons resulted in lower respiratory quotient and oxygen consumption one week after surgery, but was comparable to sham values after 3 and 5 weeks of surgery. Taken together, we report that EWcp/LepRb/Ucn1 neurons not only respond to leptin signaling but also control WAT size and fat metabolism without altering food intake. These data suggest the existence of a EWcp-WAT circuitry allowing an organism to recruit fuels without being able to eat in situations such as the fight-or-flight response.
Collapse
Affiliation(s)
- Lu Xu
- Department of Anatomy Medical Imaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands; Department of Structural and Cellular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Nóra Füredi
- Department of Anatomy and Center for Neuroscience, Medical School, Pécs University, Pécs, Hungary; Department of Translational Medicine, Medical School, Pécs University, Pécs, Hungary
| | - Christoph Lutter
- Department of Anatomy and Center for Neuroscience, Medical School, Pécs University, Pécs, Hungary
| | - Bram Geenen
- Department of Anatomy Medical Imaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Erika Pétervári
- Department of Translational Medicine, Medical School, Pécs University, Pécs, Hungary
| | - Márta Balaskó
- Department of Translational Medicine, Medical School, Pécs University, Pécs, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Krisztina J Kovács
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Budapest, Hungary
| | - Balázs Gaszner
- Department of Anatomy and Center for Neuroscience, Medical School, Pécs University, Pécs, Hungary.
| | - Tamás Kozicz
- Department of Anatomy Medical Imaging, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands; Department of Clinical Genomics, Mayo Clinic, MN, USA; Department of Laboratory Medicine and Pathology, Mayo Clinic, MN, USA; Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
33
|
Sex differences in the association between nonalcoholic fatty liver disease and Parkinson's disease. Parkinsonism Relat Disord 2021; 93:19-26. [PMID: 34763304 DOI: 10.1016/j.parkreldis.2021.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/13/2021] [Accepted: 10/31/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The association between nonalcoholic fatty liver disease (NAFLD) and Parkinson's disease (PD) remains uncertain. This study sought to assess the sex-specific association between NAFLD and PD risk considering that sex is an important factor in both conditions. METHODS We included 2,651,169 men and 2,998,904 women (≥40 years of age) who underwent health examinations in 2009 using database of the Korean National Health Insurance Service. To define NAFLD, the Fatty Liver Index (FLI) was used and a score of at least 60 points was regarded as suggesting the presence of NAFLD. Cox proportional hazards analyses were performed to evaluate the association between the presence of NAFLD/each component of FLI and the risk of PD. All analyses were stratified by sex. RESULTS The median follow-up duration was 7.3 years in both men and women. Of the total study population, 23,233 patients with PD (10,578 men and 12,655 women) were identified. Among men, a decreased risk of PD was observed in those with NAFLD [adjusted hazard ratio (aHR): 0.86, 95% confidence interval (CI): 0.82-0.91]. In contrast, among women, an increased risk of PD was observed in those with NAFLD (aHR: 1.09, 95% CI: 1.02-1.16). This different association according to sex was more prominent among younger participants but was not significant in the old age group. CONCLUSIONS NAFLD defined by FLI is differently associated with the risk of PD by sex (i.e., low risk of PD in men with NAFLD vs. high risk of PD in women with NAFLD).
Collapse
|
34
|
Hardoňová M, Šiarnik P, Siváková M, Suchá B, Vlček M, Imrich R, Turčáni P, Havranová A, Rádiková Ž, Žitňanová I, Dean Z, Penesová A, Kollár B. Autonomic Nervous System Function in Newly Diagnosed Multiple Sclerosis: Association With Lipid Levels and Insulin Resistance. Physiol Res 2021; 70:875-882. [PMID: 34717060 DOI: 10.33549/physiolres.934695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Autonomic nervous system (ANS) disorders are common in multiple sclerosis (MS). Previous studies showed differences in insulin resistance (IR) and lipoprotein levels in MS subjects compared to controls. Lipolysis caused by increased sympathetic activity could be one of the possible linking mechanisms leading to dyslipidemia in MS. Our study aimed to evaluate ANS activity in the context of glucose and lipid metabolism in people with MS. We prospectively measured short-term heart rate variability (HRV), fasting lipoprotein concentrations, and calculated IR indices based on plasma glucose and insulin levels during oral glucose tolerance test (oGTT) in 32 patients with MS and 29 healthy controls matched for age, sex and body mass index in our study. There was no significant difference in HRV parameters and lipoprotein levels between MS and controls. A significant positive correlation was found between low/high-frequency power ratio (LF/HF) and triglycerides (r=0.413, p=0.021) in MS subjects but not in controls. A significantly lower whole-body insulin sensitivity index (ISIMat) was found in patients with MS compared to the control group (7.3±3.7 vs. 9.8±5.6, p=0.041). No significant correlations were found between LF/HF and IR parameters. In MS subjects, the positive correlation of LF/HF with triglycerides could reflect the effects of sympathetic activity on lipolysis. Positive correlations of sympathetic activity with increased lipoprotein levels could rather reflect processes associated with immune system activation/inflammation, than processes involved in glucose homeostasis maintenance.
Collapse
Affiliation(s)
- M Hardoňová
- 1st Department of Neurology, Faculty of Medicine, Comenius University, Slovakia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Morin R, Goulet N, Mauger JF, Imbeault P. Physiological Responses to Hypoxia on Triglyceride Levels. Front Physiol 2021; 12:730935. [PMID: 34497541 PMCID: PMC8419320 DOI: 10.3389/fphys.2021.730935] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/03/2021] [Indexed: 11/25/2022] Open
Abstract
Hypoxia is a condition during which the body or specific tissues are deprived of oxygen. This phenomenon can occur in response to exposure to hypoxic environmental conditions such as high-altitude, or because of pathophysiological conditions such as obstructive sleep apnea. Circumstances such as these can restrict supply or increase consumption of oxygen, leading to oxyhemoglobin desaturation and tissue hypoxia. In certain cases, hypoxia may lead to severe health consequences such as an increased risk of developing cardiovascular diseases and type 2 diabetes. A potential explanation for the link between hypoxia and an increased risk of developing cardiovascular diseases lies in the disturbing effect of hypoxia on circulating blood lipids, specifically its capacity to increase plasma triglyceride concentrations. Increased circulating triglyceride levels result from the production of triglyceride-rich lipoproteins, such as very-low-density lipoproteins and chylomicrons, exceeding their clearance rate. Considerable research in murine models reports that hypoxia may have detrimental effects on several aspects of triglyceride metabolism. However, in humans, the mechanisms underlying the disturbing effect of hypoxia on triglyceride levels remain unclear. In this mini-review, we outline the available evidence on the physiological responses to hypoxia and their impact on circulating triglyceride levels. We also discuss mechanisms by which hypoxia affects various organs involved in the metabolism of triglyceride-rich lipoproteins. This information will benefit scientists and clinicians interested in the mechanistic of the regulatory cascade responsible for the response to hypoxia and how this response could lead to a deteriorated lipid profile and an increased risk of developing hypoxia-related health consequences.
Collapse
Affiliation(s)
- Renée Morin
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Nicholas Goulet
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Mauger
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Pascal Imbeault
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.,Hôpital Montfort, Institut du Savoir Montfort, Ottawa, ON, Canada
| |
Collapse
|
36
|
Assinder SJ, Boumelhem BB. Oxytocin stimulates lipolysis, prostaglandin E 2 synthesis, and leptin secretion in 3T3-L1 adipocytes. Mol Cell Endocrinol 2021; 534:111381. [PMID: 34216640 DOI: 10.1016/j.mce.2021.111381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 11/26/2022]
Abstract
A model of oxytocin in the regulation of metabolic status has described one of oxytocin synthesis and release from the neurohypophysis in response to leptin, to suppress further leptin release. In addition, a lipogenic role for oxytocin has been suggested, consistent with an insulinergic action. This model, however, may be incorrect. Oxytocin reduces fat mass in the absence of either leptin or leptin receptor signalling, thereby challenging the interdependence between leptin and oxytocin. An oxytocin induced production of the anti-lipolytic prostaglandin E2 (PGE2) might account for this. Media from 3T3-L1 differentiated adipocytes treated with oxytocin (0-50 nmol.L-1) for 24 hrs were assayed for PGE2, leptin, adiponectin, and glycerol. Harvested cells were analysed for lipid droplet triglyceride and cytosolic free fatty acid (FFA) by flow cytometry, and for altered expression of lipolytic and lipogenic associated gene ontology transcripts by cDNA array. Both PGE2 and leptin secretion were significantly increased by oxytocin treatment whilst adiponectin secretion was not. A significant increase in cytosolic FFA was detected following oxytocin treatment, similar to that determined following treatment with isoproterenol (positive control). A significant increase in glycerol release to the culture media confirmed a lipolytic effect. No enrichment of lipolytic and lipogenic associated gene ontology transcripts was determined, but significant overrepresentation of chemosensory olfactory transcripts was. In conclusion, oxytocin stimulates lipolysis in 3T3-L1 adipocytes, mediated by autocrine/paracrine actions of PGE2 and leptin. To confirm that this response is mediated solely by the oxytocin receptor, further experiments would require those effects being blocked by a specific oxytocin antagonist.
Collapse
Affiliation(s)
- Stephen J Assinder
- Discipline of Physiology, School of Medical Science and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Australia.
| | - Badwi B Boumelhem
- Discipline of Physiology, School of Medical Science and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Australia
| |
Collapse
|
37
|
Liang X, Zhang P, Luo S, Zhang G, Tang X, Liu L. The association of quality of life and personality characteristics with adolescent metabolic syndrome: a cohort study. Health Qual Life Outcomes 2021; 19:160. [PMID: 34103067 PMCID: PMC8186050 DOI: 10.1186/s12955-021-01797-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND An increased prevalence of adolescent metabolic syndrome (MS) is associated with adulthood cardiovascular diseases. This study aimed to explore the potential relationship of quality of life (QoL) and personality traits with adolescent MS. METHODS A total of 1961 participants from Chongqing with an average age of 11.68 years old from a cohort study established in 2014 and followed up through 2019 were included. QoL information, Eysenck's personality questionnaire and MS components were collected. RESULTS A higher QoL domain score of physical activity ability (PAA) was a protective factor for both MS and MS score (all P < 0.01), which was mainly negatively correlated with the MS components of central obesity, diastolic blood pressure (DBP) and triglyceride levels, as well as positively correlated with high density lipoprotein cholesterol (HDL-C) level. The total QoL score was negatively correlated with triglyceride levels and positively correlated with DBP (all P < 0.01). High extraversion personality score was a protective factor against adolescent MS (P = 0.04) and MS score (P < 0.05), which were mainly negatively correlated with the MS components of waist circumference, systolic blood pressure and TGs, and positively correlated with HDL-C (all P ≤ 0.01). CONCLUSIONS QoL score and extraversion personality score were independent protective factors against both MS prevalence and MS score, suggesting that community intervention to improve the QoL and psychological health of children are essential.
Collapse
Affiliation(s)
- Xiaohua Liang
- Clinical Epidemiology and Biostatistics Department, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center of Child Development and Critical Disorders, 136 2nd Street, Yuzhong District, Chongqing, 400016, China.
| | - Peng Zhang
- Disease Control and Prevention Center of Jiulongpo District, Chongqing, China
| | - Shunqing Luo
- Medical General Ward of Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Guifang Zhang
- Plastic Surgery Department of Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xian Tang
- Clinical Epidemiology and Biostatistics Department, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center of Child Development and Critical Disorders, 136 2nd Street, Yuzhong District, Chongqing, 400016, China
| | - Lingjuan Liu
- Clinical Epidemiology and Biostatistics Department, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center of Child Development and Critical Disorders, 136 2nd Street, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
38
|
Liang X, Zhang P, Luo S, Zhang G, Tang X, Liu L. The association of quality of life and personality characteristics with adolescent metabolic syndrome: a cohort study. Health Qual Life Outcomes 2021. [DOI: https://doi.org/10.1186/s12955-021-01797-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract
Background
An increased prevalence of adolescent metabolic syndrome (MS) is associated with adulthood cardiovascular diseases. This study aimed to explore the potential relationship of quality of life (QoL) and personality traits with adolescent MS.
Methods
A total of 1961 participants from Chongqing with an average age of 11.68 years old from a cohort study established in 2014 and followed up through 2019 were included. QoL information, Eysenck’s personality questionnaire and MS components were collected.
Results
A higher QoL domain score of physical activity ability (PAA) was a protective factor for both MS and MS score (all P < 0.01), which was mainly negatively correlated with the MS components of central obesity, diastolic blood pressure (DBP) and triglyceride levels, as well as positively correlated with high density lipoprotein cholesterol (HDL-C) level. The total QoL score was negatively correlated with triglyceride levels and positively correlated with DBP (all P < 0.01). High extraversion personality score was a protective factor against adolescent MS (P = 0.04) and MS score (P < 0.05), which were mainly negatively correlated with the MS components of waist circumference, systolic blood pressure and TGs, and positively correlated with HDL-C (all P ≤ 0.01).
Conclusions
QoL score and extraversion personality score were independent protective factors against both MS prevalence and MS score, suggesting that community intervention to improve the QoL and psychological health of children are essential.
Collapse
|
39
|
Siqueira BS, Ceglarek VM, Gomes ECZ, Vettorazzi JF, Rentz T, Nenevê JZ, Volinski KZ, Moraes SS, Malta A, de Freitas Mathias PC, de Oliveira Emilio HR, Balbo SL, Grassiolli S. Vagotomy and Splenectomy Reduce Insulin Secretion and Interleukin-1β. Pancreas 2021; 50:607-616. [PMID: 33939676 DOI: 10.1097/mpa.0000000000001809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES This study aimed to evaluate the effect of vagotomy, when associated with splenectomy, on adiposity and glucose homeostasis in Wistar rats. METHODS Rats were divided into 4 groups: vagotomized (VAG), splenectomized (SPL), VAG + SPL, and SHAM. Glucose tolerance tests were performed, and physical and biochemical parameters evaluated. Glucose-induced insulin secretion and protein expression (Glut2/glucokinase) were measured in isolated pancreatic islets. Pancreases were submitted to histological and immunohistochemical analyses, and vagus nerve neural activity was recorded. RESULTS The vagotomized group presented with reduced body weight, growth, and adiposity; high food intake; reduced plasma glucose and triglyceride levels; and insulin resistance. The association of SPL with the VAG surgery attenuated, or abolished, the effects of VAG and reduced glucose-induced insulin secretion and interleukin-1β area in β cells, in addition to lowering vagal activity. CONCLUSIONS The absence of the spleen attenuated or blocked the effects of VAG on adiposity, triglycerides and glucose homeostasis, suggesting a synergistic effect of both on metabolism. The vagus nerve and spleen modulate the presence of interleukin-1β in β cells, possibly because of the reduction of glucose-induced insulin secretion, indicating a bidirectional flow between autonomous neural firing and the spleen, with repercussions for the endocrine pancreas.
Collapse
Affiliation(s)
- Bruna Schumaker Siqueira
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Vanessa Marieli Ceglarek
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | | | | | - Thiago Rentz
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas
| | - Juliane Zanon Nenevê
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Karoline Zanella Volinski
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Sandra Schmidt Moraes
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá (UEM), Maringá
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá (UEM), Maringá
| | | | - Sandra Lucinei Balbo
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Sabrina Grassiolli
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| |
Collapse
|
40
|
Van Drunen R, Eckel-Mahan K. Circadian Rhythms of the Hypothalamus: From Function to Physiology. Clocks Sleep 2021; 3:189-226. [PMID: 33668705 PMCID: PMC7931002 DOI: 10.3390/clockssleep3010012] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
The nearly ubiquitous expression of endogenous 24 h oscillations known as circadian rhythms regulate the timing of physiological functions in the body. These intrinsic rhythms are sensitive to external cues, known as zeitgebers, which entrain the internal biological processes to the daily environmental changes in light, temperature, and food availability. Light directly entrains the master clock, the suprachiasmatic nucleus (SCN) which lies in the hypothalamus of the brain and is responsible for synchronizing internal rhythms. However, recent evidence underscores the importance of other hypothalamic nuclei in regulating several essential rhythmic biological functions. These extra-SCN hypothalamic nuclei also express circadian rhythms, suggesting distinct regions that oscillate either semi-autonomously or independent of SCN innervation. Concurrently, the extra-SCN hypothalamic nuclei are also sensitized to fluctuations in nutrient and hormonal signals. Thus, food intake acts as another powerful entrainer for the hypothalamic oscillators' mediation of energy homeostasis. Ablation studies and genetic mouse models with perturbed extra-SCN hypothalamic nuclei function reveal their critical downstream involvement in an array of functions including metabolism, thermogenesis, food consumption, thirst, mood and sleep. Large epidemiological studies of individuals whose internal circadian cycle is chronically disrupted reveal that disruption of our internal clock is associated with an increased risk of obesity and several neurological diseases and disorders. In this review, we discuss the profound role of the extra-SCN hypothalamic nuclei in rhythmically regulating and coordinating body wide functions.
Collapse
Affiliation(s)
- Rachel Van Drunen
- MD Anderson UTHealth School Graduate School of Biomedical Sciences, Houston TX 77030, USA;
- Brown Foundation Institute of Molecular Medicine University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- MD Anderson UTHealth School Graduate School of Biomedical Sciences, Houston TX 77030, USA;
- Brown Foundation Institute of Molecular Medicine University of Texas McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
41
|
Central Apolipoprotein A-IV Stimulates Thermogenesis in Brown Adipose Tissue. Int J Mol Sci 2021; 22:ijms22031221. [PMID: 33513710 PMCID: PMC7865537 DOI: 10.3390/ijms22031221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
Stimulation of thermogenesis in brown adipose tissue (BAT) could have far-reaching health benefits in combatting obesity and obesity-related complications. Apolipoprotein A-IV (ApoA-IV), produced by the gut and the brain in the presence of dietary lipids, is a well-known short-term satiating protein. While our previous studies have demonstrated reduced diet-induced thermogenesis in ApoA-IV-deficient mice, it is unclear whether this reduction is due to a loss of peripheral or central effects of ApoA-IV. We hypothesized that central administration of ApoA-IV stimulates BAT thermogenesis and that sympathetic and sensory innervation is necessary for this action. To test this hypothesis, mice with unilateral denervation of interscapular BAT received central injections of recombinant ApoA-IV protein or artificial cerebrospinal fluid (CSF). The effects of central ApoA-IV on BAT temperature and thermogenesis in mice with unilateral denervation of the intrascapular BAT were monitored using transponder probe implantation, qPCR, and immunoblots. Relative to CSF, central administration of ApoA-IV significantly increased temperature and UCP expression in BAT. However, all of these effects were significantly attenuated or prevented in mice with unilateral denervation. Together, these results clearly demonstrate that ApoA-IV regulates BAT thermogenesis centrally, and this effect is mediated through sympathetic and sensory nerves.
Collapse
|
42
|
Luo S, Ezrokhi M, Cominos N, Tsai TH, Stoelzel CR, Trubitsyna Y, Cincotta AH. Experimental dopaminergic neuron lesion at the area of the biological clock pacemaker, suprachiasmatic nuclei (SCN) induces metabolic syndrome in rats. Diabetol Metab Syndr 2021; 13:11. [PMID: 33485386 PMCID: PMC7825247 DOI: 10.1186/s13098-021-00630-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The daily peak in dopaminergic neuronal activity at the area of the biological clock (hypothalamic suprachiasmatic nuclei [SCN]) is diminished in obese/insulin resistant vs lean/insulin sensitive animals. The impact of targeted lesioning of dopamine (DA) neurons specifically at the area surrounding (and that communicate with) the SCN (but not within the SCN itself) upon glucose metabolism, adipose and liver lipid gene expression, and cardiovascular biology in normal laboratory animals has not been investigated and was the focus of this study. METHODS Female Sprague-Dawley rats received either DA neuron neurotoxic lesion by bilateral intra-cannula injection of 6-hydroxydopamine (2-4 μg/side) or vehicle treatment at the area surrounding the SCN at 20 min post protriptyline ip injection (20 mg/kg) to protect against damage to noradrenergic and serotonergic neurons. RESULTS At 16 weeks post-lesion relative to vehicle treatment, peri-SCN area DA neuron lesioning increased weight gain (34.8%, P < 0.005), parametrial and retroperitoneal fat weight (45% and 90% respectively, P < 0.05), fasting plasma insulin, leptin and norepinephrine levels (180%, 71%, and 40% respectively, P < 0.05), glucose tolerance test area under the curve (AUC) insulin (112.5%, P < 0.05), and insulin resistance (44%-Matsuda Index, P < 0.05) without altering food consumption during the test period. Such lesion also induced the expression of several lipid synthesis genes in adipose and liver and the adipose lipolytic gene, hormone sensitive lipase in adipose (P < 0.05 for all). Liver monocyte chemoattractant protein 1 (a proinflammatory protein associated with metabolic syndrome) gene expression was also significantly elevated in peri-SCN area dopaminergic lesioned rats. Peri-SCN area dopaminergic neuron lesioned rats were also hypertensive (systolic BP rose from 157 ± 5 to 175 ± 5 mmHg, P < 0.01; diastolic BP rose from 109 ± 4 to 120 ± 3 mmHg, P < 0.05 and heart rate increase from 368 ± 12 to 406 ± 12 BPM, P < 0.05) and had elevated plasma norepinephrine levels (40% increased, P < 0.05) relative to controls. CONCLUSIONS These findings indicate that reduced dopaminergic neuronal activity in neurons at the area of and communicating with the SCN contributes significantly to increased sympathetic tone and the development of metabolic syndrome, without effect on feeding.
Collapse
Affiliation(s)
- Shuqin Luo
- VeroScience LLC, 1334 Main Road, Tiverton, RI, 02878, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Effect of Adrenergic Agonists on High-Fat Diet-Induced Hepatic Steatosis in Mice. Int J Mol Sci 2020; 21:ijms21249392. [PMID: 33321735 PMCID: PMC7764675 DOI: 10.3390/ijms21249392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
The autonomic nervous system, consisting of sympathetic and parasympathetic branches, plays an important role in regulating metabolic homeostasis. The sympathetic nervous system (SNS) regulates hepatic lipid metabolism by regulating adrenergic receptor activation, resulting in the stimulation of hepatic very-low-density lipoprotein-triglyceride (TG) production in vivo. However, only a few studies on the relationship between SNS and hepatic steatosis have been reported. Here, we investigate the effect of adrenergic receptor agonists on hepatic steatosis in mice fed a high-fat diet (HFD). The α-adrenergic receptor agonist phenylephrine (10 mg/kg/d) or the β-adrenergic receptor agonist isoproterenol (30 mg/kg/d) was coadministered with HFD to male mice. After five weeks, hepatic steatosis, TG levels, and hepatic fat metabolism-related biomarkers were examined. HFD treatment induced hepatic steatosis, and cotreatment with phenylephrine, but not isoproterenol, attenuated this effect. Phenylephrine administration upregulated the mRNA levels of hepatic peroxisome proliferator-activated receptor alpha and its target genes (such as carnitine palmitoyltransferase 1) and increased hepatic β-hydroxybutyrate levels. Additionally, phenylephrine treatment increased the expression of the autophagosomal marker LC3-II but decreased that of p62, which is selectively degraded during autophagy. These results indicate that phenylephrine inhibits hepatic steatosis through stimulation of β-oxidation and autophagy in the liver.
Collapse
|
44
|
Takata K, Goto T, Kuroda M, Kimura Y, Harada I, Ueda K, Kawada T, Kioka N. Stiffness of the extracellular matrix regulates differentiation into beige adipocytes. Biochem Biophys Res Commun 2020; 532:205-210. [PMID: 32859378 DOI: 10.1016/j.bbrc.2020.08.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 11/15/2022]
Abstract
Beige/brite adipocytes, which express high levels of uncoupling protein 1 (UCP1) to generate heat using stored triglycerides, are induced under specific stimuli such as cold exposure in inguinal white adipose tissue (iWAT). Although extracellular microenvironments such as extracellular matrix (ECM) stiffness are known to regulate cell behaviors, including cell differentiation into adipocytes, the effect on iWAT cells is unknown. In this study, we show that rigid ECM promotes the cell spreading of iWAT-derived preadipocytes. Furthermore, the expression of UCP1 and other thermogenic genes in iWAT cells is promoted when the cells are cultured on rigid ECM. The expression of mTOR, a kinase known to regulate the differentiation to beige adipocytes, is decreased on rigid substrates. These results suggest that ECM stiffness plays an important role in the differentiation to beige adipocytes.
Collapse
Affiliation(s)
- Kyoko Takata
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Technology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Mito Kuroda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Yasuhisa Kimura
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Ichiro Harada
- Medical Products Technology Development Center, R&D Headquarters, Canon Inc., Ohta-ku, Tokyo, 146-8501, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo, Kyoto, 606-8501, Japan
| | - Teruo Kawada
- Division of Food Science and Technology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo, Kyoto, 606-8501, Japan.
| |
Collapse
|
45
|
Lebrun B, Barbot M, Tonon MC, Prévot V, Leprince J, Troadec JD. Glial endozepines and energy balance: Old peptides with new tricks. Glia 2020; 69:1079-1093. [PMID: 33105065 DOI: 10.1002/glia.23927] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
The contribution of neuroglial interactions to the regulation of energy balance has gained increasing acceptance in recent years. In this context, endozepines, endogenous analogs of benzodiazepine derived from diazepam-binding inhibitor, are now emerging as major players. Produced by glial cells (astrocytes and tanycytes), endozepines have been known for two decades to exert potent anorexigenic effects by acting at the hypothalamic level. However, it is only recently that their modes of action, including the mechanisms by which they modulate energy metabolism, have begun to be elucidated. The data available today are abundant, significant, and sometimes contradictory, revealing a much more complex regulation than initially expected. Several mechanisms of action of endozepines seem to coexist at the central level, particularly in the hypothalamus. The brainstem has also recently emerged as a potential site of action for endozepines. In addition to their central anorexigenic effects, endozepines may also display peripheral effects promoting orexigenic actions, adding to their complexity and raising yet more questions. In this review, we attempt to provide an overview of our current knowledge in this rapidly evolving field and to pinpoint questions that remain unanswered.
Collapse
Affiliation(s)
- Bruno Lebrun
- CNRS 7291, Laboratoire de Neurosciences Cognitives, Aix Marseille University, Marseille, France
| | - Manon Barbot
- CNRS 7291, Laboratoire de Neurosciences Cognitives, Aix Marseille University, Marseille, France
| | - Marie-Christine Tonon
- INSERM U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Normandie Université, Rouen, France
| | - Vincent Prévot
- University of Lille, INSERM, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, European Genomic Institute of Diabetes (EGID), Lille, France
| | - Jérôme Leprince
- INSERM U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Normandie Université, Rouen, France
| | - Jean-Denis Troadec
- CNRS 7291, Laboratoire de Neurosciences Cognitives, Aix Marseille University, Marseille, France
| |
Collapse
|
46
|
Xiang AS, Giles C, Loh RK, Formosa MF, Eikelis N, Lambert GW, Meikle PJ, Kingwell BA, Carey AL. Plasma Docosahexaenoic Acid and Eicosapentaenoic Acid Concentrations Are Positively Associated with Brown Adipose Tissue Activity in Humans. Metabolites 2020; 10:metabo10100388. [PMID: 32998426 PMCID: PMC7601733 DOI: 10.3390/metabo10100388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/14/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022] Open
Abstract
Brown adipose tissue (BAT) activation is a possible therapeutic strategy to increase energy expenditure and improve metabolic homeostasis in obesity. Recent studies have revealed novel interactions between BAT and circulating lipid species—in particular, the non-esterified fatty acid (NEFA) and oxylipin lipid classes. This study aimed to identify individual lipid species that may be associated with cold-stimulated BAT activity in humans. A panel of 44 NEFA and 41 oxylipin species were measured using mass-spectrometry-based lipidomics in the plasma of fourteen healthy male participants before and after 90 min of mild cold exposure. Lipid measures were correlated with BAT activity measured via 18F-fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT), along with norepinephrine (NE) concentration (a surrogate marker of sympathetic activity). The study identified a significant increase in total NEFA concentration following cold exposure that was positively associated with NE concentration change. Individually, 33 NEFA and 11 oxylipin species increased significantly in response to cold exposure. The concentration of the omega-3 NEFA, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) at baseline was significantly associated with BAT activity, and the cold-induced change in 18 NEFA species was significantly associated with BAT activity. No significant associations were identified between BAT activity and oxylipins.
Collapse
Affiliation(s)
- Angie S. Xiang
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; (A.S.X.); (R.K.C.L.); (M.F.F.); (B.A.K.); (A.L.C.)
- Central Clinical School, Monash University, Clayton, Melbourne 3004, Australia
| | - Corey Giles
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia;
- Correspondence: ; Tel.: +61-3-8532-1536
| | - Rebecca K.C. Loh
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; (A.S.X.); (R.K.C.L.); (M.F.F.); (B.A.K.); (A.L.C.)
- Department of Physiology, Monash University, Clayton, Melbourne 3800, Australia
| | - Melissa F. Formosa
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; (A.S.X.); (R.K.C.L.); (M.F.F.); (B.A.K.); (A.L.C.)
| | - Nina Eikelis
- Iverson Health Innovation Research Institute, Swinburne Institute of Technology, Melbourne 3122, Australia; (N.E.); (G.W.L.)
| | - Gavin W. Lambert
- Iverson Health Innovation Research Institute, Swinburne Institute of Technology, Melbourne 3122, Australia; (N.E.); (G.W.L.)
| | - Peter J. Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia;
| | - Bronwyn A. Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; (A.S.X.); (R.K.C.L.); (M.F.F.); (B.A.K.); (A.L.C.)
- Central Clinical School, Monash University, Clayton, Melbourne 3004, Australia
- Department of Physiology, Monash University, Clayton, Melbourne 3800, Australia
- Research Therapeutic Area, CSL Limited, Parkville 3052, Australia
| | - Andrew L. Carey
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; (A.S.X.); (R.K.C.L.); (M.F.F.); (B.A.K.); (A.L.C.)
- Department of Physiology, Monash University, Clayton, Melbourne 3800, Australia
| |
Collapse
|
47
|
Mahú I, Barateiro A, Rial-Pensado E, Martinéz-Sánchez N, Vaz SH, Cal PMSD, Jenkins B, Rodrigues T, Cordeiro C, Costa MF, Mendes R, Seixas E, Pereira MMA, Kubasova N, Gres V, Morris I, Temporão C, Olivares M, Sanz Y, Koulman A, Corzana F, Sebastião AM, López M, Bernardes GJL, Domingos AI. Brain-Sparing Sympathofacilitators Mitigate Obesity without Adverse Cardiovascular Effects. Cell Metab 2020; 31:1120-1135.e7. [PMID: 32402266 PMCID: PMC7671941 DOI: 10.1016/j.cmet.2020.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 03/03/2020] [Accepted: 04/14/2020] [Indexed: 02/02/2023]
Abstract
Anti-obesity drugs in the amphetamine (AMPH) class act in the brain to reduce appetite and increase locomotion. They are also characterized by adverse cardiovascular effects with origin that, despite absence of any in vivo evidence, is attributed to a direct sympathomimetic action in the heart. Here, we show that the cardiac side effects of AMPH originate from the brain and can be circumvented by PEGylation (PEGyAMPH) to exclude its central action. PEGyAMPH does not enter the brain and facilitates SNS activity via theβ2-adrenoceptor, protecting mice against obesity by increasing lipolysis and thermogenesis, coupled to higher heat dissipation, which acts as an energy sink to increase energy expenditure without altering food intake or locomotor activity. Thus, we provide proof-of-principle for a novel class of exclusively peripheral anti-obesity sympathofacilitators that are devoid of any cardiovascular and brain-related side effects.
Collapse
Affiliation(s)
- Inês Mahú
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Andreia Barateiro
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal; Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Eva Rial-Pensado
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, A Coruña 15782, Spain
| | - Noelia Martinéz-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Sandra H Vaz
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof., Egas Moniz, Lisbon 1649-028, Portugal; Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Pedro M S D Cal
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof., Egas Moniz, Lisbon 1649-028, Portugal
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Wellcome Trust, MRL Institute of Metabolic Science, University of Cambridge, Pathology building Level 4, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Tiago Rodrigues
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof., Egas Moniz, Lisbon 1649-028, Portugal
| | - Carlos Cordeiro
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Faculdade de Ciências da Universidade de Lisboa, Lisbon 1749-016, Portugal
| | - Miguel F Costa
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal; Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon 1049-001, Portugal
| | - Raquel Mendes
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Elsa Seixas
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Mafalda M A Pereira
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Nadiya Kubasova
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Vitka Gres
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Imogen Morris
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Carolina Temporão
- Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Marta Olivares
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council, Valencia (IATA-CSIC), Catedratico Agustin Escardino 7, 46980, Paterna, Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council, Valencia (IATA-CSIC), Catedratico Agustin Escardino 7, 46980, Paterna, Valencia, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Wellcome Trust, MRL Institute of Metabolic Science, University of Cambridge, Pathology building Level 4, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Francisco Corzana
- Departamento de Química, Universidad de La Rioja, Centro de Investigación en Síntesis Química, 26006 Logroño, Spain
| | - Ana M Sebastião
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof., Egas Moniz, Lisbon 1649-028, Portugal; Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, A Coruña 15782, Spain
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof., Egas Moniz, Lisbon 1649-028, Portugal; Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK; Obesity Laboratory, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal; Howard Hughes Medical Institute, IGC, Oeiras, Portugal.
| |
Collapse
|
48
|
Zhu Q, Weng J, Shen M, Fish J, Shen Z, Coschigano KT, Davidson WS, Tso P, Shi H, Lo CC. Apolipoprotein A-IV Enhances Fatty Acid Uptake by Adipose Tissues of Male Mice via Sympathetic Activation. Endocrinology 2020; 161:5802681. [PMID: 32157301 PMCID: PMC7100924 DOI: 10.1210/endocr/bqaa042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/04/2020] [Indexed: 12/31/2022]
Abstract
Apolipoprotein A-IV (ApoA-IV) synthesized by the gut regulates lipid metabolism. Sympathetic innervation of adipose tissues also controls lipid metabolism. We hypothesized that ApoA-IV required sympathetic innervation to increase fatty acid (FA) uptake by adipose tissues and brown adipose tissue (BAT) thermogenesis. After 3 weeks feeding of either a standard chow diet or a high-fat diet (HFD), mice with unilateral denervation of adipose tissues received intraperitoneal administration of recombinant ApoA-IV protein and intravenous infusion of lipid mixture with radioactive triolein. In chow-fed mice, ApoA-IV administration increased FA uptake by intact BAT but not the contralateral denervated BAT or intact white adipose tissue (WAT). Immunoblots showed that, in chow-fed mice, ApoA-IV increased expression of lipoprotein lipase and tyrosine hydroxylase in both intact BAT and inguinal WAT (IWAT), while ApoA-IV enhanced protein levels of β3 adrenergic receptor, adipose triglyceride lipase, and uncoupling protein 1 in the intact BAT only. In HFD-fed mice, ApoA-IV elevated FA uptake by intact epididymal WAT (EWAT) but not intact BAT or IWAT. ApoA-IV increased sympathetic activity assessed by norepinephrine turnover (NETO) rate in BAT and EWAT of chow-fed mice, whereas it elevated NETO only in EWAT of HFD-fed mice. These observations suggest that, in chow-fed mice, ApoA-IV activates sympathetic activity of BAT and increases FA uptake by BAT via innervation, while in HFD-fed mice, ApoA-IV stimulates sympathetic activity of EWAT to shunt FAs into the EWAT.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Biology, Miami University, Oxford, OH
| | - Jonathan Weng
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Minqian Shen
- Department of Biology, Miami University, Oxford, OH
| | - Jace Fish
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Zhujun Shen
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Karen T Coschigano
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH
| | - Chunmin C Lo
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
- Correspondence: Chunmin C Lo, Department of Biomedical Sciences, Irvine Hall 228, 1 Ohio University, Athens, OH 45701-2979. E-mail:
| |
Collapse
|
49
|
Straat ME, Schinkelshoek MS, Fronczek R, Lammers GJ, Rensen PCN, Boon MR. Role of Brown Adipose Tissue in Adiposity Associated With Narcolepsy Type 1. Front Endocrinol (Lausanne) 2020; 11:145. [PMID: 32373062 PMCID: PMC7176868 DOI: 10.3389/fendo.2020.00145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/02/2020] [Indexed: 11/23/2022] Open
Abstract
Narcolepsy type 1 is a neurological sleep-wake disorder caused by the destruction of orexin (hypocretin)-producing neurons. These neurons are particularly located in the lateral hypothalamus and have widespread projections throughout the brain, where they are involved, e.g., in the regulation of the sleep-wake cycle and appetite. Interestingly, a higher prevalence of obesity has been reported in patients with narcolepsy type 1 compared to healthy controls, despite a normal to decreased food intake and comparable physical activity. This suggests the involvement of tissues implicated in total energy expenditure, including skeletal muscle, liver, white adipose tissue (WAT), and brown adipose tissue (BAT). Recent evidence from pre-clinical studies with orexin knock-out mice demonstrates a crucial role for the orexin system in the functionality of brown adipose tissue (BAT), probably through multiple pathways. Since BAT is a highly metabolically active organ that combusts fatty acids and glucose toward heat, thereby contributing to energy metabolism, this raises the question of whether BAT plays a role in the development of obesity and related metabolic diseases in narcolepsy type 1. BAT is densely innervated by the sympathetic nervous system that activates BAT, for instance, following cold exposure. The sympathetic outflow toward BAT is mainly mediated by the dorsomedial, ventromedial, arcuate, and paraventricular nuclei in the hypothalamus. This review focuses on the current knowledge on the role of the orexin system in the control of energy balance, with specific focus on BAT metabolism and adiposity in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Maaike E. Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Maaike E. Straat
| | - Mink S. Schinkelshoek
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Rolf Fronczek
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Gerrit Jan Lammers
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
- Sleep Wake Centre SEIN, Heemstede, Netherlands
| | - Patrick C. N. Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
50
|
Chamarthi B, Vinik A, Ezrokhi M, Cincotta AH. Circadian-timed quick-release bromocriptine lowers elevated resting heart rate in patients with type 2 diabetes mellitus. Endocrinol Diabetes Metab 2020; 3:e00101. [PMID: 31922028 PMCID: PMC6947713 DOI: 10.1002/edm2.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/09/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Sympathetic nervous system (SNS) overactivity is a risk factor for insulin resistance and cardiovascular disease (CVD). We evaluated the impact of bromocriptine-QR, a dopamine-agonist antidiabetes medication, on elevated resting heart rate (RHR) (a marker of SNS overactivity in metabolic syndrome), blood pressure (BP) and the relationship between bromocriptine-QR's effects on RHR and HbA1c in type 2 diabetes subjects. DESIGN AND SUBJECTS RHR and BP changes were evaluated in this post hoc analysis of data from a randomized controlled trial in 1014 type 2 diabetes subjects randomized to bromocriptine-QR vs placebo added to standard therapy (diet ± ≤2 oral antidiabetes medications) for 24 weeks without concomitant antihypertensive or antidiabetes medication changes, stratified by baseline RHR (bRHR). RESULTS In subjects with bRHR ≥70 beats/min, bromocriptine-QR vs placebo reduced RHR by -3.4 beats/min and reduced BP (baseline 130/79; systolic, diastolic, mean arterial BP reductions [mm Hg]: -3.6 [P = .02], -1.9 [P = .05], -2.5 [P = .02]). RHR reductions increased with higher baseline HbA1c (bHbA1c) (-2.7 [P = .03], -5 [P = .002], -6.1 [P = .002] with bHbA1c ≤7, >7, ≥7.5%, respectively] in the bRHR ≥70 group and more so with bRHR ≥80 (-4.5 [P = .07], -7.8 [P = .015], -9.9 [P = .005]). Subjects with bRHR <70 had no significant change in RHR or BP. With bHbA1c ≥7.5%, %HbA1c reductions with bromocriptine-QR vs placebo were -0.50 (P = .04), -0.73 (P = .005) and -1.22 (P = .008) with bRHR <70, ≥70 and ≥80, respectively. With bRHR ≥70, the magnitude of bromocriptine-QR-induced RHR reduction was an independent predictor of bromocriptine-QR's HbA1c lowering effect. CONCLUSION Bromocriptine-QR lowers elevated RHR with concurrent decrease in BP and hyperglycaemia. These findings suggest a potential sympatholytic mechanism contributing to bromocriptine-QR's antidiabetes effect and potentially its previously demonstrated effect to reduce CVD events.
Collapse
Affiliation(s)
| | - Aaron Vinik
- Eastern Virginia Medical School Strelitz Diabetes CenterNorfolkVirginia
| | | | | |
Collapse
|