1
|
Shen D, Wu C, Chen M, Zhou Z, Li H, Tong X, Chen Z, Guo Y. Prognosis prediction and drug guidance of ovarian serous cystadenocarcinoma through mitochondria gene-based model. Cancer Genet 2025; 292-293:1-13. [PMID: 39754905 DOI: 10.1016/j.cancergen.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Mitochondrial dysregulation contributes to the chemoresistance of multiple cancer types. Yet, the functions of mitochondrial dysregulation in Ovarian serous cystadenocarcinoma (OSC) remain largely unknown. AIM We sought to investigate the function of mitochondrial dysregulation in OSC from the bioinformatics perspective. We aimed to establish a model for prognosis prediction and chemosensitivity evaluation of the OSC patients by targeting mitochondrial dysregulation. METHODS Differentially expressed genes (DEGs) were screened from the Cancer Genome Atlas (TCGA)-OV dataset and the mitochondrial-related DEGs were identified from the Human MitoCarta 3.0 database. Prognosis-related mitochondria-related genes (MRGs) were screened to establish the MRGs-based risk score model for prognosis prediction. To validate the risk score model, the risk score model was then evaluated by IHC staining intensity and survival curves from clinical specimens of OSC patients. Migration and proliferation assays were performed to elucidate the role of carcinogenic gene ACSS3 in serous ovarian cancer cell lines. RESULTS Using consensus clustering algorithm, we identified 341 MRGs and two subtypes of OSC patients. Moreover, we established a novel prognostic risk score model by combining the transcription level, intensity and extent scores of MRGs for prognosis prediction purpose. The model was established using 7 MRGs (ACOT13, ACSS3, COA6, HINT2, MRPL14, NDUFC2, and NDUFV2) significantly correlated to the prognosis of OSC. Importantly, by performing the drug sensitivity analysis, we found that the OSC patients in the low-risk group were more sensitive to cisplatin, paclitaxel and docetaxel than those in the high-risk group, while the latter ones were more sensitive to VEGFR inhibitor Axitinib and BRAF inhibitors Vemurafenib and SB590885. In addition, patients in the low-risk group were predicted to have better response in anti-PD-1 immunotherapy than those in the high-risk group. The risk score model was then validated by survival curves of high-risk and low-risk groups determined by IHC staining scores of OSC clinical samples. The carcinogenic effect of ACSS3 in OSC was confirmed through the knockdown of ACSS3 in SKOV3 and HO-8910 cells. CONCLUSION To summarize, we established a novel 7 MRGs - based risk score model that could be utilized for prognosis prediction and chemosensitivity assessment in OSC patients.
Collapse
Affiliation(s)
- Dongsheng Shen
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200120, PR China; Department of Obstetrics and Gynecology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Chenghao Wu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Meiyi Chen
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200120, PR China
| | - Zixuan Zhou
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Burn Institute of PLA, Shanghai, 200433, PR China
| | - Huaifang Li
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200120, PR China
| | - Xiaowen Tong
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200120, PR China
| | - Zhenghu Chen
- College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, PR China.
| | - Yi Guo
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200120, PR China; Department of Obstetrics and Gynecology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China.
| |
Collapse
|
2
|
Zou Y, Ye H, Xu Z, Yang Q, Zhu J, Li T, Cheng Y, Zhu Y, Zhang J, Bo Y, Wang P. Obesity, Sarcopenia, Sarcopenic Obesity, and Hypertension: Mediating Role of Inflammation and Insulin Resistance. J Gerontol A Biol Sci Med Sci 2025; 80:glae284. [PMID: 39918389 DOI: 10.1093/gerona/glae284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND This study aimed to assess the association between obesity, sarcopenia, and sarcopenic obesity with hypertension and to explore the potential mediation of inflammation indicators and insulin resistance. METHODS Data from the UK Biobank, a large-scale prospective cohort, were utilized. Obesity was defined using percentage of fat mass, whereas sarcopenia was defined as low muscle mass and low muscle strength. The primary outcome assessed was new-onset hypertension within a 5-year follow-up period. The association analysis was examined using a Cox regression model. RESULTS A total of 183 091 participants were enrolled in this study. During 5 years of follow-up, 3 812 (2.08%) developed hypertension. In the fully adjusted model, compared to men without these conditions, those with obesity, sarcopenia, and sarcopenic obesity had 2.32 times (95% confidence interval [95% CI], 2.12-2.55), 3.10 times (95% CI, 2.35-4.08), and 3.66 times (95% CI, 2.98-4.50) higher risks of developing hypertension, respectively. Women with obesity, sarcopenia, and sarcopenic obesity had 2.27 times (95% CI, 2.03-2.54), 2.93 times (95% CI, 1.95-4.39), and 4.04 times (95% CI, 3.32-4.91) higher risks of hypertension, respectively. Significant mediating effects of C-reactive protein, neutrophils, white blood cells, triglyceride-glucose index, and triglyceride-to-high-density lipoprotein cholesterol ratio were found, with mediations ranging from 6% to 13% for men and 2% to 21% for women in the association between sarcopenic obesity and hypertension. CONCLUSIONS Obesity, sarcopenia, and sarcopenic obesity significantly increased the risk of hypertension. Inflammation and insulin resistance appeared to mediate the association between sarcopenic obesity and hypertension.
Collapse
Affiliation(s)
- Yuanlin Zou
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Ziqing Xu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Qian Yang
- Prenatal Diagnosis Center, The Third Affiliated Hospital of Zhengzhou University/Maternal and Child Health Hospital of Henan Province, Zhengzhou 450052, Henan Province, China
| | - Jicun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yifan Cheng
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yongjian Zhu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Junxi Zhang
- NHC Key Laboratory of Birth Defects Prevention and Henan Key Laboratory of Population Defects Prevention, Zhengzhou 450002, Henan Province, China
| | - Yacong Bo
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- NHC Key Laboratory of Birth Defects Prevention and Henan Key Laboratory of Population Defects Prevention, Zhengzhou 450002, Henan Province, China
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
3
|
Shannon CE, Bakewell T, Fourcaudot MJ, Ayala I, Smelter AA, Hinostroza EA, Romero G, Asmis M, Freitas Lima LC, Wallace M, Norton L. The mitochondrial pyruvate carrier regulates adipose glucose partitioning in female mice. Mol Metab 2024; 88:102005. [PMID: 39137831 PMCID: PMC11382204 DOI: 10.1016/j.molmet.2024.102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
OBJECTIVE The mitochondrial pyruvate carrier (MPC) occupies a critical node in intermediary metabolism, prompting interest in its utility as a therapeutic target for the treatment of obesity and cardiometabolic disease. Dysregulated nutrient metabolism in adipose tissue is a prominent feature of obesity pathophysiology, yet the functional role of adipose MPC has not been explored. We investigated whether the MPC shapes the adaptation of adipose tissue to dietary stress in female and male mice. METHODS The impact of pharmacological and genetic disruption of the MPC on mitochondrial pathways of triglyceride assembly (lipogenesis and glyceroneogenesis) was assessed in 3T3L1 adipocytes and murine adipose explants, combined with analyses of adipose MPC expression in metabolically compromised humans. Whole-body and adipose-specific glucose metabolism were subsequently investigated in male and female mice lacking adipocyte MPC1 (Mpc1AD-/-) and fed either standard chow, high-fat western style, or high-sucrose lipid restricted diets for 24 weeks, using a combination of radiolabeled tracers and GC/MS metabolomics. RESULTS Treatment with UK5099 or siMPC1 impaired the synthesis of lipids and glycerol-3-phosphate from pyruvate and blunted triglyceride accumulation in 3T3L1 adipocytes, whilst MPC expression in human adipose tissue was negatively correlated with indices of whole-body and adipose tissue metabolic dysfunction. Mature adipose explants from Mpc1AD-/- mice were intrinsically incapable of incorporating pyruvate into triglycerides. In vivo, MPC deletion restricted the incorporation of circulating glucose into adipose triglycerides, but only in female mice fed a zero fat diet, and this associated with sex-specific reductions in tricarboxylic acid cycle pool sizes and compensatory transcriptional changes in lipogenic and glycerol metabolism pathways. However, whole-body adiposity and metabolic health were preserved in Mpc1AD-/- mice regardless of sex, even under conditions of zero dietary fat. CONCLUSIONS These findings highlight the greater capacity for mitochondrially driven triglyceride assembly in adipose from female versus male mice and expose a reliance upon MPC-gated metabolism for glucose partitioning in female adipose under conditions of dietary lipid restriction.
Collapse
Affiliation(s)
- Christopher E Shannon
- UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland; Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
| | - Terry Bakewell
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Marcel J Fourcaudot
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Iriscilla Ayala
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Annie A Smelter
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Edgar A Hinostroza
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Giovanna Romero
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mara Asmis
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Leandro C Freitas Lima
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Martina Wallace
- UCD Conway Institute, School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Luke Norton
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
4
|
Fu Q, Lv R, Wang S, Wang W, Li Y, Qiu G, Chen X, Sun C. Ndufa8 promotes white fat Browning by improving mitochondrial respiratory chain complex I function to ameliorate obesity by in vitro and in vivo. Cell Signal 2024; 122:111340. [PMID: 39127135 DOI: 10.1016/j.cellsig.2024.111340] [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: 03/26/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Obesity and its complications have become a global health problem that needs to be addressed urgently. White adipose tissue (WAT) browning contributes to consuming excess energy in WAT, which is important for improving obesity and maintaining a healthy energy homeostasis. Mitochondria, as the energy metabolism center of cells, are extensively involved in many metabolic processes, including the browning of WAT. NADH: Ubiquinone oxidoreductase subunit A8 (NDUFA8) is a constituent subunit of respiratory chain complex I (CI), which has been found to participate in a wide range of physiological processes by affecting the activity of respiratory CI. However, the regulatory effect of Ndufa8 on the browning of WAT has not been reported. Here, we used β3-adrenergic agonis CL316, 243 to construct WAT browning models in vivo and in vitro to investigate the role and mechanism of Ndufa8 in the regulation of WAT browning. Briefly, Ndufa8 significantly increased CI activity and suppressed mitochondrial ROS levels in vitro, thereby improving mitochondrial function. Ndufa8 also increased the transcriptional levels and protein levels of UCP1 in vitro and in vivo, which promoted WAT browning. Our findings provide a new molecular approach for the research of browning of WAT in animals, as well as a new target for animal metabolism improvement and obesity treatments.
Collapse
Affiliation(s)
- Qinghua Fu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Rui Lv
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Simeng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wentao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yizhou Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Guiping Qiu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xinhao Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
5
|
Al-Diab O, Sünkel C, Blanc E, Catar RA, Ashraf MI, Zhao H, Wang P, Rinschen MM, Fritsche-Guenther R, Grahammer F, Bachmann S, Beule D, Kirwan JA, Rajewsky N, Huber TB, Gürgen D, Kusch A. Sex-specific molecular signature of mouse podocytes in homeostasis and in response to pharmacological challenge with rapamycin. Biol Sex Differ 2024; 15:72. [PMID: 39278930 PMCID: PMC11404044 DOI: 10.1186/s13293-024-00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/30/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Sex differences exist in the prevalence and progression of major glomerular diseases. Podocytes are the essential cell-type in the kidney which maintain the physiological blood-urine barrier, and pathological changes in podocyte homeostasis are critical accelerators of impairment of kidney function. However, sex-specific molecular signatures of podocytes under physiological and stress conditions remain unknown. This work aimed at identifying sexual dimorphic molecular signatures of podocytes under physiological condition and pharmacologically challenged homeostasis with mechanistic target of rapamycin (mTOR) inhibition. mTOR is a crucial regulator involved in a variety of physiological and pathological stress responses in the kidney and inhibition of this pathway may therefore serve as a general stress challenger to get fundamental insights into sex differences in podocytes. METHODS The genomic ROSAmT/mG-NPHS2 Cre mouse model was used which allows obtaining highly pure podocyte fractions for cell-specific molecular analyses, and vehicle or pharmacologic treatment with the mTOR inhibitor rapamycin was performed for 3 weeks. Subsequently, deep RNA sequencing and proteomics were performed of the isolated podocytes to identify intrinsic sex differences. Studies were supplemented with metabolomics from kidney cortex tissues. RESULTS Although kidney function and morphology remained normal in all experimental groups, RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways. Interestingly, rapamycin abolished prominent sex-specific clustering of podocyte gene expression and induced major changes only in male transcriptome. Several sex-biased transcription factors could be identified as possible upstream regulators of these sexually dimorphic responses. Concordant to transcriptomics, metabolomic changes were more prominent in males. Remarkably, high number of previously reported kidney disease genes showed intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. CONCLUSIONS Our results highlight remarkable intrinsic sex-differences and sex-specific response patterns towards pharmacological challenged podocyte homeostasis which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. This work provides rationale and an in-depth database for novel targets to be tested in specific kidney disease models to advance with sex-specific treatment strategies.
Collapse
Affiliation(s)
- Ola Al-Diab
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christin Sünkel
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Rusan Ali Catar
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pinchao Wang
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Markus M Rinschen
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Raphaela Fritsche-Guenther
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jennifer A Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Dennis Gürgen
- Experimental Pharmacology & Oncology Berlin-Buch GmbH, 13125 Berlin-Buch, Germany
| | - Angelika Kusch
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- BIH Biomedical Innovation Academy (BIA), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
6
|
Natarajan N, Florentin J, Johny E, Xiao H, O'Neil SP, Lei L, Shen J, Ohayon L, Johnson AR, Rao K, Li X, Zhao Y, Zhang Y, Tavakoli S, Shiva S, Das J, Dutta P. Aberrant mitochondrial DNA synthesis in macrophages exacerbates inflammation and atherosclerosis. Nat Commun 2024; 15:7337. [PMID: 39187565 PMCID: PMC11347661 DOI: 10.1038/s41467-024-51780-1] [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: 09/22/2023] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
There is a large body of evidence that cellular metabolism governs inflammation, and that inflammation contributes to the progression of atherosclerosis. However, whether mitochondrial DNA synthesis affects macrophage function and atherosclerosis pathology is not fully understood. Here we show, by transcriptomic analyzes of plaque macrophages, spatial single cell transcriptomics of atherosclerotic plaques, and functional experiments, that mitochondrial DNA (mtDNA) synthesis in atherosclerotic plaque macrophages are triggered by vascular cell adhesion molecule 1 (VCAM-1) under inflammatory conditions in both humans and mice. Mechanistically, VCAM-1 activates C/EBPα, which binds to the promoters of key mitochondrial biogenesis genes - Cmpk2 and Pgc1a. Increased CMPK2 and PGC-1α expression triggers mtDNA synthesis, which activates STING-mediated inflammation. Consistently, atherosclerosis and inflammation are less severe in Apoe-/- mice lacking Vcam1 in macrophages. Downregulation of macrophage-specific VCAM-1 in vivo leads to decreased expression of LYZ1 and FCOR, involved in STING signalling. Finally, VCAM-1 expression in human carotid plaque macrophages correlates with necrotic core area, mitochondrial volume, and oxidative damage to DNA. Collectively, our study highlights the importance of macrophage VCAM-1 in inflammation and atherogenesis pathology and proposes a self-acerbating pathway involving increased mtDNA synthesis.
Collapse
Affiliation(s)
- Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ebin Johny
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Hanxi Xiao
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD program in Computational Biology, Pittsburgh, PA, USA
| | - Scott Patrick O'Neil
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Liqun Lei
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jixing Shen
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Lee Ohayon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Aaron R Johnson
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Krithika Rao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Xiaoyun Li
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yanwu Zhao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yingze Zhang
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sina Tavakoli
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
- University of Pittsburgh School of Medicine Department of Pharmacology & Chemical Biology, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Wang F, Huynh PM, An YA. Protocol for Seahorse analysis of ex vivo mouse brown and white adipose tissues. STAR Protoc 2024; 5:103042. [PMID: 38850537 PMCID: PMC11215091 DOI: 10.1016/j.xpro.2024.103042] [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: 02/14/2024] [Revised: 03/12/2024] [Accepted: 04/12/2024] [Indexed: 06/10/2024] Open
Abstract
The mitochondrial stress test is a gold-standard approach for assessing adipose tissue physiological functions and pathological changes. Here, we present a protocol for conducting Seahorse assays using ex vivo mouse brown and white adipose depots. We describe steps for rehydrating the cartridge, preparing freshly harvested fat depots, placing them onto an islet capture plate, and incubating them in a non-CO2 incubator. We then detail procedures for adding mitochondrial stressor solutions and conducting the mitochondrial stress test using the Seahorse XFe24 Analyzer. For complete details on the use and execution of this protocol, please refer to An et al.1.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Shao H, Zhang H, Jia D. The Role of Exerkines in Obesity-Induced Disruption of Mitochondrial Homeostasis in Thermogenic Fat. Metabolites 2024; 14:287. [PMID: 38786764 PMCID: PMC11122964 DOI: 10.3390/metabo14050287] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
There is a notable correlation between mitochondrial homeostasis and metabolic disruption. In this review, we report that obesity-induced disruption of mitochondrial homeostasis adversely affects lipid metabolism, adipocyte differentiation, oxidative capacity, inflammation, insulin sensitivity, and thermogenesis in thermogenic fat. Elevating mitochondrial homeostasis in thermogenic fat emerges as a promising avenue for developing treatments for metabolic diseases, including enhanced mitochondrial function, mitophagy, mitochondrial uncoupling, and mitochondrial biogenesis. The exerkines (e.g., myokines, adipokines, batokines) released during exercise have the potential to ameliorate mitochondrial homeostasis, improve glucose and lipid metabolism, and stimulate fat browning and thermogenesis as a defense against obesity-associated metabolic diseases. This comprehensive review focuses on the manifold benefits of exercise-induced exerkines, particularly emphasizing their influence on mitochondrial homeostasis and fat thermogenesis in the context of metabolic disorders associated with obesity.
Collapse
Affiliation(s)
- Hui Shao
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (H.S.); (H.Z.)
- Graduate School of Harbin Sport University, Harbin Sport University, Harbin 150006, China
| | - Huijie Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (H.S.); (H.Z.)
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (H.S.); (H.Z.)
| |
Collapse
|
9
|
Talari NK, Mattam U, Kaminska D, Sotomayor-Rodriguez I, Rahman AP, Péterfy M, Pajukanta P, Pihlajamäki J, Chella Krishnan K. Hepatokine ITIH3 protects against hepatic steatosis by downregulating mitochondrial bioenergetics and de novo lipogenesis. iScience 2024; 27:109709. [PMID: 38689636 PMCID: PMC11059128 DOI: 10.1016/j.isci.2024.109709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/16/2024] [Accepted: 04/06/2024] [Indexed: 05/02/2024] Open
Abstract
Recent studies demonstrate that liver secretory proteins, also known as hepatokines, regulate normal development, obesity, and simple steatosis to non-alcoholic steatohepatitis (NASH) progression. Using a panel of ∼100 diverse inbred strains of mice and a cohort of bariatric surgery patients, we found that one such hepatokine, inter-trypsin inhibitor heavy chain 3 (ITIH3), was progressively lower in severe non-alcoholic fatty liver disease (NAFLD) disease states highlighting an inverse relationship between Itih3/ITIH3 expression and NAFLD severity. Follow-up animal and cell culture models demonstrated that hepatic ITIH3 overexpression lowered liver triglyceride and lipid droplet accumulation, respectively. Conversely, ITIH3 knockdown in mice increased the liver triglyceride in two independent NAFLD models. Mechanistically, ITIH3 reduced mitochondrial respiration and this, in turn, reduced liver triglycerides, via downregulated de novo lipogenesis. This was accompanied by increased STAT1 signaling and Stat3 expression, both of which are known to protect against NAFLD/NASH. Our findings indicate hepatokine ITIH3 as a potential biomarker and/or treatment for NAFLD.
Collapse
Affiliation(s)
- Noble Kumar Talari
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ushodaya Mattam
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Dorota Kaminska
- Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Irene Sotomayor-Rodriguez
- Medical Sciences Baccalaureate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Afra P. Rahman
- Medical Sciences Baccalaureate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Miklós Péterfy
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, USA
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
10
|
Wu Y, Zhang C, Duan S, Li Y, Lu L, Bajpai A, Yang C, Mi J, Tian G, Xu F, Qi D, Xu Z, Chi XD. TEAD1, MYO7A and NDUFC2 are novel functional genes associated with glucose metabolism in BXD recombinant inbred population. Diabetes Obes Metab 2024; 26:1775-1788. [PMID: 38385898 DOI: 10.1111/dom.15491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/23/2024]
Abstract
AIM The liver is an important metabolic organ that governs glucolipid metabolism, and its dysfunction may cause non-alcoholic fatty liver disease, type 2 diabetes mellitus, dyslipidaemia, etc. We aimed to systematic investigate the key factors related to hepatic glucose metabolism, which may be beneficial for understanding the underlying pathogenic mechanisms for obesity and diabetes mellitus. MATERIALS AND METHODS Oral glucose tolerance test (OGTT) phenotypes and liver transcriptomes of BXD mice under chow and high-fat diet conditions were collected from GeneNetwork. QTL mapping was conducted to pinpoint genomic regions associated with glucose homeostasis. Candidate genes were further nominated using a multi-criteria approach and validated to confirm their functional relevance in vitro. RESULTS Our results demonstrated that plasma glucose levels in OGTT were significantly affected by both diet and genetic background, with six genetic regulating loci were mapped on chromosomes 1, 4, and 7. Moreover, TEAD1, MYO7A and NDUFC2 were identified as the candidate genes. Functionally, siRNA-mediated TEAD1, MYO7A and NDUFC2 knockdown significantly decreased the glucose uptake and inhibited the transcription of genes related to insulin and glucose metabolism pathways. CONCLUSIONS Our study contributes novel insights to the understanding of hepatic glucose metabolism, demonstrating the impact of TEAD1, MYO7A and NDUFC2 on mitochondrial function in the liver and their regulatory role in maintaining in glucose homeostasis.
Collapse
Affiliation(s)
- Yingying Wu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, China
| | - Chao Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shaofei Duan
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Yushan Li
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Lu Lu
- The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Akhilesh Bajpai
- The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Donglai Qi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Zhaowei Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Xiao Dong Chi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| |
Collapse
|
11
|
Liu Y, You S, Ding L, Yuan F, Sun Y. Hepatotoxic effects of chronic exposure to environmentally relevant concentrations of Di-(2-ethylhexyl) phthalate (DEHP) on crucian carp: Insights from multi-omics analyses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 923:171447. [PMID: 38447714 DOI: 10.1016/j.scitotenv.2024.171447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is an extensively used phthalate esters (PAEs) that raise growing ecotoxicological concerns due to detrimental effects on living organisms and ecosystems. This study performed hepatotoxic investigations on crucian carp under chronic low-dosage (CLD) exposure to DEHP at environmentally relevant concentrations (20-500 μg/L). The results demonstrated that the CLD exposure induced irreversible damage to the liver tissue. Multi-omics (transcriptomics and metabolomics) analyses revealed the predominant toxicological mechanisms underlying DEHP-induced hepatotoxicity by inhibiting energy production pathways and the up-regulation of the purine metabolism. Disruption of metabolic pathways led to excessive reactive oxygen species (ROS) production and subsequent oxidative stress. The adverse metabolic effects were exacerbated by an interplay between oxidative stress and endoplasmic reticulum stress. This study not only provides new mechanistic insights into the ecotoxicological effects of DEHP under chronic low-dosage exposure, but also suggests a potential strategy for further ecological risk assessment of PAEs.
Collapse
Affiliation(s)
- Yingjie Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Shijie You
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Lu Ding
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Fangying Yuan
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Yanchun Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China.
| |
Collapse
|
12
|
Zhang J, Na X, Li Z, Ji JS, Li G, Yang H, Yang Y, Tan Y, Zhang J, Xi M, Su D, Zeng H, Wu L, Zhao A. Sarcopenic obesity is part of obesity paradox in dementia development: evidence from a population-based cohort study. BMC Med 2024; 22:133. [PMID: 38520024 PMCID: PMC10960494 DOI: 10.1186/s12916-024-03357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Sarcopenic obesity, a clinical and functional condition characterized by the coexistence of obesity and sarcopenia, has not been investigated in relation to dementia risk and its onset. METHODS We included 208,867 participants from UK biobank, who aged 60 to 69 years at baseline. Dementia diagnoses were identified using hospital records and death register data. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox proportional hazards models to evaluate the associations of obesity, sarcopenia, and sarcopenic obesity with dementia risk, stratified by sex. Stratified analyses were performed across dementia-related polygenic risk score (PRS). Restricted mean survival time models were established to estimate the difference and 95%CIs of dementia onset across different status. Additionally, linear regression models were employed to estimate associations of different status with brain imaging parameters. The mediation effects of chronic diseases were also examined. RESULTS Obese women with high PRS had a decreased risk (HR = 0.855 [0.761-0.961]), but obese men with low PRS had an increased risk (HR = 1.223 [1.045-1.431]). Additionally, sarcopenia was associated with elevated dementia risk (HRwomen = 1.323 [1.064-1.644]; HRmen = 2.144 [1.753-2.621]) in those with low PRS. Among those with high PRS, however, the association was only significant in early-life (HRwomen = 1.679 [1.355-2.081]; HRmen = 2.069 [1.656-2.585]). Of note, sarcopenic obesity was associated with higher dementia risk (HRwomen = 1.424 [1.227-1.653]; HRmen = 1.989 [1.702-2.323]), and results remained similar stratified by PRS. Considering dementia onset, obesity was associated with dementia by 1.114 years delayed in women, however, 0.170 years advanced in men. Sarcopenia (women: 0.080 years; men: 0.192 years) and sarcopenic obesity (women: 0.109 years; men: 0.511 years) respectively advanced dementia onset. Obesity, sarcopenia, and sarcopenic obesity were respectively related to alterations in different brain regions. Association between sarcopenic obesity and dementia was mediated by chronic diseases. CONCLUSIONS Sarcopenic obesity and sarcopenia were respectively associated with increased dementia risk and advanced dementia onset to vary degree. The role of obesity in dementia may differ by sex and genetic background.
Collapse
Affiliation(s)
- Junhan Zhang
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Xiaona Na
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Zhihui Li
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - John S Ji
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Guowei Li
- Center for Clinical Epidemiology and Methodology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Haibing Yang
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Yucheng Yang
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Yuefeng Tan
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Jian Zhang
- School of Public Health, Peking University, Beijing, China
| | - Menglu Xi
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Donghan Su
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Huatang Zeng
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Shenzhen Health Development Research and Data Management Center, Shenzhen, Guangdong, China
| | - Liqun Wu
- Shenzhen Health Development Research and Data Management Center, Shenzhen, Guangdong, China
| | - Ai Zhao
- Vanke School of Public Health, Tsinghua University, Beijing, China.
- Institute for Healthy China, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Wu H, Xie J, Peng W, Ji F, Qian J, Shen Q, Hou G. Effects of guanidinoacetic acid supplementation on liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks. Front Vet Sci 2024; 11:1364815. [PMID: 38435369 PMCID: PMC10904544 DOI: 10.3389/fvets.2024.1364815] [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: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Exogenous supplementation of guanidinoacetic acid can mechanistically regulate the energy distribution in muscle cells. This study aimed to investigate the effects of guanidinoacetic acid supplementation on liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks. We randomly divided 480 42 days-old female Jiaji ducks into four groups with six replicates and 20 ducks for each replicate. The control group was fed the basal diet, and the experimental groups were fed the basal diet with 400, 600, and 800 mg/kg (GA400, GA600, and GA800) guanidinoacetic acid, respectively. Compared with the control group, (1) the total cholesterol (p = 0.0262), triglycerides (p = 0.0357), malondialdehyde (p = 0.0452) contents were lower in GA400, GA600 and GA800 in the liver; (2) the total cholesterol (p = 0.0365), triglycerides (p = 0.0459), and malondialdehyde (p = 0.0326) contents in breast muscle were decreased in GA400, GA600 and GA800; (3) the high density lipoprotein (p = 0.0356) and apolipoprotein-A1 (p = 0.0125) contents were increased in GA600 in the liver; (4) the apolipoprotein-A1 contents (p = 0.0489) in breast muscle were higher in GA600 and GA800; (5) the lipoprotein lipase contents (p = 0.0325) in the liver were higher in GA600 and GA800; (6) the malate dehydrogenase contents (p = 0.0269) in breast muscle were lower in GA400, GA600, and GA800; (7) the insulin induced gene 1 (p = 0.0326), fatty acid transport protein 1 (p = 0.0412), and lipoprotein lipase (p = 0.0235) relative expression were higher in GA400, GA600, and GA800 in the liver; (8) the insulin induced gene 1 (p = 0.0269), fatty acid transport protein 1 (p = 0.0234), and lipoprotein lipase (p = 0.0425) relative expression were increased in GA400, GA600, and GA800 in breast muscle. In this study, the optimum dosage of 600 mg/kg guanidinoacetic acid improved the liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks.
Collapse
Affiliation(s)
- Hongzhi Wu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Jiajun Xie
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Weiqi Peng
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Fengjie Ji
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Jinyu Qian
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Qian Shen
- Hainan Xuhuai Technology Co., Ltd., Haikou, China
| | - Guanyu Hou
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| |
Collapse
|
14
|
Hoang MT, Jung SJ, Lee H, Kim HC. Parent-Offspring Associations of Ideal Cardiovascular Health Metrics: Findings From the 2014 to 2021 Korea National Health and Nutrition Examination Survey. J Am Heart Assoc 2024; 13:e030995. [PMID: 38214252 PMCID: PMC10926814 DOI: 10.1161/jaha.123.030995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Studies have reported the strength of cardiovascular health (CVH) metrics in parent-offspring relationships. This study aimed to describe the sex-specific associations between CVH in parents and adult offspring. METHODS AND RESULTS This study was conducted on the Korea National Health and Nutrition Examination Survey data set, which analyzed trios of mother-father-child, with the child's age from 20 to 39 years. To use the nature of sampling design, survey weighting was applied to all our analyses. Ideal CVH was defined as a cluster of at least 5 ideal individual CVH metrics. We examined the association between parents and their adult offspring regarding clustering CVH and individual CVH metrics through odds ratios and 95% CIs using multiple logistic regression with standard errors adjusted for within-family clustering. The study included 1267 married couples comprising 748 sons and 819 daughters. After adjusting for household income and offspring's sex, age, education, and alcohol consumption, an offspring with either parent attaining a nonideal CVH was 3.52 times more likely to have nonideal CVH. Fathers' nonideal CVH was significantly positively associated with the daughters' nonideal CVH. Maternal nonideal CVH was significantly positively associated with the son's nonideal CVH. When analyzing individual CVH metrics, ideal status in fathers or mothers reduced the likelihood of their offspring having a nonideal status. CONCLUSIONS This cross-sectional study showed positive and differential associations of CVH and its components between parents' and offsprings' nonideal status. Our hypothesis-generating results suggest the relevance of using CVH as a composite indicator in family-centered approaches and heart-health interventions.
Collapse
Affiliation(s)
- Manh Thang Hoang
- Department of Public HealthGraduate School, Yonsei UniversitySeoulKorea
- Department of Preventive MedicineHanyang University College of MedicineSeoulKorea
| | - Sun Jae Jung
- Department of Preventive MedicineYonsei University College of MedicineSeoulKorea
| | - Hokyou Lee
- Department of Preventive MedicineYonsei University College of MedicineSeoulKorea
| | - Hyeon Chang Kim
- Department of Preventive MedicineYonsei University College of MedicineSeoulKorea
- Institute for Innovation in Digital Healthcare, Yonsei UniversitySeoulKorea
| |
Collapse
|
15
|
Miranda CS, Silva-Veiga FM, Santana-Oliveira DA, Vasques-Monteiro IML, Daleprane JB, Souza-Mello V. PPARα/γ synergism activates UCP1-dependent and -independent thermogenesis and improves mitochondrial dynamics in the beige adipocytes of high-fat fed mice. Nutrition 2024; 117:112253. [PMID: 37944411 DOI: 10.1016/j.nut.2023.112253] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/11/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the role of peroxisome proliferator-activated receptor (PPAR) activation (single PPARα or PPARγ, and dual PPARα/γ) on UCP1-dependent and -independent thermogenic pathways and mitochondrial metabolism in the subcutaneous white adipose tissue of mice fed a high-fat diet. METHODS Male C57BL/6 mice received either a control diet (10% lipids) or a high-fat diet (HF; 50% lipids) for 12 wk. The HF group was divided to receive the treatments for 4 wk: HFγ (pioglitazone, 10 mg/kg), HFα (WY-14643, 3.5 mg/kg), and HFα/γ (tesaglitazar, 4 mg/kg). RESULTS The HF group was overweight, insulin resistant, and had subcutaneous white adipocyte dysfunction. Treatment with PPARα and PPARα/γ reduced body mass, mitigated insulin resistance, and induced browning with increased UCP1-dependent and -independent thermogenesis activation and improved mitochondrial metabolism to support the beige adipocyte phenotype. CONCLUSION PPARα and dual PPARα/γ activation recruited UCP1+ beige adipocytes and favored UCP1-independent thermogenesis, yielding body mass and insulin sensitivity normalization. Preserved mitochondrial metabolism emerges as a potential target for obesity treatment using PPAR agonists, with possible clinical applications.
Collapse
Affiliation(s)
- Carolline Santos Miranda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Isabela Macedo Lopes Vasques-Monteiro
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics (LEING), Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Mauvais-Jarvis F. Sex differences in energy metabolism: natural selection, mechanisms and consequences. Nat Rev Nephrol 2024; 20:56-69. [PMID: 37923858 DOI: 10.1038/s41581-023-00781-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/06/2023]
Abstract
Metabolic homeostasis operates differently in men and women. This sex asymmetry is the result of evolutionary adaptations that enable women to resist loss of energy stores and protein mass while remaining fertile in times of energy deficit. During starvation or prolonged exercise, women rely on oxidation of lipids, which are a more efficient energy source than carbohydrates, to preserve glucose for neuronal and placental function and spare proteins necessary for organ function. Carbohydrate reliance in men could be an evolutionary adaptation related to defence and hunting, as glucose, unlike lipids, can be used as a fuel for anaerobic high-exertion muscle activity. The larger subcutaneous adipose tissue depots in healthy women than in healthy men provide a mechanism for lipid storage. As female mitochondria have higher functional capacity and greater resistance to oxidative damage than male mitochondria, uniparental inheritance of female mitochondria may reduce the transmission of metabolic disorders. However, in women, starvation resistance and propensity to obesity have evolved in tandem, and the current prevalence of obesity is greater in women than in men. The combination of genetic sex, programming by developmental testosterone in males, and pubertal sex hormones defines sex-specific biological systems in adults that produce phenotypic sex differences in energy homeostasis, metabolic disease and drug responses.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine and Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA.
- Endocrine service, Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
| |
Collapse
|
17
|
Guerrier L, Malpuech-Brugère C, Richard R, Touron J. Mitochondrial Function in Healthy Human White Adipose Tissue: A Narrative Review. Nutrients 2023; 15:4430. [PMID: 37892505 PMCID: PMC10609723 DOI: 10.3390/nu15204430] [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: 09/07/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
As ¾ of the global population either have excess or insufficient fat, it has become increasingly critical to understand the functions and dysfunctions of adipose tissue (AT). AT serves as a key organ in energy metabolism, and recently, attention has been focused on white AT, particularly its mitochondria, as the literature evidence links their functions to adiposity. This narrative review provides an overview of mitochondrial functionality in human white AT. Firstly, it is noteworthy that the two primary AT depots, subcutaneous AT (scAT) and visceral AT (vAT), exhibit differences in mitochondrial density and activity. Notably, vAT tends to have a higher mitochondrial activity compared to scAT. Subsequently, studies have unveiled a negative correlation between mitochondrial activity and body mass index (BMI), indicating that obesity is associated with a lower mitochondrial function. While the impact of exercise on AT mitochondria remains uncertain, dietary interventions have demonstrated varying effects on AT mitochondria. This variability holds promise for the modulation of AT mitochondrial activity. In summary, AT mitochondria exert a significant influence on health outcomes and can be influenced by factors such as obesity and dietary interventions. Understanding the mechanisms underlying these responses can offer potential insights into managing conditions related to AT and overall health.
Collapse
Affiliation(s)
- Lisa Guerrier
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRAe, 63000 Clermont-Ferrand, France; (C.M.-B.); (R.R.); (J.T.)
| | - Corinne Malpuech-Brugère
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRAe, 63000 Clermont-Ferrand, France; (C.M.-B.); (R.R.); (J.T.)
- CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Ruddy Richard
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRAe, 63000 Clermont-Ferrand, France; (C.M.-B.); (R.R.); (J.T.)
- CRNH Auvergne, 63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Julianne Touron
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRAe, 63000 Clermont-Ferrand, France; (C.M.-B.); (R.R.); (J.T.)
- CRNH Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
18
|
Guo H, Yang B, Kiryu S, Wang Q, Yu D, Sun Z, Chen Y, Li X, Wang F, Ba X. Evaluation of the relations between reproduction-related pituitary and ovarian hormones and abdominal fat area-related variables determined with computed tomography in overweight or obese women who have undergone bariatric surgery: a cross-sectional study. Quant Imaging Med Surg 2023; 13:7065-7076. [PMID: 37869350 PMCID: PMC10585523 DOI: 10.21037/qims-22-1283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 08/22/2023] [Indexed: 10/24/2023]
Abstract
Background An understanding of the associations between midregion fat depots and systemic hormone levels will be crucial for developing health-promotion messages aimed at overweight or obese women. However, related research in this area is rare. The present study was performed to identify and quantify fat-related reproduction pituitary and ovarian hormones in overweight or obese women. Methods A total of 250 eligible overweight or obese women scheduled to undergo laparoscopic sleeve gastrectomy (LSG) from a single center were retrospectively included in this study. Computed tomography (CT) images at the level of the umbilicus were selected, and abdominal fat areas were measured and calculated. The reproduction-related pituitary and ovarian hormones were also measured. The correlations among the parameters were examined using Spearman correlation test. Multiple linear regression analysis was performed after log and β-transformation of the hormone levels and fat area-related variables. Results Positive correlations were detected for prolactin (PRL) with total fat area (TFA) [β=0.045; P=0.029; 95% confidence interval (CI): 0.004-0.085] and subcutaneous fat area (SFA) (β=0.066; P=0.023; 95% CI: 0.009-0.123), whereas estradiol showed a negative correlation with visceral fat area (VFA) (β=-0.056, P=0.005; 95% CI: -0.096 to -0.017) and relative VFA (rVFA) (β=-0.068; P=0.001; 95% CI: -0.109 to -0.027) and a positive correlation with SFA (β=0.036; P=0.042; 95% CI: 0.001-0.071). Progesterone (PROG) was negatively correlated with both VFA (β=-0.037; P=0.002; 95% CI: -0.061 to -0.013) and rVFA (β=-0.039; P=0.002; 95% CI: -0.063 to -0.014). The final results revealed that TFA was increased by 3.1% and SFA was increased by 4.7% with a doubling of PRL concentration; VFA was reduced by 2.5% and rVFA was reduced by 2.6% with a doubling of PROG concentration; and VFA was reduced by 3.8%, rVFA was reduced by 4.6%, and SFA was increased by 2.5% with a doubling of estradiol concentration. Conclusions There exist certain associations between some reproduction-related pituitary and ovarian hormones and fat areas. Our findings provide new insights into the associations between midregion fat depots and systemic hormone levels in overweight or obese women.
Collapse
Affiliation(s)
- Hao Guo
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, China
| | - Bo Yang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Radiology, Qingzhou People’s Hospital, Qingzhou, China
| | - Shigeru Kiryu
- Department of Radiology, Narita Hospital, International University of Health and Welfare, Narita City, Chiba, Japan
| | - Qing Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, China
| | - Yang Chen
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University School of Medicine, Yantai, China
- Department of Medical Imaging, Weifang Medical University, Weifang, China
| | - Xin Li
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Radiology, Tengzhou Central People’s Hospital, Tengzhou, China
| | - Fang Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinru Ba
- Department of Radiology, Yantaishan Hospital, Yantai, China
| |
Collapse
|
19
|
Yang M, Abudureyimu M, Wang X, Zhou Y, Zhang Y, Ren J. PHB2 ameliorates Doxorubicin-induced cardiomyopathy through interaction with NDUFV2 and restoration of mitochondrial complex I function. Redox Biol 2023; 65:102812. [PMID: 37451140 PMCID: PMC10366351 DOI: 10.1016/j.redox.2023.102812] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Doxorubicin (DOX) is among the most widely employed antitumor agents, although its clinical applications have been largely hindered by severe cardiotoxicity. Earlier studies described an essential role of mitochondrial injury in the pathogenesis of DOX cardiomyopathy. PHB2 (Prohibitin 2) is perceived as an essential regulator for mitochondrial dynamics and oxidative phosphorylation (OXPHOS) although its involvement in DOX cardiomyopathy remains elusive. METHODS To decipher the possible role of PHB2 in DOX cardiomyopathy, tamoxifen-induced cardiac-specific PHB2 conditional knockout mice were generated and subjected to DOX challenge. Cardiac function and mitochondrial profiles were examined. Screening of downstream mediators of PHB2 was performed using proteomic profiling and bioinformatic analysis, and was further verified using co-immunoprecipitation and pulldown assays. RESULTS Our data revealed significantly downregulated PHB2 expression in DOX-challenged mouse hearts. PHB2CKO mice were more susceptible to DOX cardiotoxicity compared with PHB2flox/flox mice, as evidenced by more pronounced cardiac atrophy, interstitial fibrosis and decrease in left ventricular ejection fraction and fractional shortening. Mechanistically, PHB2 deficiency resulted in the impairment of mitochondrial bioenergetics and oxidative phosphorylation in DOX cardiotoxicity. Proteomic profiling and interactome analyses revealed that PHB2 interacted with NDUFV2 (NADH-ubiquinone oxidoreductase core subunit V2), a key subunit of mitochondrial respiratory Complex I to mediate regulatory property of PHB2 on mitochondrial metabolism. PHB2 governed the expression of NDUFV2 by promoting its stabilization, while PHB2 deficiency significantly downregulated NDUFV2 in DOX-challenged hearts. Cardiac overexpression of PHB2 alleviated mitochondrial defects in DOX cardiomyopathy both in vivo and in vitro. CONCLUSIONS Our study defined a novel role for PHB2 in mitochondrial dynamics and energetic metabolism through interacting with NDUFV2 in DOX-challenged hearts. Forced overexpression of PHB2 may be considered a promising therapeutic approach for patients with DOX cardiomyopathy.
Collapse
Affiliation(s)
- Mingjie Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China
| | - Xiang Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
20
|
Abstract
In this review, we provide a brief synopsis of the connections between adipose tissue and metabolic health and highlight some recent developments in understanding and exploiting adipocyte biology. Adipose tissue plays critical roles in the regulation of systemic glucose and lipid metabolism and secretes bioactive molecules possessing endocrine, paracrine, and autocrine functions. Dysfunctional adipose tissue has a detrimental impact on metabolic health and is intimately involved in key aspects of metabolic diseases such as insulin resistance, lipid overload, inflammation, and organelle stress. Differences in the distribution of fat depots and adipose characteristics relate to divergent degrees of metabolic dysfunction found in metabolically healthy and unhealthy obese individuals. Thermogenic adipocytes increase energy expenditure via mitochondrial uncoupling or adenosine triphosphate-consuming futile substrate cycles, while functioning as a metabolic sink and participating in crosstalk with other metabolic organs. Manipulation of adipose tissue provides a wealth of opportunities to intervene and combat the progression of associated metabolic diseases. We discuss current treatment modalities for obesity including incretin hormone analogs and touch upon emerging strategies with therapeutic potential including exosome-based therapy, pharmacological activation of brown and beige adipocyte thermogenesis, and administration or inhibition of adipocyte-derived factors.
Collapse
Affiliation(s)
- Sung-Min An
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Seung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - John C. Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
21
|
Chen L, Zhou M, Li H, Liu D, Liao P, Zong Y, Zhang C, Zou W, Gao J. Mitochondrial heterogeneity in diseases. Signal Transduct Target Ther 2023; 8:311. [PMID: 37607925 PMCID: PMC10444818 DOI: 10.1038/s41392-023-01546-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 08/24/2023] Open
Abstract
As key organelles involved in cellular metabolism, mitochondria frequently undergo adaptive changes in morphology, components and functions in response to various environmental stresses and cellular demands. Previous studies of mitochondria research have gradually evolved, from focusing on morphological change analysis to systematic multiomics, thereby revealing the mitochondrial variation between cells or within the mitochondrial population within a single cell. The phenomenon of mitochondrial variation features is defined as mitochondrial heterogeneity. Moreover, mitochondrial heterogeneity has been reported to influence a variety of physiological processes, including tissue homeostasis, tissue repair, immunoregulation, and tumor progression. Here, we comprehensively review the mitochondrial heterogeneity in different tissues under pathological states, involving variant features of mitochondrial DNA, RNA, protein and lipid components. Then, the mechanisms that contribute to mitochondrial heterogeneity are also summarized, such as the mutation of the mitochondrial genome and the import of mitochondrial proteins that result in the heterogeneity of mitochondrial DNA and protein components. Additionally, multiple perspectives are investigated to better comprehend the mysteries of mitochondrial heterogeneity between cells. Finally, we summarize the prospective mitochondrial heterogeneity-targeting therapies in terms of alleviating mitochondrial oxidative damage, reducing mitochondrial carbon stress and enhancing mitochondrial biogenesis to relieve various pathological conditions. The possibility of recent technological advances in targeted mitochondrial gene editing is also discussed.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Zhou
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China.
| |
Collapse
|
22
|
Ding M, Xu HY, Zhou WY, Xia YF, Li BY, Shi YJ, Dou X, Yang QQ, Qian SW, Tang Y, Pan DN, Liu Y, Tang QQ. CLCF1 signaling restrains thermogenesis and disrupts metabolic homeostasis by inhibiting mitochondrial biogenesis in brown adipocytes. Proc Natl Acad Sci U S A 2023; 120:e2305717120. [PMID: 37549287 PMCID: PMC10433725 DOI: 10.1073/pnas.2305717120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/07/2023] [Indexed: 08/09/2023] Open
Abstract
Great progress has been made in identifying positive regulators that activate adipocyte thermogenesis, but negative regulatory signaling of thermogenesis remains poorly understood. Here, we found that cardiotrophin-like cytokine factor 1 (CLCF1) signaling led to loss of brown fat identity, which impaired thermogenic capacity. CLCF1 levels decreased during thermogenic stimulation but were considerably increased in obesity. Adipocyte-specific CLCF1 transgenic (CLCF1-ATG) mice showed impaired energy expenditure and severe cold intolerance. Elevated CLCF1 triggered whitening of brown adipose tissue by suppressing mitochondrial biogenesis. Mechanistically, CLCF1 bound and activated ciliary neurotrophic factor receptor (CNTFR) and augmented signal transducer and activator of transcription 3 (STAT3) signaling. STAT3 transcriptionally inhibited both peroxisome proliferator-activated receptor-γ coactivator (PGC) 1α and 1β, which thereafter restrained mitochondrial biogenesis in adipocytes. Inhibition of CNTFR or STAT3 could diminish the inhibitory effects of CLCF1 on mitochondrial biogenesis and thermogenesis. As a result, CLCF1-TG mice were predisposed to develop metabolic dysfunction even without external metabolic stress. Our findings revealed a brake signal on nonshivering thermogenesis and suggested that targeting this pathway could be used to restore brown fat activity and systemic metabolic homeostasis in obesity.
Collapse
Affiliation(s)
- Meng Ding
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Hong-yu Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Wei-yu Zhou
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yi-fan Xia
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Bai-yu Li
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yi-jie Shi
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Xin Dou
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Qi-qi Yang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Shu-wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Dong-ning Pan
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Qi-qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai200032, China
| |
Collapse
|
23
|
Fried SK. Adipose 'neighborhoods' collaborate to maintain metabolic health. Curr Opin Genet Dev 2023; 81:102079. [PMID: 37406429 PMCID: PMC10867982 DOI: 10.1016/j.gde.2023.102079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023]
Abstract
Body fat is stored in anatomically distinct adipose depots that vary in their cell composition and play specialized roles in systemic metabolic homeostasis via secreted products. Their local effects on nearby tissues (e.g. the gut and visceral adipose tissues) are increasingly recognized and this local crosstalk is being elucidated. The major subcutaneous fat depots, abdominal and gluteal-femoral, exert opposite effects on the risk of metabolic disease. The pace of research into developmental, sex, and genetic determinants of human adipose depot growth and function is rapidly accelerating, providing insight into the pathogenesis of metabolic dysfunction in persons with obesity.
Collapse
Affiliation(s)
- Susan K Fried
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1152, New York, NY 10029, USA.
| |
Collapse
|
24
|
Chella Krishnan K, El Hachem EJ, Keller MP, Patel SG, Carroll L, Vegas AD, Gerdes Gyuricza I, Light C, Cao Y, Pan C, Kaczor-Urbanowicz KE, Shravah V, Anum D, Pellegrini M, Lee CF, Seldin MM, Rosenthal NA, Churchill GA, Attie AD, Parker B, James DE, Lusis AJ. Genetic architecture of heart mitochondrial proteome influencing cardiac hypertrophy. eLife 2023; 12:e82619. [PMID: 37276142 PMCID: PMC10241513 DOI: 10.7554/elife.82619] [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: 08/11/2022] [Accepted: 05/18/2023] [Indexed: 06/07/2023] Open
Abstract
Mitochondria play an important role in both normal heart function and disease etiology. We report analysis of common genetic variations contributing to mitochondrial and heart functions using an integrative proteomics approach in a panel of inbred mouse strains called the Hybrid Mouse Diversity Panel (HMDP). We performed a whole heart proteome study in the HMDP (72 strains, n=2-3 mice) and retrieved 848 mitochondrial proteins (quantified in ≥50 strains). High-resolution association mapping on their relative abundance levels revealed three trans-acting genetic loci on chromosomes (chr) 7, 13 and 17 that regulate distinct classes of mitochondrial proteins as well as cardiac hypertrophy. DAVID enrichment analyses of genes regulated by each of the loci revealed that the chr13 locus was highly enriched for complex-I proteins (24 proteins, P=2.2E-61), the chr17 locus for mitochondrial ribonucleoprotein complex (17 proteins, P=3.1E-25) and the chr7 locus for ubiquinone biosynthesis (3 proteins, P=6.9E-05). Follow-up high resolution regional mapping identified NDUFS4, LRPPRC and COQ7 as the candidate genes for chr13, chr17 and chr7 loci, respectively, and both experimental and statistical analyses supported their causal roles. Furthermore, a large cohort of Diversity Outbred mice was used to corroborate Lrpprc gene as a driver of mitochondrial DNA (mtDNA)-encoded gene regulation, and to show that the chr17 locus is specific to heart. Variations in all three loci were associated with heart mass in at least one of two independent heart stress models, namely, isoproterenol-induced heart failure and diet-induced obesity. These findings suggest that common variations in certain mitochondrial proteins can act in trans to influence tissue-specific mitochondrial functions and contribute to heart hypertrophy, elucidating mechanisms that may underlie genetic susceptibility to heart failure in human populations.
Collapse
Affiliation(s)
- Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Elie-Julien El Hachem
- Department of Integrative Biology and Physiology, Field Systems Biology, Sciences Sorbonne UniversitéParisFrance
| | - Mark P Keller
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Sanjeet G Patel
- Department of Surgery/Division of Cardiac Surgery, University of Southern California Keck School of MedicineLos AngelesUnited States
| | - Luke Carroll
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Alexis Diaz Vegas
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | | | - Christine Light
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
| | - Yang Cao
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Division of Oral Biology and Medicine, UCLA School of DentistryLos AngelesUnited States
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Varun Shravah
- Department of Chemistry, University of CaliforniaLos AngelesUnited States
| | - Diana Anum
- Department of Integrative Biology and Physiology, University of CaliforniaLos AngelesUnited States
| | - Matteo Pellegrini
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
- Department of Physiology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Marcus M Seldin
- Center for Epigenetics and MetabolismIrvineUnited States
- Department of Biological Chemistry, University of CaliforniaIrvineUnited States
| | | | | | - Alan D Attie
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
| | - David E James
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, University of CaliforniaLos AngelesUnited States
- Department of Microbiology, Immunology and Molecular Genetics, University of CaliforniaLos AngelesUnited States
| |
Collapse
|
25
|
Sun Y, Zhang J, Hong J, Zhang Z, Lu P, Gao A, Ni M, Zhang Z, Yang H, Shen J, Lu J, Xue W, Lv Q, Bi Y, Zeng YA, Gu W, Ning G, Wang W, Liu R, Wang J. Human RSPO1 Mutation Represses Beige Adipocyte Thermogenesis and Contributes to Diet-Induced Adiposity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207152. [PMID: 36755192 PMCID: PMC10131814 DOI: 10.1002/advs.202207152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/15/2023] [Indexed: 06/18/2023]
Abstract
Recent genetic evidence has linked WNT downstream mutations to fat distribution. However, the roles of WNTs in human obesity remain unclear. Here, the authors screen all Wnt-related paracrine factors in 1994 obese cases and 2161 controls using whole-exome sequencing (WES) and identify that 12 obese patients harbor the same mutations in RSPO1 (p.R219W/Q) predisposing to human obesity. RSPO1 is predominantly expressed in visceral fat, primarily in the fibroblast cluster, and is increased with adiposity. Mice overexpressing human RSPO1 in adipose tissues develop obesity under a high-fat diet (HFD) due to reduced brown/beige fat thermogenesis. In contrast, Rspo1 ablation resists HFD-induced adiposity by increasing thermogenesis. Mechanistically, RSPO1 overexpression or administration significantly inhibits adipocyte mitochondrial respiration and thermogenesis via LGR4-Wnt/β-catenin signaling pathway. Importantly, humanized knockin mice carrying the hotspot mutation (p.R219W) display suppressed thermogenesis and recapitulate the adiposity feature of obese carriers. The mutation disrupts RSPO1's electrostatic interaction with the extracellular matrix, leading to excessive RSPO1 release that activates LGR4-Wnt/β-catenin signaling and attenuates thermogenic capacity in differentiated beige adipocytes. Therefore, these findings identify that gain-of-function mutations and excessive expression of RSPO1, acting as a paracrine Wnt activator, suppress fat thermogenesis and contribute to obesity in humans.
Collapse
Affiliation(s)
- Yingkai Sun
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Juan Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Jie Hong
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Zhongyun Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Peng Lu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Aibo Gao
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Mengshan Ni
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Zhiyin Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Huanjie Yang
- BGI GenomicsBGI‐ShenzhenShenzhen860755P. R. China
| | - Juan Shen
- BGI GenomicsBGI‐ShenzhenShenzhen860755P. R. China
| | - Jieli Lu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Wenzhi Xue
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Qianqian Lv
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Yufang Bi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Yi Arial Zeng
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell ScienceInstitute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031P. R. China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Guang Ning
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Weiqing Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Ruixin Liu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai National Center for Translational MedicineShanghai200025P. R. China
| |
Collapse
|
26
|
Jurrjens AW, Seldin MM, Giles C, Meikle PJ, Drew BG, Calkin AC. The potential of integrating human and mouse discovery platforms to advance our understanding of cardiometabolic diseases. eLife 2023; 12:e86139. [PMID: 37000167 PMCID: PMC10065800 DOI: 10.7554/elife.86139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
Cardiometabolic diseases encompass a range of interrelated conditions that arise from underlying metabolic perturbations precipitated by genetic, environmental, and lifestyle factors. While obesity, dyslipidaemia, smoking, and insulin resistance are major risk factors for cardiometabolic diseases, individuals still present in the absence of such traditional risk factors, making it difficult to determine those at greatest risk of disease. Thus, it is crucial to elucidate the genetic, environmental, and molecular underpinnings to better understand, diagnose, and treat cardiometabolic diseases. Much of this information can be garnered using systems genetics, which takes population-based approaches to investigate how genetic variance contributes to complex traits. Despite the important advances made by human genome-wide association studies (GWAS) in this space, corroboration of these findings has been hampered by limitations including the inability to control environmental influence, limited access to pertinent metabolic tissues, and often, poor classification of diseases or phenotypes. A complementary approach to human GWAS is the utilisation of model systems such as genetically diverse mouse panels to study natural genetic and phenotypic variation in a controlled environment. Here, we review mouse genetic reference panels and the opportunities they provide for the study of cardiometabolic diseases and related traits. We discuss how the post-GWAS era has prompted a shift in focus from discovery of novel genetic variants to understanding gene function. Finally, we highlight key advantages and challenges of integrating complementary genetic and multi-omics data from human and mouse populations to advance biological discovery.
Collapse
Affiliation(s)
- Aaron W Jurrjens
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, United States
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
27
|
García-Llorca A, Kararigas G. Sex-Related Effects of Gut Microbiota in Metabolic Syndrome-Related Diabetic Retinopathy. Microorganisms 2023; 11:microorganisms11020447. [PMID: 36838411 PMCID: PMC9967826 DOI: 10.3390/microorganisms11020447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
The metabolic syndrome (MetS) is a complex disease of metabolic abnormalities, including obesity, insulin resistance, hypertension and dyslipidaemia, and it is associated with an increased risk of cardiovascular disease (CVD). Diabetic retinopathy (DR) is the leading cause of vision loss among working-aged adults around the world and is the most frequent complication in type 2 diabetic (T2D) patients. The gut microbiota are a complex ecosystem made up of more than 100 trillion of microbial cells and their composition and diversity have been identified as potential risk factors for the development of several metabolic disorders, including MetS, T2D, DR and CVD. Biomarkers are used to monitor or analyse biological processes, therapeutic responses, as well as for the early detection of pathogenic disorders. Here, we discuss molecular mechanisms underlying MetS, the effects of biological sex in MetS-related DR and gut microbiota, as well as the latest advances in biomarker research in the field. We conclude that sex may play an important role in gut microbiota influencing MetS-related DR.
Collapse
|
28
|
Rudnicki M, Pislaru A, Rezvan O, Rullman E, Fawzy A, Nwadozi E, Roudier E, Gustafsson T, Haas TL. Transcriptomic profiling reveals sex-specific molecular signatures of adipose endothelial cells under obesogenic conditions. iScience 2022; 26:105811. [PMID: 36624843 PMCID: PMC9823135 DOI: 10.1016/j.isci.2022.105811] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/13/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Female mice display greater adipose angiogenesis and maintain healthier adipose tissue than do males upon high-fat diet feeding. Through transcriptome analysis of endothelial cells (EC) from the white adipose tissue of male and female mice high-fat-fed for 7 weeks, we found that adipose EC exhibited pronouncedly sex-distinct transcriptomes. Genes upregulated in female adipose EC were associated with proliferation, oxidative phosphorylation, and chromatin remodeling contrasting the dominant enrichment for genes related to inflammation and a senescence-associated secretory of male EC. Similar sex-biased phenotypes of adipose EC were detectable in a dataset of aged EC. The highly proliferative phenotype of female EC was observed also in culture conditions. In turn, male EC displayed greater inflammatory potential than female EC in culture, based on basal and tumor necrosis factor alpha-stimulated patterns of gene expression. Our study provides insights into molecular programs that distinguish male and female EC responses to pathophysiological conditions.
Collapse
Affiliation(s)
- Martina Rudnicki
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada,Corresponding author
| | | | - Omid Rezvan
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Eric Rullman
- Department Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Department Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Aly Fawzy
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Emmanuel Nwadozi
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Emilie Roudier
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada
| | - Thomas Gustafsson
- Department Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Department Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Tara L. Haas
- School of Kinesiology and Health Science & Muscle Health Research Centre, York University, Toronto, Canada,Department of Biology, York University, Toronto, Canada,Corresponding author
| |
Collapse
|
29
|
Moore TM, Cheng L, Wolf DM, Ngo J, Segawa M, Zhu X, Strumwasser AR, Cao Y, Clifford BL, Ma A, Scumpia P, Shirihai OS, Vallim TQDA, Laakso M, Lusis AJ, Hevener AL, Zhou Z. Parkin regulates adiposity by coordinating mitophagy with mitochondrial biogenesis in white adipocytes. Nat Commun 2022; 13:6661. [PMID: 36333379 PMCID: PMC9636263 DOI: 10.1038/s41467-022-34468-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Parkin, an E3 ubiquitin ligase, plays an essential role in mitochondrial quality control. However, the mechanisms by which Parkin connects mitochondrial homeostasis with cellular metabolism in adipose tissue remain unclear. Here, we demonstrate that Park2 gene (encodes Parkin) deletion specifically from adipose tissue protects mice against high-fat diet and aging-induced obesity. Despite a mild reduction in mitophagy, mitochondrial DNA content and mitochondrial function are increased in Park2 deficient white adipocytes. Moreover, Park2 gene deletion elevates mitochondrial biogenesis by increasing Pgc1α protein stability through mitochondrial superoxide-activated NAD(P)H quinone dehydrogenase 1 (Nqo1). Both in vitro and in vivo studies show that Nqo1 overexpression elevates Pgc1α protein level and mitochondrial DNA content and enhances mitochondrial activity in mouse and human adipocytes. Taken together, our findings indicate that Parkin regulates mitochondrial homeostasis by balancing mitophagy and Pgc1α-mediated mitochondrial biogenesis in white adipocytes, suggesting a potential therapeutic target in adipocytes to combat obesity and obesity-associated disorders.
Collapse
Affiliation(s)
- Timothy M Moore
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lijing Cheng
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dane M Wolf
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jennifer Ngo
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Mayuko Segawa
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Xiaopeng Zhu
- Division of Pediatric Endocrinology, Department of Pediatrics UCLA Children's Discovery and Innovation Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Alexander R Strumwasser
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Yang Cao
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Bethan L Clifford
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alice Ma
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Philip Scumpia
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Orian S Shirihai
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Thomas Q de Aguiar Vallim
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Los Angeles, CA, USA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
30
|
Chen Y, He LX, Chen JL, Xu X, Wang JJ, Zhan XH, Jiao JW, Dong G, Li EM, Xu LY. L2Δ13, a splicing isoform of lysyl oxidase-like 2, causes adipose tissue loss via the gut microbiota and lipid metabolism. iScience 2022; 25:104894. [PMID: 36060061 PMCID: PMC9436769 DOI: 10.1016/j.isci.2022.104894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/16/2022] [Accepted: 08/03/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity is primarily characterized by the dysregulation of lipid metabolism and gut microbiota. Here, we found that the body weight of transgenic mice overexpressing L2Δ13, a selectively spliced isoform of lysyl oxidase-like 2 (LOXL2), was lower than that of wild-type (WT) mice. Numerous microbiotas were significantly changed and most microbial metabolites were abnormal in L2Δ13 mice. Lipid metabolites in feces were negatively correlated with those in plasma, suggesting that L2Δ13 may affect lipid uptake, and potentially, adipose tissue homeostasis. This was supported by the weight loss and decreased area of adipose tissue in L2Δ13 mice. Adipogenic differentiation of primary stromal vascular fraction cells showed that the lipid droplets of L2Δ13 cells were significantly smaller than those of WT cells. Adipocyte differentiation-associated genes were also downregulated in adipose tissue from L2Δ13 mice. Thus, L2Δ13 can induce adipose tissue loss in mice by affecting gut microbiota homeostasis and multi-tissue lipid metabolism.
Collapse
Affiliation(s)
- Yang Chen
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Li-Xia He
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Jin-Ling Chen
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Xin Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Juan-Juan Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Xiu-Hui Zhan
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Ji-Wei Jiao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Geng Dong
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Medical Informatics Research Center, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| |
Collapse
|
31
|
Das BK, Wang L, Fujiwara T, Zhou J, Aykin-Burns N, Krager KJ, Lan R, Mackintosh SG, Edmondson R, Jennings ML, Wang X, Feng JQ, Barrientos T, Gogoi J, Kannan A, Gao L, Xing W, Mohan S, Zhao H. Transferrin receptor 1-mediated iron uptake regulates bone mass in mice via osteoclast mitochondria and cytoskeleton. eLife 2022; 11:73539. [PMID: 35758636 PMCID: PMC9352353 DOI: 10.7554/elife.73539] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Increased intracellular iron spurs mitochondrial biogenesis and respiration to satisfy high-energy demand during osteoclast differentiation and bone-resorbing activities. Transferrin receptor 1 (Tfr1) mediates cellular iron uptake through endocytosis of iron-loaded transferrin, and its expression increases during osteoclast differentiation. Nonetheless, the precise functions of Tfr1 and Tfr1-mediated iron uptake in osteoclast biology and skeletal homeostasis remain incompletely understood. To investigate the role of Tfr1 in osteoclast lineage cells in vivo and in vitro, we crossed Tfrc (encoding Tfr1)-floxed mice with Lyz2 (LysM)-Cre and Cathepsin K (Ctsk)-Cre mice to generate Tfrc conditional knockout mice in myeloid osteoclast precursors (Tfr1ΔLysM) or differentiated osteoclasts (Tfr1ΔCtsk), respectively. Skeletal phenotyping by µCT and histology unveiled a significant increase in trabecular bone mass with normal osteoclast number in long bones of 10-week-old young and 6-month-old adult female but not male Tfr1ΔLysM mice. Although high trabecular bone volume in long bones was observed in both male and female Tfr1ΔCtsk mice, this phenotype was more pronounced in female knockout mice. Consistent with this gender-dependent phenomena, estrogen deficiency induced by ovariectomy decreased trabecular bone mass in Tfr1ΔLysM mice. Mechanistically, disruption of Tfr1 expression attenuated mitochondrial metabolism and cytoskeletal organization in mature osteoclasts in vitro by attenuating mitochondrial respiration and activation of the Src-Rac1-WAVE regulatory complex axis, respectively, leading to decreased bone resorption with little impact on osteoclast differentiation. These results indicate that Tfr1-mediated iron uptake is specifically required for osteoclast function and is indispensable for bone remodeling in a gender-dependent manner.
Collapse
Affiliation(s)
- Bhaba K Das
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Lei Wang
- Department of Orthopedics, Anhui Medical University, Hefei, China
| | - Toshifumi Fujiwara
- Department of Orthopedic Surgery, Kyushu University Hospital, Fukuoka, Japan
| | - Jian Zhou
- Department of Orthopedics, Anhui Medical University, HeFei, China
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Kimberly J Krager
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Renny Lan
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Ricky Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Michael L Jennings
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | | | - Jyoti Gogoi
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Aarthi Kannan
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Ling Gao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Haibo Zhao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| |
Collapse
|
32
|
Sandovici I, Fernandez-Twinn DS, Hufnagel A, Constância M, Ozanne SE. Sex differences in the intergenerational inheritance of metabolic traits. Nat Metab 2022; 4:507-523. [PMID: 35637347 DOI: 10.1038/s42255-022-00570-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/05/2022] [Indexed: 02/02/2023]
Abstract
Strong evidence suggests that early-life exposures to suboptimal environmental factors, including those in utero, influence our long-term metabolic health. This has been termed developmental programming. Mounting evidence suggests that the growth and metabolism of male and female fetuses differ. Therefore, sexual dimorphism in response to pre-conception or early-life exposures could contribute to known sex differences in susceptibility to poor metabolic health in adulthood. However, until recently, many studies, especially those in animal models, focused on a single sex, or, often in the case of studies performed during intrauterine development, did not report the sex of the animal at all. In this review, we (a) summarize the evidence that male and females respond differently to a suboptimal pre-conceptional or in utero environment, (b) explore the potential biological mechanisms that underlie these differences and (c) review the consequences of these differences for long-term metabolic health, including that of subsequent generations.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonia Hufnagel
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
33
|
Velez LM, Van C, Moore T, Zhou Z, Johnson C, Hevener AL, Seldin MM. Genetic variation of putative myokine signaling is dominated by biological sex and sex hormones. eLife 2022; 11:e76887. [PMID: 35416774 PMCID: PMC9094747 DOI: 10.7554/elife.76887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022] Open
Abstract
Skeletal muscle plays an integral role in coordinating physiological homeostasis, where signaling to other tissues via myokines allows for coordination of complex processes. Here, we aimed to leverage natural genetic correlation structure of gene expression both within and across tissues to understand how muscle interacts with metabolic tissues. Specifically, we performed a survey of genetic correlations focused on myokine gene regulation, muscle cell composition, cross-tissue signaling, and interactions with genetic sex in humans. While expression levels of a majority of myokines and cell proportions within skeletal muscle showed little relative differences between males and females, nearly all significant cross-tissue enrichments operated in a sex-specific or hormone-dependent fashion; in particular, with estradiol. These sex- and hormone-specific effects were consistent across key metabolic tissues: liver, pancreas, hypothalamus, intestine, heart, visceral, and subcutaneous adipose tissue. To characterize the role of estradiol receptor signaling on myokine expression, we generated male and female mice which lack estrogen receptor α specifically in skeletal muscle (MERKO) and integrated with human data. These analyses highlighted potential mechanisms of sex-dependent myokine signaling conserved between species, such as myostatin enriched for divergent substrate utilization pathways between sexes. Several other putative sex-dependent mechanisms of myokine signaling were uncovered, such as muscle-derived tumor necrosis factor alpha (TNFA) enriched for stronger inflammatory signaling in females compared to males and GPX3 as a male-specific link between glycolytic fiber abundance and hepatic inflammation. Collectively, we provide a population genetics framework for inferring muscle signaling to metabolic tissues in humans. We further highlight sex and estradiol receptor signaling as critical variables when assaying myokine functions and how changes in cell composition are predicted to impact other metabolic organs.
Collapse
Affiliation(s)
- Leandro M Velez
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Cassandra Van
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Timothy Moore
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Zhenqi Zhou
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Casey Johnson
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine at UCLALos AngelesUnited States
- Iris Cantor-UCLA Women’s Health Research Center, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| |
Collapse
|
34
|
Savva C, Helguero LA, González-Granillo M, Melo T, Couto D, Buyandelger B, Gustafsson S, Liu J, Domingues MR, Li X, Korach-André M. Maternal high-fat diet programs white and brown adipose tissue lipidome and transcriptome in offspring in a sex- and tissue-dependent manner in mice. Int J Obes (Lond) 2022; 46:831-842. [PMID: 34997206 PMCID: PMC8960419 DOI: 10.1038/s41366-021-01060-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 12/10/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The prevalence of overweight and obesity among children has drastically increased during the last decades and maternal obesity has been demonstrated as one of the ultimate factors. Nutrition-stimulated transgenerational regulation of key metabolic genes is fundamental to the developmental origins of the metabolic syndrome. Fetal nutrition may differently influence female and male offspring. METHODS Mice dam were fed either a control diet or a high-fat diet (HFD) for 6-week prior mating and continued their respective diet during gestation and lactation. At weaning, female and male offspring were fed the HFD until sacrifice. White (WAT) and brown (BAT) adipose tissues were investigated in vivo by nuclear magnetic resonance at two different timepoints in life (midterm and endterm) and tissues were collected at endterm for lipidomic analysis and RNA sequencing. We explored the sex-dependent metabolic adaptation and gene programming changes by maternal HFD in visceral AT (VAT), subcutaneous AT (SAT) and BAT of offspring. RESULTS We show that the triglyceride profile varies between adipose depots, sexes and maternal diet. In female offspring, maternal HFD remodels the triglycerides profile in SAT and BAT, and increases thermogenesis and cell differentiation in BAT, which may prevent metabolic complication later in life. Male offspring exhibit whitening of BAT and hyperplasia in VAT when born from high-fat mothers, with impaired metabolic profile. Maternal HFD differentially programs gene expression in WAT and BAT of female and male offspring. CONCLUSION Maternal HFD modulates metabolic profile in offspring in a sex-dependent manner. A sex- and maternal diet-dependent gene programming exists in VAT, SAT, and BAT which may be key player in the sexual dimorphism in the metabolic adaptation later in life.
Collapse
Affiliation(s)
- Christina Savva
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luisa A Helguero
- Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Marcela González-Granillo
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tânia Melo
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Daniela Couto
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Byambajav Buyandelger
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sonja Gustafsson
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Jianping Liu
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maria Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Xidan Li
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Marion Korach-André
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden.
- Department of Medicine, Metabolism Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden.
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
35
|
Bao J, Wang L, Hu P, Liu J, Tu J, Wang J, Li J, Ning X. Burden of Metabolic Syndrome Among a Low-Income Population in China: A Population-Based Cross-Sectional Study. Diabetes Metab Syndr Obes 2022; 15:2713-2723. [PMID: 36091482 PMCID: PMC9451033 DOI: 10.2147/dmso.s377490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Metabolic syndrome (MetS) is a chronic and complex disease associated with all-cause mortality, cardiovascular disease, and type 2 diabetes. The present study aimed to evaluate the prevalence of MetS and its risk factors among middle-aged and older adults in low-income, low-education rural areas with a high incidence of stroke. METHODS This cross-sectional study of the general population was performed from April 2019 to June 2019 in rural areas of Tianjin, China. All eligible residents aged ≥ 45 years and without active malignant tumors, hepatic failure, and severe renal disease underwent routine medical examinations, which included a questionnaire, physical examination, and routine blood and biochemical tests. The modified International Diabetes Federation criteria for the Asian population was used to identify patients with MetS. RESULTS A total of 3175 individuals (44.8% men, 55.2% women) were included in the final analysis. The prevalence of MetS was 52.8%, with higher prevalence in women than in men (62.4%and 40.9%, respectively). Of the five MetS components, high blood pressure and abdominal obesity were the two most prevalent in both women and men, accounting for 89.3% and 62.0%, respectively, followed by elevated fasting plasma glucose, low high-density lipoprotein cholesterol, and elevated triglycerides. Multivariate logistic regression analysis revealed the following traits to be risk factors for MetS: female sex, self-reported smoking, self-reported snoring, high body mass index, high waist-to-hip ratio, and high serum urate level. CONCLUSION The prevalence of MetS was quite high in rural areas with a low-income, low-education population. Implementing preventive and therapeutic interventions based on these risk factors is essential to prevent metabolic abnormalities.
Collapse
Affiliation(s)
- Jie Bao
- Department of Rehabilitation Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Lifeng Wang
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
| | - Peng Hu
- Department of Acupuncture Encephalopathy, Binhai New Area Hospital of TCM, Tianjin, 300451, People’s Republic of China
| | - Jie Liu
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
| | - Jun Tu
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
| | - Jinghua Wang
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
| | - Jidong Li
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Department of Neurosurgery, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Jidong Li, Department of Neurosurgery, Tianjin Jizhou People’s Hospital, Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, 18 Nanhuan Road, Jizhou District, Tianjin, 301900, People’s Republic of China, Tel/Fax +86-22- 60733586, Email
| | - Xianjia Ning
- Center of Clinical Epidemiology & Evidence-Based Medicine, Tianjin Jizhou People’s Hospital, Tianjin, 301900, People’s Republic of China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
- Correspondence: Xianjia Ning, Laboratory of Epidemiology, Tianjin Neurological Institute & Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China, Tel +86-22-60817505, Fax +86-22-60817448, Email
| |
Collapse
|
36
|
Ding J, Zhang Y. Relationship between Egg Consumption and Metabolic Syndrome. A Meta-Analysis of Observational Studies. J Nutr Health Aging 2022; 26:373-382. [PMID: 35450994 DOI: 10.1007/s12603-022-1765-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Aims: To explore the association between egg consumption and metabolic syndrome (MetS) in the adult population. METHODS The PubMed, Web of Science and Embase electronic databases were searched up to December 2021 for observational studies on the association between egg consumption and MetS in the adult population. The pooled relative risk (RR) of MetS for the highest versus lowest category of egg consumption, and the standard mean difference (SMD) of egg consumption for MetS versus control subjects were calculated. Egg consumption was assessed by a food-frequency questionnaire (FFQ) and the 24-hour recall method. The criteria for MetS were National Cholesterol Education Program-Adult Treatment Panel III (NCEP ATP III), International Diabetes Federation (IDF), Joint Interim Statement (JIS) and American Heart Association (AHA). RESULTS A total of 19 articles with 20 observational studies (331667 participants) were included in this meta-analysis. The overall multivariable adjusted RR (18 studies included) demonstrated that higher egg consumption was associated with a lower probability of having MetS (RR=0.92, 95%CI: 0.88 to 0.96; P<0.001). Subgroup analysis confirmed these findings in cross-sectional studies (RR=0.91, 95% CI: 0.88 to 0.95; P<0.001), studies that used the NCEP ATP III criteria (RR=0.94, 95% CI: 0.89 to 0.99; P=0.02), Asia (RR=0.92, 95% CI: 0.86 to 0.99; P=0.02), studies with samples sizes >5000 (RR=0.92, 95% CI: 0.86 to 0.96; P<0.001), studies that adjusted body mass index (BMI) (RR=0.90, 95% CI: 0.86 to 0.95; P<0.001) and energy intake (RR=0.89, 95% CI: 0.83 to 0.94; P<0.001) and high-quality studies (RR=0.91, 95% CI: 0.88 to 0.95; P<0.001). Moreover, the overall combined SMD (5 studies included) showed that the level of egg consumption in subjects with MetS was also lower than that in control subjects. (SMD=-0.22, 95% CI: -0.25 to -0.20; P<0.001). Sensitivity analysis demonstrated that this finding only existed in studies with samples sizes >5000 (SMD=-0.21, 95% CI: -0.29 to -0.12; P<0.001) and high-quality studies (SMD=-0.23, 95% CI: -0.26 to -0.20; P<0.001). CONCLUSION Our results suggest that higher egg consumption is associated with a lower probability of having MetS in the adult population. However, due to the limited evidence, more global well-designed prospective cohort studies are still needed.
Collapse
Affiliation(s)
- J Ding
- Dr. Yi Zhang, Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China, 410008;
| | | |
Collapse
|