1
|
Shan B, Zhao C, Peng C, Miao Y, Lei S, Zhao L, Jia M, Pan S, Gong J, Wang Q. Theabrownin from Pu-erh tea attenuated high-fat diet-induced metabolic syndrome in rat by regulating microRNA and affecting gut microbiota. Int J Biol Macromol 2024; 285:138368. [PMID: 39638201 DOI: 10.1016/j.ijbiomac.2024.138368] [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: 08/28/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Theabrownin (TB), the primary pigment in Pu-erh tea, has shown potential in alleviating metabolic syndrome (MS), though its precise mechanisms remain unclear. This study investigated the effects of Pu-erh tea water extract (WE) and TB on high-fat diet (HFD)-induced MS in rats, focusing on miRNA regulation and gut microbiota composition. Both WE and TB significantly improved markers of MS, including dyslipidemia, insulin resistance, and inflammation. These improvements were linked to the normalization of specific miRNAs (miR-125b-5p, miR-223-3p_R + 2, miR-148b-3p, and miR-1247-5p), which activated the PI3K/AKT/FOXO signaling pathway, subsequently modulating key genes involved in glucolipid metabolism (SREBP-1C, PEPCK, PGC-1α, and G6pc). Additionally, WE and TB restored gut microbiota balance by decreasing the Firmicutes/Bacteroidetes ratio and increasing beneficial bacteria such as Bacteroides, Lactobacillus, and Bifidobacterium, while reducing harmful bacteria like Pseudomonas. These findings underscore the potential of theabrownin as a functional food component for MS prevention, offering new insights into its miRNA-mediated and microbiota-related mechanisms.
Collapse
Affiliation(s)
- Bo Shan
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunyan Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunxiu Peng
- College of Horticulture and Landscape, Yunnan Agricultural University, Kunming 650201, China
| | - Yue Miao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Shuwen Lei
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Lei Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; College of Science, Yunnan Agricultural University, Kunming 650201, China.
| | - Man Jia
- Fermentation Engineering Research Center for Yunnan Pu-erh Tea, Kunming 650217, China
| | - Shukang Pan
- Fermentation Engineering Research Center for Yunnan Pu-erh Tea, Kunming 650217, China.
| | - Jiashun Gong
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Agro-Products Processing Research Institute, Yunnan Academy of Agricultural Sciences, Kunming 650223, China.
| | - Qiuping Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
2
|
Azari H, George M, Albracht-Schulte K. Gut Microbiota-microRNA Interactions and Obesity Pathophysiology: A Systematic Review of Integrated Studies. Int J Mol Sci 2024; 25:12836. [PMID: 39684547 DOI: 10.3390/ijms252312836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is the fifth leading cause of death globally and its comorbidities put a high burden on societies and cause disability. In this review, we aim to summarize the interactions and crosstalk between gut microbiota and micro-RNA (miRNA) in obesity. We searched for the relevant literature through PubMed, Web of Science, Scopus, and Science Direct. The study design is registered in the international prospective register of systematic reviews (Prospero). According to the inclusion criteria, eight studies were eligible for assessment (two studies including human subjects and six studies including animal subjects). We report that the interactions of miRNA and gut microbiota in the context of obesity are diverse and in some cases tissue specific. However, the interactions mediate obesity-associated pathways including the inflammatory response, oxidative stress, insulin signaling, gut permeability, and lipogenesis. To mention the most meaningful results, the expression of adipose tissue miRNA-378a-3p/5p was associated with Bifidobacterium and Akkermansia abundance, the expression of hepatic miRNA-34a was related to the Firmicutes phylum, and the expression of miRNA-122-5p and miRNA-375 was associated with the Bacteroides genus. miRNA-microbiota-associated pathological pathways seem to provide an intricate, but promising field for future research directed toward the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Hushyar Azari
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Megan George
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Kembra Albracht-Schulte
- Department of Kinesiology and Sport Management and Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
3
|
Park S, Cha HN, Shin MG, Park S, Kim Y, Kim MS, Shin KH, Thoudam T, Lee EJ, Wolfe RR, Dan J, Koh JH, Kim IY, Choi I, Lee IK, Sung HK, Park SY. Inhibitory Regulation of FOXO1 in PPARδ Expression Drives Mitochondrial Dysfunction and Insulin Resistance. Diabetes 2024; 73:1084-1098. [PMID: 38656552 DOI: 10.2337/db23-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
Forkhead box O1 (FOXO1) regulates muscle growth, but the metabolic role of FOXO1 in skeletal muscle and its mechanisms remain unclear. To explore the metabolic role of FOXO1 in skeletal muscle, we generated skeletal muscle-specific Foxo1 inducible knockout (mFOXO1 iKO) mice and fed them a high-fat diet to induce obesity. We measured insulin sensitivity, fatty acid oxidation, mitochondrial function, and exercise capacity in obese mFOXO1 iKO mice and assessed the correlation between FOXO1 and mitochondria-related protein in the skeletal muscle of patients with diabetes. Obese mFOXO1 iKO mice exhibited improved mitochondrial respiratory capacity, which was followed by attenuated insulin resistance, enhanced fatty acid oxidation, and improved skeletal muscle exercise capacity. Transcriptional inhibition of FOXO1 in peroxisome proliferator-activated receptor δ (PPARδ) expression was confirmed in skeletal muscle, and deletion of PPARδ abolished the beneficial effects of FOXO1 deficiency. FOXO1 protein levels were higher in the skeletal muscle of patients with diabetes and negatively correlated with PPARδ and electron transport chain protein levels. These findings highlight FOXO1 as a new repressor in PPARδ gene expression in skeletal muscle and suggest that FOXO1 links insulin resistance and mitochondrial dysfunction in skeletal muscle via PPARδ. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Min-Gyeong Shin
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Min-Seob Kim
- Department of Fundamental Environment Research, Environmental Measurement and Analysis Center, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Kyung-Hoon Shin
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Robert R Wolfe
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Jinmyoung Dan
- Department of Orthopedic Surgery, College of Medicine, CHA University, Gumi, Republic of Korea
| | - Jin-Ho Koh
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hoon-Ki Sung
- The Hospital for Sick Children Research Institute & Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
4
|
Paylar B, Pramanik S, Bezabhe YH, Olsson PE. Temporal sex specific brain gene expression pattern during early rat embryonic development. Front Cell Dev Biol 2024; 12:1343800. [PMID: 38961864 PMCID: PMC11219815 DOI: 10.3389/fcell.2024.1343800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Background: The classical concept of brain sex differentiation suggests that steroid hormones released from the gonads program male and female brains differently. However, several studies indicate that steroid hormones are not the only determinant of brain sex differentiation and that genetic differences could also be involved. Methods: In this study, we have performed RNA sequencing of rat brains at embryonic days 12 (E12), E13, and E14. The aim was to identify differentially expressed genes between male and female rat brains during early development. Results: Analysis of genes expressed with the highest sex differences showed that Xist was highly expressed in females having XX genotype with an increasing expression over time. Analysis of genes expressed with the highest male expression identified three early genes, Sry2, Eif2s3y, and Ddx3y. Discussion: The observed sex-specific expression of genes at early development confirms that the rat brain is sexually dimorphic prior to gonadal action on the brain and identifies Sry2 and Eif2s3y as early genes contributing to male brain development.
Collapse
Affiliation(s)
| | | | | | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
5
|
De Sousa-Coelho AL, Gacias M, O'Neill BT, Relat J, Link W, Haro D, Marrero PF. FOXO1 represses PPARα-Mediated induction of FGF21 gene expression. Biochem Biophys Res Commun 2023; 644:122-129. [PMID: 36640666 DOI: 10.1016/j.bbrc.2023.01.012] [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: 12/26/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Fibroblast growth factor 21 (FGF21) has emerged as a metabolic regulator that exerts potent anti-diabetic and lipid-lowering effects in animal models of obesity and type 2 diabetes, showing a protective role in fatty liver disease and hepatocellular carcinoma progression. Hepatic expression of FGF21 is regulated by PPARα and is induced by fasting. Ablation of FoxO1 in liver has been shown to increase FGF21 expression in hyperglycemia. To better understand the role of FOXO1 in the regulation of FGF21 expression we have modified HepG2 human hepatoma cells to overexpress FoxO1 and PPARα. Here we show that FoxO1 represses PPARα-mediated FGF21 induction, and that the repression acts on the FGF21 gene promoter without affecting other PPARα target genes. Additionally, we demonstrate that FoxO1 physically interacts with PPARα and that FoxO1/3/4 depletion in skeletal muscle contributes to increased Fgf21 tissue levels. Taken together, these data indicate that FOXO1 is a PPARα-interacting protein that antagonizes PPARα activity on the FGF21 promoter. Because other PPARα target genes remained unaffected, these results suggest a highly specific mechanism implicated in FGF21 regulation. We conclude that FGF21 can be specifically modulated by FOXO1 in a PPARα-dependent manner.
Collapse
Affiliation(s)
- Ana Luísa De Sousa-Coelho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, Edifício 2, 8005-139, Faro, Portugal; Algarve Biomedical Center (ABC), Campus de Gambelas, Edifício 2, 8005-139, Faro, Portugal; Escola Superior de Saúde, Universidade do Algarve, Campus de Gambelas, Edifício 1, 8005-139, Faro, Portugal.
| | - Mar Gacias
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain
| | - Brian T O'Neill
- Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, 52242, Iowa, USA
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - Diego Haro
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain
| | - Pedro F Marrero
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain.
| |
Collapse
|
6
|
Penniman CM, Bhardwaj G, Nowers CJ, Brown CU, Junck TL, Boyer CK, Jena J, Fuqua JD, Lira VA, O'Neill BT. Loss of FoxOs in muscle increases strength and mitochondrial function during aging. J Cachexia Sarcopenia Muscle 2023; 14:243-259. [PMID: 36442857 PMCID: PMC9891940 DOI: 10.1002/jcsm.13124] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Muscle mitochondrial decline is associated with aging-related muscle weakness and insulin resistance. FoxO transcription factors are targets of insulin action and deletion of FoxOs improves mitochondrial function in diabetes. However, disruptions in proteostasis and autophagy are hallmarks of aging and the effect of chronic inhibition of FoxOs in aged muscle is unknown. This study investigated the role of FoxOs in regulating muscle strength and mitochondrial function with age. METHODS We measured muscle strength, cross-sectional area, muscle fibre-type, markers of protein synthesis/degradation, central nuclei, glucose/insulin tolerance, and mitochondrial bioenergetics in 4.5-month (Young) and 22-24-month-old (Aged) muscle-specific FoxO1/3/4 triple KO (TKO) and littermate control (Ctrl) mice. RESULTS Lean mass was increased in Aged TKO compared with both Aged Ctrl and younger groups by 26-33% (P < 0.01). Muscle strength, measured by max force of tibialis anterior (TA) contraction, was 20% lower in Aged Ctrl compared with Young Ctrls (P < 0.01) but was not decreased in Aged TKOs. Increased muscle strength in Young and Aged TKO was associated with 18-48% increased muscle weights compared with Ctrls (P < 0.01). Muscle cross-sectional analysis of TA, soleus, and plantaris revealed increases in fibre size distribution and a 2.5-10-fold increase in central nuclei in Young and Aged TKO mice, without histologic signs of muscle damage. Age-dependent increases in Gadd45a and Ube4a expression as well accumulation of K48 polyubiquitinated proteins were observed in quad and TA but were prevented by FoxO deletion. Young and Aged TKO muscle showed minimal changes in autophagy flux and no accumulation of autophagosomes compared with Ctrl groups. Increased strength in Young and Aged TKO was associated with a 10-20% increase in muscle mitochondrial respiration using glutamate/malate/succinate compared with controls (P < 0.05). OXPHOS subunit expression and complex I activity were decreased 16-34% in Aged Ctrl compared with Young Ctrl but were prevented in Aged TKO. Both Aged Ctrl and Aged TKO showed impaired glucose tolerance by 33% compared to young groups (P < 0.05) indicating improved strength and mitochondrial respiration are not due to improved glycemia. CONCLUSIONS FoxO deletion increases muscle strength even during aging. Deletion of FoxOs maintains muscle strength in part by mild suppression of atrophic pathways, including inhibition of Gadd45a and Ube4a expression, without accumulation of autophagosomes in muscle. Deletion of FoxOs also improved mitochondrial function by maintenance of OXPHOS in both young and aged TKO.
Collapse
Affiliation(s)
- Christie M Penniman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Colette J Nowers
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Chandler U Brown
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Taylor L Junck
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Jayashree Jena
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Jordan D Fuqua
- Fraternal Order of Eagles Diabetes Research Center and Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, USA
| | - Vitor A Lira
- Fraternal Order of Eagles Diabetes Research Center and Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, USA
| | - Brian T O'Neill
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA.,Veterans Affairs Health Care System, 52242, Iowa City, Iowa, USA
| |
Collapse
|
7
|
Paloma Álvarez-Rendón J, Manuel Murillo-Maldonado J, Rafael Riesgo-Escovar J. The insulin signaling pathway a century after its discovery: Sexual dimorphism in insulin signaling. Gen Comp Endocrinol 2023; 330:114146. [PMID: 36270337 DOI: 10.1016/j.ygcen.2022.114146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
Abstract
Since practically a century ago, the insulin pathway was discovered in both vertebrates and invertebrates, implying an evolutionarily ancient origin. After a century of research, it is now clear that the insulin signal transduction pathway is a critical, flexible and pleiotropic pathway, evolving into multiple anabolic functions besides glucose homeostasis. It regulates paramount aspects of organismal well-being like growth, longevity, intermediate metabolism, and reproduction. Part of this diversification has been attained by duplications and divergence of both ligands and receptors riding on a common general signal transduction system. One of the aspects that is strikingly different is its usage in reproduction, particularly in male versus female development and fertility within the same species. This review highlights sexual divergence in metabolism and reproductive tract differences, the occurrence of sexually "exaggerated" traits, and sex size differences that are due to the sexes' differential activity/response to the insulin signaling pathway.
Collapse
Affiliation(s)
- Jéssica Paloma Álvarez-Rendón
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Mexico
| | - Juan Manuel Murillo-Maldonado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Mexico
| | - Juan Rafael Riesgo-Escovar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Mexico.
| |
Collapse
|
8
|
Arioglu-Inan E, Kayki-Mutlu G. Sex Differences in Glucose Homeostasis. Handb Exp Pharmacol 2023; 282:219-239. [PMID: 37439847 DOI: 10.1007/164_2023_664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Sexual dimorphism has been demonstrated to have an effect on various physiological functions. In this regard, researchers have investigated its impact on glucose homeostasis in both preclinical and clinical studies. Sex differences mainly arise from physiological factors such as sex hormones, body fat and muscle distribution, and sex chromosomes. The sexual dimorphism has also been studied in the context of diabetes. Reflecting the prevalence of the disease among the population, studies focusing on the sex difference in type 1 diabetes (T1D) are not common as the ones in type 2 diabetes (T2D). T1D is reported as the only major specific autoimmune disease that exhibits a male predominance. Clinical studies have demonstrated that impaired fasting glucose is more frequent in men whereas women more commonly exhibit impaired glucose tolerance. Understanding the sex difference in glucose homeostasis becomes more attractive when focusing on the findings that highlight sexual dimorphism on the efficacy or adverse effect profile of antidiabetic medications. Thus, in this chapter, we aimed to discuss the impact of sex on the glucose homeostasis both in health and in diabetes.
Collapse
Affiliation(s)
- Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey.
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
9
|
In-depth investigations of the molecular basis underlying sex differences among middle-aged and elderly schizophrenia populations. Psychiatr Genet 2022; 32:178-187. [PMID: 36125368 DOI: 10.1097/ypg.0000000000000322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sex can influence almost all aspects of schizophrenia. However, the molecular mechanisms underlying sex differences in schizophrenia remain poorly understood. In this project, the dataset GSE107638 containing neuronal RNA-seq data and age/sex information of individuals with or without schizophrenia were retrieved. Schizophrenia samples were divided into young male (M-1), young female (F-1), middle-aged and elderly male (M-2) and middle-aged and elderly female (F-2) groups. Next, green/yellow/turquoise modules related to the M-2 trait and turquoise module correlated with the F-2 trait were identified by weighted correlation network analysis (WGCNA) analysis (soft thresholding power: 13; min module size: 200). Crucial genes in the M-2 green, M-2 turquoise and F-2 turquoise modules were identified by WGCNA, gene significance/module membership, and protein-protein interaction (PPI) analysis. Moreover, 2067 and 934 differentially expressed genes (|log2 fold-change| ≥0.58 and P-value < 0.05) in M-2 and F-2 schizophrenia subgroups versus same-age and same-sex counterparts were identified, respectively. Additionally, 82 core genes in the M-2 turquoise module and 4 hub genes in the F-2 turquoise module were differentially expressed in M-2 and F-2 schizophrenia subgroups versus their counterparts, respectively. Among the 82 hub genes, 15 genes were found to be correlated with neuronal development by the Kyoto Encyclopedia of Genes and Genomes enrichment analysis. Also, 2 potential PPI networks related to neuronal development were identified. Taken together, multiple potential hub genes and 2 potential neurobiological networks related to schizophrenia sex differences and disease progression were identified among middle-aged and elderly schizophrenia populations.
Collapse
|
10
|
Sabir U, Irfan HM, Alamgeer, Umer I, Niazi ZR, Asjad HMM. Phytochemicals targeting NAFLD through modulating the dual function of forkhead box O1 (FOXO1) transcription factor signaling pathways. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:741-755. [PMID: 35357518 DOI: 10.1007/s00210-022-02234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
Literature evidence reveals that natural compounds are potential candidates for ameliorating obesity-associated non-alcoholic fatty liver disease (NAFLD) by targeting forkhead box O1 (FOXO1) transcription factor. FOXO1 has a dual and complex role in regulating both increase and decrease in lipid accumulation in hepatocytes and adipose tissues (AT) at different stages of NAFLD. In insulin resistance (IR), it is constitutively expressed, resulting in increased hepatic glucose output and lipid metabolism irregularity. The studies on different phytochemicals indicate that dysregulation of FOXO1 causes disturbance in cellular nutrients homeostasis, and the natural entities have an enduring impact on the mitigation of these abnormalities. The current review communicates and evaluates certain phytochemicals through different search engines, targeting FOXO1 and its downstream cellular pathways to find lead compounds as potential therapeutic agents for treating NAFLD and related metabolic disorders. The findings of this review confirm that polyphenols, flavonoids, alkaloids, terpenoids, and anthocyanins are capable of modulating FOXO1 and associated signaling pathways, and they are potential therapeutic agents for NAFLD and related complications. HIGHLIGHTS: • FOXO1 has the potential to be targeted by novel drugs from natural sources for the treatment of NAFLD and obesity. • FOXO1 regulates cellular autophagy, inflammation, oxidative stress, and lipogenesis through alternative mechanisms. • Phytochemicals treat NAFLD by acting on FOXO1 or SREBP1c and PPARγ transcription factor signaling pathways.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Hafiz Muhammad Irfan
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan.
| | - Alamgeer
- Punjab University College of Pharmacy, University of the Punjab Lahore, Lahore, Pakistan
| | - Ihtisham Umer
- Pharmacy Department, Comsat International University Lahore Campus, Lahore, Pakistan
| | | | | |
Collapse
|
11
|
Tadinada SM, Weatherford ET, Collins GV, Bhardwaj G, Cochran J, Kutschke W, Zimmerman K, Bosko A, O'Neill BT, Weiss RM, Abel ED. Functional resilience of C57BL/6J mouse heart to dietary fat overload. Am J Physiol Heart Circ Physiol 2021; 321:H850-H864. [PMID: 34477461 PMCID: PMC8616610 DOI: 10.1152/ajpheart.00419.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 01/22/2023]
Abstract
Molecular mechanisms underlying cardiac dysfunction and subsequent heart failure in diabetic cardiomyopathy are incompletely understood. Initially we intended to test the role of G protein-coupled receptor kinase 2 (GRK2), a potential mediator of cardiac dysfunction in diabetic cardiomyopathy, but found that control animals on HFD did not develop cardiomyopathy. Cardiac function was preserved in both wild-type and GRK2 knockout animals fed high-fat diet as indicated by preserved left ventricular ejection fraction (LVEF) although heart mass was increased. The absence of cardiac dysfunction led us to rigorously evaluate the utility of diet-induced obesity to model diabetic cardiomyopathy in mice. Using pure C57BL/6J animals and various diets formulated with different sources of fat-lard (32% saturated fat, 68% unsaturated fat) or hydrogenated coconut oil (95% saturated fat), we consistently observed left ventricular hypertrophy, preserved LVEF, and preserved contractility measured by invasive hemodynamics in animals fed high-fat diet. Gene expression patterns that characterize pathological hypertrophy were not induced, but a modest induction of various collagen isoforms and matrix metalloproteinases was observed in heart with high-fat diet feeding. PPARα-target genes that enhance lipid utilization such as Pdk4, CD36, AcadL, and Cpt1b were induced, but mitochondrial energetics was not impaired. These results suggest that although long-term fat feeding in mice induces cardiac hypertrophy and increases cardiac fatty acid metabolism, it may not be sufficient to activate pathological hypertrophic mechanisms that impair cardiac function or induce cardiac fibrosis. Thus, additional factors that are currently not understood may contribute to the cardiac abnormalities previously reported by many groups.NEW & NOTEWORTHY Dietary fat overload (DFO) is widely used to model diabetic cardiomyopathy but the utility of this model is controversial. We comprehensively characterized cardiac contractile and mitochondrial function in C57BL6/J mice fed with lard-based or saturated fat-enriched diets initiated at two ages. Despite cardiac hypertrophy, contractile and mitochondrial function is preserved, and molecular adaptations likely limit lipotoxicity. The resilience of these hearts to DFO underscores the need to develop robust alternative models of diabetic cardiomyopathy.
Collapse
MESH Headings
- Age Factors
- Animals
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Female
- Fibrosis
- G-Protein-Coupled Receptor Kinase 2/genetics
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocardium/enzymology
- Myocardium/pathology
- Obesity/complications
- Stroke Volume
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
- Mice
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Eric T Weatherford
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Greg V Collins
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jesse Cochran
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - William Kutschke
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Kathy Zimmerman
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Alyssa Bosko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Brian T O'Neill
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| | - Robert M Weiss
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Division of Cardiology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
12
|
Bhardwaj G, Penniman CM, Jena J, Suarez Beltran PA, Foster C, Poro K, Junck TL, Hinton AO, Souvenir R, Fuqua JD, Morales PE, Bravo-Sagua R, Sivitz WI, Lira VA, Abel ED, O'Neill BT. Insulin and IGF-1 receptors regulate complex-I dependent mitochondrial bioenergetics and supercomplexes via FoxOs in muscle. J Clin Invest 2021; 131:e146415. [PMID: 34343133 DOI: 10.1172/jci146415] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
Decreased skeletal muscle strength and mitochondrial dysfunction are characteristic of diabetes. Action of insulin and IGF-1 through insulin receptor (IR) and IGF-1 receptor (IGF1R) maintain muscle mass via suppression of FoxOs, but whether FoxO activation coordinates atrophy in concert with mitochondrial dysfunction is unknown. We show that mitochondrial respiration and complex-I activity were decreased in streptozotocin (STZ) diabetic muscle, but these defects were reversed following muscle-specific FoxO1/3/4 triple knockout in STZ-FoxO TKO. In the absence of systemic glucose or lipid abnormalities, muscle-specific IR knockout (M-IR-/-) or combined IR/IGF1R knockout (MIGIRKO) impaired mitochondrial respiration, decreased ATP production, and increased ROS. These mitochondrial abnormalities were not present in muscle-specific IR/IGF1R and FoxO1/3/4 quintuple knockout mice (M-QKO). Acute tamoxifen-inducible deletion of IR/IGF1R also decreased muscle pyruvate respiration, complex-I activity, and supercomplex assembly. Although autophagy was increased when IR/IGF1R were deleted in muscle, mitophagy was not increased. Mechanistically, RNA-seq revealed that complex-I core subunits were decreased in STZ-diabetic and MIGIRKO muscle, and these changes were not present with FoxO knockout in STZ-FoxO TKO and M-QKO. Thus, insulin-deficient diabetes or loss of insulin/IGF-1 action in muscle decreases complex-I driven mitochondrial respiration and supercomplex assembly, in part by FoxO-mediated repression of Complex-I subunit expression.
Collapse
Affiliation(s)
- Gourav Bhardwaj
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Christie M Penniman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Jayashree Jena
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Pablo A Suarez Beltran
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Collin Foster
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Kennedy Poro
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Taylor L Junck
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Antentor O Hinton
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Rhonda Souvenir
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Jordan D Fuqua
- Department of Health and Human Physiology, University of Iowa, Iowa City, United States of America
| | - Pablo E Morales
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - William I Sivitz
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Vitor A Lira
- Department of Health and Human Physiology, University of Iowa, Iowa City, United States of America
| | - E Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Brian T O'Neill
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| |
Collapse
|
13
|
Lu RJ, Taylor S, Contrepois K, Kim M, Bravo JI, Ellenberger M, Sampathkumar NK, Benayoun BA. Multi-omic profiling of primary mouse neutrophils predicts a pattern of sex and age-related functional regulation. NATURE AGING 2021; 1:715-733. [PMID: 34514433 PMCID: PMC8425468 DOI: 10.1038/s43587-021-00086-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Neutrophils are the most abundant human white blood cell and constitute a first line of defense in the innate immune response. Neutrophils are short-lived cells, and thus the impact of organismal aging on neutrophil biology, especially as a function of biological sex, remains poorly understood. Here, we describe a multi-omic resource of mouse primary bone marrow neutrophils from young and old female and male mice, at the transcriptomic, metabolomic and lipidomic levels. We identify widespread regulation of neutrophil 'omics' landscapes with organismal aging and biological sex. In addition, we leverage our resource to predict functional differences, including changes in neutrophil responses to activation signals. To date, this dataset represents the largest multi-omics resource for neutrophils across sex and ages. This resource identifies neutrophil characteristics which could be targeted to improve immune responses as a function of sex and/or age.
Collapse
Affiliation(s)
- Ryan J. Lu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Graduate program in the Biology of Aging, University of Southern California, Los Angeles, CA 90089, USA
| | - Shalina Taylor
- Departments of Pediatrics and of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Minhoo Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Juan I. Bravo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Graduate program in the Biology of Aging, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Nirmal K. Sampathkumar
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Present Address: UK-Dementia Research Institute, Institute of Psychiatry, Psychology and Neuroscience, Basic and Clinical Neuroscience Institute, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, UK
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA 90089
- USC Stem Cell Initiative, Los Angeles, CA 90089, USA
| |
Collapse
|
14
|
Salekeen R, Diaconeasa AG, Billah MM, Islam KMD. Energy Metabolism Focused Analysis of Sexual Dimorphism in Biological Aging and Hypothesized Sex-specificity in Sirtuin Dependency. Mitochondrion 2021; 60:85-100. [PMID: 34332101 DOI: 10.1016/j.mito.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 01/09/2023]
Abstract
The process of biological aging or senescence refers to the gradual loss of homeostasis and subsequent loss of function - leading to higher chances of mortality. Many mechanisms and driving forces have been suggested to facilitate the evolution of a molecular circuit acting as a trade-off between survival and proliferation, resulting in senescence. A major observation on biological aging and longevity in humans and model organisms is the prevalence of significant sexual divergence in the onset, mechanisms and effects of aging associated processes. In the current account, we describe possible mechanisms by which aging, sex and reproduction are evolutionarily intertwined in order to maintain systemic energy homeostasis. We also interrogate existing literature on the sexual dimorphism of genetic, cellular, metabolic, endocrine and epigenetic processes driving cellular and systemic aging. Subsequently, based on available evidence, we propose a hypothetic model of sex-limited decoupling of female longevity from sirtuins, a major family of regulator proteins of the survival-proliferation trade-off. We also provide necessary considerations to be made in order to test the hypothesis and explore the physiological and therapeutic implications of this decoupling event in male and female longevity after reaching reproductive maturity. HYPOTHESIS STATEMENT: Sirtuins provide survival benefits in a sex-nonspecific manner but the dependency on sirtuins in driving metabolic networks after reaching reproductive maturity is evolutionarily decoupled from female longevity.
Collapse
Affiliation(s)
- Rahagir Salekeen
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, Bangladesh.
| | - Amalia Gabriela Diaconeasa
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania.
| | - Md Morsaline Billah
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, Bangladesh.
| | - Kazi Mohammed Didarul Islam
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, Bangladesh.
| |
Collapse
|
15
|
Li X, Wan T, Li Y. Role of FoxO1 in regulating autophagy in type 2 diabetes mellitus (Review). Exp Ther Med 2021; 22:707. [PMID: 34007316 PMCID: PMC8120662 DOI: 10.3892/etm.2021.10139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major chronic disease that is characterized by pancreatic β-cell dysfunction and insulin resistance. Autophagy is a highly conserved intracellular recycling pathway and is involved in regulating intracellular homeostasis. Transcription factor Forkhead box O1 (FoxO1) also regulates fundamental cellular processes, including cell differentiation, metabolism and apoptosis, and proliferation to cellular stress. Increasing evidence suggest that autophagy and FoxO1 are involved in the pathogenesis of T2DM, including β-cell viability, apoptosis, insulin secretion and peripheral insulin resistance. Recent studies have demonstrated that FoxO1 improves insulin resistance by regulating target tissue autophagy. The present review summarizes current literature on the role of autophagy and FoxO1 in T2DM. The participation of FoxO1 in the development and occurrence of T2DM via autophagy is also discussed.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tingting Wan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
16
|
Ortiz-Huidobro RI, Velasco M, Larqué C, Escalona R, Hiriart M. Molecular Insulin Actions Are Sexually Dimorphic in Lipid Metabolism. Front Endocrinol (Lausanne) 2021; 12:690484. [PMID: 34220716 PMCID: PMC8251559 DOI: 10.3389/fendo.2021.690484] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
The increment in energy-dense food and low physical activity has contributed to the current obesity pandemic, which is more prevalent in women than in men. Insulin is an anabolic hormone that regulates the metabolism of lipids, carbohydrates, and proteins in adipose tissue, liver, and skeletal muscle. During obesity, nutrient storage capacity is dysregulated due to a reduced insulin action on its target organs, producing insulin resistance, an early marker of metabolic dysfunction. Insulin resistance in adipose tissue is central in metabolic diseases due to the critical role that this tissue plays in energy homeostasis. We focused on sexual dimorphism on the molecular mechanisms of insulin actions and their relationship with the physiology and pathophysiology of adipose tissue. Until recently, most of the physiological and pharmacological studies were done in males without considering sexual dimorphism, which is relevant. There is ample clinical and epidemiological evidence of its contribution to the establishment and progression of metabolic diseases. Sexual dimorphism is a critical and often overlooked factor that should be considered in design of sex-targeted therapeutic strategies and public health policies to address obesity and diabetes.
Collapse
Affiliation(s)
- Rosa Isela Ortiz-Huidobro
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Myrian Velasco
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Larqué
- Department of Embryology and Genetics, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rene Escalona
- Department of Embryology and Genetics, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marcia Hiriart
- Neurosciences Division, Department of Cognitive Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- *Correspondence: Marcia Hiriart,
| |
Collapse
|
17
|
Li L, Li C, Lv M, Hu Q, Guo L, Xiong D. Correlation between alterations of gut microbiota and miR-122-5p expression in patients with type 2 diabetes mellitus. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1481. [PMID: 33313226 PMCID: PMC7729379 DOI: 10.21037/atm-20-6717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background To investigate the correlation between gut microbiota and circulating microRNAs (miRNAs) in patients with primary diagnosis of type 2 diabetes mellitus (T2DM) and to explore the possible mechanisms of miRNA-gut microbiota crosstalke network in the regulation of the insulin signaling pathway and glucose homeostasis in T2DM. Methods T2DM patients and normal controls were recruited. Fasting plasma and fecal samples were collected from the subjects, and their biochemical indexes including fasting blood glucose (FBG), glycated hemoglobin (HbAlc), cholesterol (TC), total triglycerides (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and insulin were recorded. The variations in intestinal microbiota in the two groups were analyzed using 16S rRNA third-generation sequencing technology, and the differential expression of miRNAs between the groups was screened using miRNA high-throughput sequencing. The correlation and association between specifically changed intestinal microbiota and miRNA expressions were analyzed using a combination of bioinformatics analysis and statistical methods. Finally, 16S functional gene prediction analysis and target gene enrichment pathway analysis were carried out to predict relevant gut microbiota and miRNAs. Results Compared with normal controls, the biochemical indexes of HAlbc, FBG, TG, TC, LDL, HDL, and insulin were significantly different in T2DM patients (P<0.001, P<0.001, P=0.0125, P=0.98, P<0.001 P=0.022, and P=0.0013, respectively). The two groups also showed significantly different intestinal microbiota distribution and miRNA expression characteristics, including in the counts of Bacteriodes. uniformis and Phascolarctobacterium. Faecium (P=0.023, 0.031), which were negatively correlated (P=0.014, FC = -2.36) with the expression levels of serum miR-122-5p (r=−0.68, −0.60, P=0.01, 0.01). Conclusions This study discovered specific gut microbiota and miRNA characteristics in patients with a primary diagnosis of T2DM. A negative correlation between miR-122-5p and the intestinal bacteria Bacteriodes. uniformis and Phascolarctobacterium. Faecium was also revealed, suggesting that the crosstalke between miRNA and gut microbiota may regulate the insulin secretion and signal transduction by controling key genes of glucose metabolism during the development of T2DM.
Collapse
Affiliation(s)
- Lisha Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomin Li
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meijun Lv
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lixuan Guo
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Daqian Xiong
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Skurski J, Penniman CM, Geesala R, Dixit G, Pulipati P, Bhardwaj G, Meyerholz DK, Issuree PD, O'Neill BT, Maretzky T. Loss of iRhom2 accelerates fat gain and insulin resistance in diet-induced obesity despite reduced adipose tissue inflammation. Metabolism 2020; 106:154194. [PMID: 32135161 DOI: 10.1016/j.metabol.2020.154194] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Low-grade inflammation and metabolic dysregulation are common comorbidities of obesity, both of which are associated with alterations in iRhom2-regulated pro-inflammatory cytokine and epidermal growth factor receptor (EGFR) ligand signaling. OBJECTIVE Our objective was to determine the role of iRhom2 in the regulation of low-grade inflammation and metabolic dysregulation in a murine model of diet-induced obesity. METHODS Wild type (WT) and iRhom2-deficient mice were fed normal chow (NC) or a high-fat diet (HFD) starting at 5 weeks of age for up to 33 weeks. Body composition, glucose and insulin tolerance, feeding behavior, and indirect calorimetry were measured at defined time points. Adipose tissue cytokine expression and inflammatory lesions known as crown-like structures (CLS) were analyzed at the end-point of the study. RESULTS iRhom2-deficient mice show accelerated fat gain on a HFD, accompanied by insulin resistance. Indirect calorimetry did not demonstrate changes in energy expenditure or food intake, but locomotor activity was significantly reduced in HFD iRhom2-deficient mice. Interestingly, CLS, macrophage infiltration, and tumor necrosis factor (TNF) production were decreased in adipose tissue from HFD iRhom2-deficient mice, but circulating cytokines were unchanged. In inguinal and perigonadal fat, the EGFR ligand amphiregulin was markedly induced in HFD controls but completely prevented in iRhom2-deficient mice, suggesting a potentially dominant role of EGFR-dependent mechanisms over TNF in the modulation of insulin sensitivity. CONCLUSIONS This study elucidates a novel role for iRhom2 as an immuno-metabolic regulator that affects adipose tissue inflammation independent of insulin resistance.
Collapse
Affiliation(s)
- Joseph Skurski
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Immunology Graduate Program, Iowa City, IA, USA
| | - Christie M Penniman
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - Ramasatyaveni Geesala
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - Garima Dixit
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - Priyanjali Pulipati
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - Gourav Bhardwaj
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - David K Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
| | - Priya D Issuree
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA
| | - Brian T O'Neill
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA.
| | - Thorsten Maretzky
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, IA, USA; Immunology Graduate Program, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|