1
|
Liao Z, Wang Y, Hu C, Gu Q, Peng T, Wu L, Wang Y, Zhu L, Wang Q, Ran L, Xiao X. Adipocyte ZAG improves obesity-triggered insulin resistance by reshaping macrophages populations in adipose tissue. Int Immunopharmacol 2025; 152:114414. [PMID: 40068519 DOI: 10.1016/j.intimp.2025.114414] [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: 12/04/2024] [Revised: 02/14/2025] [Accepted: 03/02/2025] [Indexed: 03/24/2025]
Abstract
Adipose tissues macrophages (ATMs) serve as a critical effector in the mediating occurrence of metabolic inflammation to impact whole-body insulin sensitivity in obesity. Discovering the key adipokines mediating crosstalk of adipocytes-macrophages and understanding the molecular mechanism of ATMs polarization and function have become hot topic issues in the immunometabolism fields. Zinc-α2-glycoprotein (ZAG) as a anti-inflammatory adipokines plays important roles in obesity-related metabolic diseases. We attempt to explore the precise role of adipose ZAG in metabolic inflammation and obesity-associated insulin resistance. Here we showed that Omental ZAG was positively associated with insulin sensitivity and M2 macrophages markers. ZAG-specific ablation in adipocyte aggravated insulin resistance and adipose tissues inflammation as evidenced by enhanced M1 macrophages proportion and inhibited AKT signaling pathway in mice fed with a high-fat diet. Exogenous ZAG inhibits PA-induced M1 macrophage polarization via β3-AR/PKA/STAT3 signaling in RAW264.7 macrophages.These findings suggest that adipocyte ZAG maintain insulin sensitivity via the cross talk with adipose-resident macrophages.
Collapse
Affiliation(s)
- Zhezhen Liao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yadi Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Can Hu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qianqian Gu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting Peng
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liangliang Wu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuanyuan Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liyong Zhu
- The Third Xiangya Hospital Affiliated to Central South University, Department of Gastrointestinal Surgery, Changsha, Hunan 410000, China
| | - Qiyu Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Li Ran
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xinhua Xiao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
2
|
Qin H, Zhong Y, Huang J, Miao Y, Du M, Huang K. TRIM56 Promotes White Adipose Tissue Browning to Attenuate Obesity by Degrading TLE3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414073. [PMID: 39928840 PMCID: PMC11967773 DOI: 10.1002/advs.202414073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/28/2025] [Indexed: 02/12/2025]
Abstract
In mammals, the activation of thermogenic adipocytes, such as beige and brown adipocytes, can significantly increase overall energy expenditure, offering a promising strategy to combat metabolic diseases. Despite its considerable potential, the regulatory mechanisms governing this activation remain largely elusive. This study bridges this gap by elucidating that tripartite motif 56 (TRIM56), an E3 ubiquitin ligase, is upregulated in response to cold stimuli, thereby promoting the recruitment of beige adipocytes. Notably, the overexpression of TRIM56 in adipocytes is shown to help mice maintain a core temperature under cold conditions, as well as confer protection against diet-induced obesity. Mechanistically, TRIM56 facilitates the degradation of the transducin-like enhancer protein 3 (TLE3) protein by promoting its K48-linked ubiquitination, which subsequently triggers the activation of thermogenic genes in subcutaneousl white adipose tissue and improved the metabolic profiles. These findings unveil a novel function for TRIM56 in adipocyte browning, suggesting its potential as a therapeutic target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Haojie Qin
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yi Zhong
- Department of Rheumatology and ImmunologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jinhui Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yanli Miao
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Meng Du
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Kai Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular AgingHuazhong University of Science and TechnologyWuhan430022China
- Hubei Clinical Research Center of Metabolic and Cardiovascular DiseaseHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
3
|
Cahyadi DD, Warita K, Takeda-Okuda N, Tamura JI, Hosaka YZ. Cartilaginous fishes-derived chondroitin sulfates potentially suppress lipid droplet accumulation in the differentiated 3T3-L1 adipocytes. Glycoconj J 2025; 42:77-86. [PMID: 40063299 DOI: 10.1007/s10719-025-10183-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025]
Abstract
In this study, we investigated for cell proliferative and adipogenic differentiation inhibitory activities of chondroitin sulfate (CS) from cartilaginous fish: mako shark (Isurus oxyrinchus, spine part, Ms-CS), blue shark (Prionace glauca, spine part, Bs-CS), sharpspine skate (Okamejei acutispina, head and tail parts, Sp-CS) and stingray (Dasyatis akajei, head part, St-CS) on 3T3-L1 cells. Most of the CSs from cartilaginous fish showed concentration-dependent cell proliferative activity of 3T3-L1 cells within the retrieved concentration range (0-1,000 μg/mL), while under induction of adipocyte differentiation, they inhibited lipid accumulation. In particular, Ms-CS and Sp-CS were highly active in inhibiting lipid accumulation in the cells. The present study revealed that cartilaginous fish-derived CS has inhibitory activity on 3T3-L1 adipocyte differentiation by suppressing lipid droplet accumulation, although the degree of suppression varied depending on the composition of the CS and its origin. In addition, a significant increase in chondroitin sulfate N-acetylgalactosaminyltransferase 2 (Csgalnact2) expression of the Sp-CS group at the concentration of 500 µg/mL was observed. Csgalnact2 expression is associated with chondroitin N-acetylgalactosaminyltransferase-2 (ChGn-2), one of the glycosyltransferases that catalyzes the chain initiation and elongation of the CS backbone in its biosynthesis. Exogenous CS from cartilaginous fishes increased Csgalnact2 expression, although further studies are needed to confirm changes in CS biosynthesis. We observed reduced lipid accumulation in differentiated 3T3-L1 cells. Our findings highlight the role of CS polysaccharides, in inhibiting adipogenesis, even though further investigation is required to understand the underlying mechanism.
Collapse
Affiliation(s)
- Danang Dwi Cahyadi
- Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
- Division of Anatomy Histology and Embryology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, 16680, Indonesia
| | - Katsuhiko Warita
- Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Naoko Takeda-Okuda
- Department of Life and Environmental Agricultural Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Jun-Ichi Tamura
- Department of Life and Environmental Agricultural Sciences, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Yoshinao Z Hosaka
- Department of Bioresource Sciences, Faculty of Agriculture, Kyushu University, Nishi-Ku Fukuoka, 744 Motooka, Fukuoka, 819-0395, Japan.
| |
Collapse
|
4
|
Liu J, Aye Y. Tools to Dissect Lipid Droplet Regulation, Players, and Mechanisms. ACS Chem Biol 2025; 20:539-552. [PMID: 40035358 PMCID: PMC11934092 DOI: 10.1021/acschembio.4c00835] [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: 12/13/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025]
Abstract
Spurred by the authors' own recent discovery of reactive metabolite-regulated nexuses involving lipid droplets (LDs), this perspective discusses the latest knowledge and multifaceted approaches toward deconstructing the function of these dynamic organelles, LD-associated localized signaling networks, and protein players. Despite accumulating knowledge surrounding protein families and pathways of conserved importance for LD homeostasis surveillance and maintenance across taxa, much remains to be understood at the molecular level. In particular, metabolic stress-triggered contextual changes in LD-proteins' localized functions, crosstalk with other organelles, and feedback signaling loops and how these are specifically rewired in disease states remain to be illuminated with spatiotemporal precision. We hope this perspective promotes an increased interest in these essential organelles and innovations of new tools and strategies to better understand context-specific LD regulation critical for organismal health.
Collapse
Affiliation(s)
- Jinmin Liu
- University
of Oxford, Oxford OX1 3TA, United
Kingdom
| | - Yimon Aye
- University
of Oxford, Oxford OX1 3TA, United
Kingdom
| |
Collapse
|
5
|
Yeh YS, Evans TD, Iwase M, Jeong SJ, Zhang X, Liu Z, Park A, Ghasemian A, Dianati B, Javaheri A, Kratky D, Kawarasaki S, Goto T, Zhang H, Dutta P, Schopfer FJ, Straub AC, Cho J, Lodhi IJ, Razani B. Identification of lysosomal lipolysis as an essential noncanonical mediator of adipocyte fasting and cold-induced lipolysis. J Clin Invest 2025; 135:e185340. [PMID: 40091840 PMCID: PMC11910232 DOI: 10.1172/jci185340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/16/2025] [Indexed: 03/19/2025] Open
Abstract
Adipose tissue lipolysis is the process by which triglycerides in lipid stores are hydrolyzed into free fatty acids (FFAs), serving as fuel during fasting or cold-induced thermogenesis. Although cytosolic lipases are considered the predominant mechanism of liberating FFAs, lipolysis also occurs in lysosomes via lysosomal acid lipase (LIPA), albeit with unclear roles in lipid storage and whole-body metabolism. We found that adipocyte LIPA expression increased in adipose tissue of mice when lipolysis was stimulated during fasting, cold exposure, or β-adrenergic agonism. This was functionally important, as inhibition of LIPA genetically or pharmacologically resulted in lower plasma FFAs under lipolytic conditions. Furthermore, adipocyte LIPA deficiency impaired thermogenesis and oxygen consumption and rendered mice susceptible to diet-induced obesity. Importantly, lysosomal lipolysis was independent of adipose triglyceride lipase, the rate-limiting enzyme of cytosolic lipolysis. Our data suggest a significant role for LIPA and lysosomal lipolysis in adipocyte lipid metabolism beyond classical cytosolic lipolysis.
Collapse
Affiliation(s)
- Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mari Iwase
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ziyang Liu
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ali Ghasemian
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Borna Dianati
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ali Javaheri
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Hanrui Zhang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Partha Dutta
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Francisco J. Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, Pennsylvania, USA
| | - Jaehyung Cho
- Division of Hematology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Lai H, Tu Y, Liao C, Zhang S, He L, Li J. Joint assessment of abdominal obesity and non-traditional lipid parameters for primary prevention of cardiometabolic multimorbidity: insights from the China health and retirement longitudinal study 2011-2018. Cardiovasc Diabetol 2025; 24:109. [PMID: 40057762 PMCID: PMC11890515 DOI: 10.1186/s12933-025-02667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Obesity and abnormal lipid metabolism increase the risk of various cardiometabolic diseases, including diabetes, heart disease, and stroke. However, the impact of abdominal obesity (AO) and non-traditional lipid parameters on the risk of cardiometabolic multimorbidity (CMM) remains unclear. This study aims to investigate the separate and combined effects of AO and non-traditional lipid parameters on the incidence risk of CMM. METHODS This study enrolled 7,597 eligible participants from the China health and retirement longitudinal study (CHARLS). Cox proportional hazards models were used to perform adjusted regression analyses and mediation analyses, with Kaplan-Meier analysis used for cumulative hazards. Restricted cubic splines were utilized to evaluate the nonlinear relationship between non-traditional lipid parameters and the risk of CMM among participants with AO. Subgroup analyses were conducted with stratification by age, gender, BMI, smoking status, drinking status, and hypertension to investigate interaction effects across different populations. Additionally, sensitivity analyses were further performed to evaluate the impact of various subgroups on diabetes, heart disease, and stroke. RESULTS During the 7-year follow-up period, a total of 699 participants (9.20%) were newly diagnosed with CMM. Kaplan-Meier curves revealed that the subgroup with both AO and high levels of non-traditional lipid parameters had the highest cumulative hazard for developing CMM. In the fully adjusted model, Cox regression analysis revealed that participants with both high levels of non-traditional lipid parameters and AO exhibited the highest risk of developing CMM. Subgroup and sensitivity analyses further confirmed the robustness of these findings, showing consistent results across different demographic groups and under various analytical conditions. Furthermore, AO was found to significantly mediated the associations between non-traditional lipid parameters and the risk of developing CMM. CONCLUSION The separate and combined effects of AO and non-traditional lipid parameters were significantly associated with the risk of developing CMM. Notably, AO may induce CMM by partially mediating the effects of serum lipids in human metabolism. The findings highlighted the importance of joint evaluation of AO and non-traditional lipid parameters for primary prevention of CMM.
Collapse
Affiliation(s)
- Hurong Lai
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yansong Tu
- Faculty of Science, University of Melbourne, Grattan Street, Parkville, VIC, 3010, Australia
| | - Caifeng Liao
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shan Zhang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ling He
- Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jian Li
- Department of Geriatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
Tuerhongjiang G, Li Y, Meng Z, Gao X, Wei Y, Muhetaer G, Li P, Zhang Y, Zhang J, Wu Y, Liu J. Deoxycholic acid ameliorates obesity and insulin resistance by enhancing lipolysis and thermogenesis. Lipids Health Dis 2025; 24:70. [PMID: 40001126 PMCID: PMC11852518 DOI: 10.1186/s12944-025-02485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Bile acids are essential for energy metabolism. Deoxycholic acid (DCA) in particular is associated with metabolic disorders such as type 2 diabetes mellitus (T2DM) and obesity. However, the direct effects of DCA on metabolism and body composition have yet to be studied in depth. METHODS Targeted metabolomics analysis of human feces was performed. C57BL/6J mice fed a high-fat diet (HFD) were gavaged with DCA, and the effects were measured by metabolic tolerance tests and metabolic cages. Body composition was evaluated by echoMRI. To evaluate the beneficial function of DCA on thermogenesis and lipolysis, histological staining and qPCR were carried out. RESULTS There was negative correlation between fecal DCA levels and serum glucose levels, as well as the Homeostatic Model Assessment for Insulin Resistance (HOMA) index in humans. Our findings confirmed that DCA could ameliorate glucose metabolism and insulin sensitivity in mice fed with HFD. DCA supplementation alleviated HFD-induced obesity and decreased the fat mass significantly by promoting lipolysis. Moreover, DCA significantly enhanced energy expenditure and thermogenesis in brown adipose tissue in mice with obesity induced by HFD. CONCLUSIONS Based on the results of our mouse model, DCA may have applications in alleviating obesity and its related metabolic disorders in humans.
Collapse
Affiliation(s)
- Gulinigaer Tuerhongjiang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yang Li
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Zixuan Meng
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Xiyu Gao
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yuanyuan Wei
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gulinigaer Muhetaer
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peiqi Li
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yi Zhang
- Center for Immunological and Metabolic Diseases (CIMD), MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, XianYang, China
| | - Jiaming Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yue Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China.
| | - Junhui Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
8
|
Malaguarnera M, Cauli O, Cabrera-Pastor A. Obesity and Adipose-Derived Extracellular Vesicles: Implications for Metabolic Regulation and Disease. Biomolecules 2025; 15:231. [PMID: 40001534 PMCID: PMC11853251 DOI: 10.3390/biom15020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity, a global epidemic, is a major risk factor for chronic diseases such as type 2 diabetes, cardiovascular disorders, and metabolic syndrome. Adipose tissue, once viewed as a passive fat storage site, is now recognized as an active endocrine organ involved in metabolic regulation and inflammation. In obesity, adipose tissue dysfunction disrupts metabolic balance, leading to insulin resistance and increased production of adipose-derived extracellular vesicles (AdEVs). These vesicles play a key role in intercellular communication and contribute to metabolic dysregulation, affecting organs such as the heart, liver, and brain. AdEVs carry bioactive molecules, including microRNAs, which influence inflammation, insulin sensitivity, and tissue remodeling. In the cardiovascular system, AdEVs can promote atherosclerosis and vascular dysfunction, while those derived from brown adipose tissue offer cardioprotective effects. In type 2 diabetes, AdEVs exacerbate insulin resistance and contribute to complications such as diabetic cardiomyopathy and cognitive decline. Additionally, AdEVs are implicated in metabolic liver diseases, including fatty liver disease, by transferring inflammatory molecules and lipotoxic microRNAs to hepatocytes. These findings highlight the role of AdEVs in obesity-related metabolic disorders and their promise as therapeutic targets for related diseases.
Collapse
Affiliation(s)
- Michele Malaguarnera
- Psychobiology Department, University of Valencia, 46010 Valencia, Spain;
- Nursing Department, University of Valencia, 46010 Valencia, Spain
| | - Omar Cauli
- Nursing Department, University of Valencia, 46010 Valencia, Spain
- Frailty Research Organized Group (FROG), University of Valencia, 46010 Valencia, Spain
| | - Andrea Cabrera-Pastor
- Pharmacology Department, University of Valencia, 46010 Valencia, Spain;
- Fundación de Investigación del Hospital Clínico Universitario de Valencia (INCLIVA), 46010 Valencia, Spain
| |
Collapse
|
9
|
Chen Y, Liu L, Calhoun R, Cheng L, Merrick D, Steger DJ, Seale P. Transcriptional regulation of adipocyte lipolysis by IRF2BP2. SCIENCE ADVANCES 2025; 11:eads5963. [PMID: 39752494 PMCID: PMC11698119 DOI: 10.1126/sciadv.ads5963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025]
Abstract
Adipocyte lipolysis controls systemic energy levels and metabolic homeostasis. Lipolysis is regulated by posttranslational modifications of key lipolytic enzymes. However, less is known about the transcriptional mechanisms that regulate lipolysis. Here, we identify interferon regulatory factor-2 binding protein 2 (IRF2BP2) as a transcriptional repressor of adipocyte lipolysis. Deletion of IRF2BP2 in human adipocytes increases lipolysis without affecting glucose uptake, whereas IRF2BP2 overexpression decreases lipolysis. RNA sequencing, and chromatin immunoprecipitation sequencing analyses show that IRF2BP2 represses lipolysis-related genes, including LIPE, which encodes hormone sensitive lipase, the rate-limiting enzyme in lipolysis. Adipocyte-selective deletion of Irf2bp2 in mice increases Lipe expression and free fatty acid levels, resulting in adipose tissue inflammation and glucose intolerance. Together, these findings demonstrate that IRF2BP2 restrains adipocyte lipolysis and opens avenues to target lipolysis for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Yang Chen
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Liu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Calhoun
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lan Cheng
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Merrick
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Steger
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Chen Y, Liu L, Calhoun R, Cheng L, Merrick D, Steger DJ, Seale P. Transcriptional regulation of adipocyte lipolysis by IRF2BP2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605689. [PMID: 39211193 PMCID: PMC11360913 DOI: 10.1101/2024.07.31.605689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Adipocyte lipolysis controls systemic energy levels and metabolic homeostasis. Lipolysis is regulated by post-translational modifications of key lipolytic enzymes. However, less is known about the transcriptional mechanisms that regulate lipolysis. Here, we identify the transcriptional factor interferon regulatory factor-2 binding protein 2 (IRF2BP2) as a repressor of adipocyte lipolysis. Deletion of IRF2BP2 in primary human adipocytes increases lipolysis without affecting glucose uptake, whereas IRF2BP2 overexpression decreases lipolysis. RNA-seq and ChIP-seq analyses reveal that IRF2BP2 directly represses several lipolysis-related genes, including LIPE ( HSL , hormone sensitive lipase), which encodes the rate-limiting enzyme in lipolysis. Adipocyte-selective deletion of Irf2bp2 in mice increases Lipe expression and free fatty acid levels, resulting in elevated adipose tissue inflammation and glucose intolerance. Altogether, these findings demonstrate that IRF2BP2 restrains adipocyte lipolysis and opens new avenues to target lipolysis for the treatment of metabolic disease.
Collapse
|
11
|
Liang Y, Kaushal D, Wilson RB. Cellular Senescence and Extracellular Vesicles in the Pathogenesis and Treatment of Obesity-A Narrative Review. Int J Mol Sci 2024; 25:7943. [PMID: 39063184 PMCID: PMC11276987 DOI: 10.3390/ijms25147943] [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: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This narrative review explores the pathophysiology of obesity, cellular senescence, and exosome release. When exposed to excessive nutrients, adipocytes develop mitochondrial dysfunction and generate reactive oxygen species with DNA damage. This triggers adipocyte hypertrophy and hypoxia, inhibition of adiponectin secretion and adipogenesis, increased endoplasmic reticulum stress and maladaptive unfolded protein response, metaflammation, and polarization of macrophages. Such feed-forward cycles are not resolved by antioxidant systems, heat shock response pathways, or DNA repair mechanisms, resulting in transmissible cellular senescence via autocrine, paracrine, and endocrine signaling. Senescence can thus affect preadipocytes, mature adipocytes, tissue macrophages and lymphocytes, hepatocytes, vascular endothelium, pancreatic β cells, myocytes, hypothalamic nuclei, and renal podocytes. The senescence-associated secretory phenotype is closely related to visceral adipose tissue expansion and metaflammation; inhibition of SIRT-1, adiponectin, and autophagy; and increased release of exosomes, exosomal micro-RNAs, pro-inflammatory adipokines, and saturated free fatty acids. The resulting hypernefemia, insulin resistance, and diminished fatty acid β-oxidation lead to lipotoxicity and progressive obesity, metabolic syndrome, and physical and cognitive functional decline. Weight cycling is related to continuing immunosenescence and exposure to palmitate. Cellular senescence, exosome release, and the transmissible senescence-associated secretory phenotype contribute to obesity and metabolic syndrome. Targeted therapies have interrelated and synergistic effects on cellular senescence, obesity, and premature aging.
Collapse
Affiliation(s)
- Yicong Liang
- Bankstown Hospital, University of New South Wales, Sydney, NSW 2560, Australia;
| | - Devesh Kaushal
- Campbelltown Hospital, Western Sydney University, Sydney, NSW 2560, Australia;
| | - Robert Beaumont Wilson
- School of Clinical Medicine, University of New South Wales, High St., Kensington, Sydney, NSW 2052, Australia
| |
Collapse
|
12
|
Liu X, Shen D, Liu L, Peng Y, Lu Q. Diosgenin improves post-myocardial infarction cardiac function via HAND2-induced angiogenesis. Biochem Biophys Res Commun 2024; 712-713:149941. [PMID: 38643718 DOI: 10.1016/j.bbrc.2024.149941] [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: 03/31/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
While diosgenin has been demonstrated effective in various cardiovascular diseases, its specific impact on treating heart attacks remains unclear. Our research revealed that diosgenin significantly improved cardiac function in a myocardial infarction (MI) mouse model, reducing cardiac fibrosis and cell apoptosis while promoting angiogenesis. Mechanistically, diosgenin upregulated the Hand2 expression, promoting the proliferation and migration of endothelial cells under hypoxic conditions. Acting as a transcription factor, HAND2 activated the angiogenesis-related gene Aggf1. Conversely, silencing Hand2 inhibited the diosgenin-induced migration of hypoxic endothelial cells and angiogenesis. In summary, these findings provide new insights into the protective role of diosgenin in MI, validating its effect on angiogenic activity and providing a theoretical basis for clinical treatment strategies.
Collapse
Affiliation(s)
- Xuehua Liu
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, China; Cardiac Department, Sir Runrun Hospital Affiliated to Nanjing Medical University, Nanjing, 211166, China
| | - Dehong Shen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Longfei Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yuzhu Peng
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, China.
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
13
|
Bays HE. Obesity, dyslipidemia, and cardiovascular disease: A joint expert review from the Obesity Medicine Association and the National Lipid Association 2024. OBESITY PILLARS 2024; 10:100108. [PMID: 38706496 PMCID: PMC11066689 DOI: 10.1016/j.obpill.2024.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 05/07/2024]
Abstract
Background This joint expert review by the Obesity Medicine Association (OMA) and National Lipid Association (NLA) provides clinicians an overview of the pathophysiologic and clinical considerations regarding obesity, dyslipidemia, and cardiovascular disease (CVD) risk. Methods This joint expert review is based upon scientific evidence, clinical perspectives of the authors, and peer review by the OMA and NLA leadership. Results Among individuals with obesity, adipose tissue may store over 50% of the total body free cholesterol. Triglycerides may represent up to 99% of lipid species in adipose tissue. The potential for adipose tissue expansion accounts for the greatest weight variance among most individuals, with percent body fat ranging from less than 5% to over 60%. While population studies suggest a modest increase in blood low-density lipoprotein cholesterol (LDL-C) levels with excess adiposity, the adiposopathic dyslipidemia pattern most often described with an increase in adiposity includes elevated triglycerides, reduced high density lipoprotein cholesterol (HDL-C), increased non-HDL-C, elevated apolipoprotein B, increased LDL particle concentration, and increased small, dense LDL particles. Conclusions Obesity increases CVD risk, at least partially due to promotion of an adiposopathic, atherogenic lipid profile. Obesity also worsens other cardiometabolic risk factors. Among patients with obesity, interventions that reduce body weight and improve CVD outcomes are generally associated with improved lipid levels. Given the modest improvement in blood LDL-C with weight reduction in patients with overweight or obesity, early interventions to treat both excess adiposity and elevated atherogenic cholesterol (LDL-C and/or non-HDL-C) levels represent priorities in reducing the risk of CVD.
Collapse
Affiliation(s)
- Harold Edward Bays
- Corresponding author. Louisville Metabolic and Atherosclerosis Research Center, Louisville, KY, 40213, USA.
| |
Collapse
|
14
|
O'Reilly ME, Ho S, Coronel J, Zhu L, Liu W, Xue C, Kim E, Cynn E, Matias CV, Soni RK, Wang C, Ionita-Laza I, Bauer RC, Ross L, Zhang Y, Corvera S, Fried SK, Reilly MP. linc-ADAIN, a human adipose lincRNA, regulates adipogenesis by modulating KLF5 and IL-8 mRNA stability. Cell Rep 2024; 43:114240. [PMID: 38753486 PMCID: PMC11334222 DOI: 10.1016/j.celrep.2024.114240] [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: 07/07/2023] [Revised: 03/01/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Adipose tissue remodeling and dysfunction, characterized by elevated inflammation and insulin resistance, play a central role in obesity-related development of type 2 diabetes (T2D) and cardiovascular diseases. Long intergenic non-coding RNAs (lincRNAs) are important regulators of cellular functions. Here, we describe the functions of linc-ADAIN (adipose anti-inflammatory), an adipose lincRNA that is downregulated in white adipose tissue of obese humans. We demonstrate that linc-ADAIN knockdown (KD) increases KLF5 and interleukin-8 (IL-8) mRNA stability and translation by interacting with IGF2BP2. Upregulation of KLF5 and IL-8, via linc-ADAIN KD, leads to an enhanced adipogenic program and adipose tissue inflammation, mirroring the obese state, in vitro and in vivo. KD of linc-ADAIN in human adipose stromal cell (ASC) hTERT adipocytes implanted into mice increases adipocyte size and macrophage infiltration compared to implanted control adipocytes, mimicking hallmark features of obesity-induced adipose tissue remodeling. linc-ADAIN is an anti-inflammatory lincRNA that limits adipose tissue expansion and lipid storage.
Collapse
Affiliation(s)
- Marcella E O'Reilly
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Sebastian Ho
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Johana Coronel
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Lucie Zhu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Wen Liu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Chenyi Xue
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Eunyoung Kim
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Esther Cynn
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Caio V Matias
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Chen Wang
- Department of Statistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Iuliana Ionita-Laza
- Department of Statistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Robert C Bauer
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Leila Ross
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Muredach P Reilly
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA; Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
15
|
Liu C, Lin Y, Wang Y, Lin S, Zhou J, Tang H, Yi X, Ma Z, Xia T, Jiang B, Tian F, Ju Z, Liu B, Gu X, Yang Z, Wang W. HuR promotes triglyceride synthesis and intestinal fat absorption. Cell Rep 2024; 43:114238. [PMID: 38748875 DOI: 10.1016/j.celrep.2024.114238] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/02/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
Triacylglyceride (TAG) synthesis in the small intestine determines the absorption of dietary fat, but the underlying mechanisms remain to be further studied. Here, we report that the RNA-binding protein HuR (ELAVL1) promotes TAG synthesis in the small intestine. HuR associates with the 3' UTR of Dgat2 mRNA and intron 1 of Mgat2 pre-mRNA. Association of HuR with Dgat2 3' UTR stabilizes Dgat2 mRNA, while association of HuR with intron 1 of Mgat2 pre-mRNA promotes the processing of Mgat2 pre-mRNA. Intestinal epithelium-specific HuR knockout reduces the expression of DGAT2 and MGAT2, thereby reducing the dietary fat absorption through TAG synthesis and mitigating high-fat-diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) and obesity. Our findings highlight a critical role of HuR in promoting dietary fat absorption.
Collapse
Affiliation(s)
- Cihang Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Yunping Lin
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Shuyong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jing Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital and Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan 450003, China
| | - Xia Yi
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing 210093, China
| | - Bin Jiang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Feng Tian
- Department of Laboratory Animal Science, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Zhongzhou Yang
- Medical School, Nanjing University, Nanjing 210093, China.
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China; Center for Healthy Aging, Changzhi Medical College, Changzhi 046000, China; Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranostics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, Liaoning, China.
| |
Collapse
|
16
|
Li X, Su Y, Xu Y, Hu T, Lu X, Sun J, Li W, Zhou J, Ma X, Yang Y, Bao Y. Adipocyte-Specific Hnrnpa1 Knockout Aggravates Obesity-Induced Metabolic Dysfunction via Upregulation of CCL2. Diabetes 2024; 73:713-727. [PMID: 38320300 PMCID: PMC11043064 DOI: 10.2337/db23-0609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/28/2024] [Indexed: 02/08/2024]
Abstract
Heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) is involved in lipid and glucose metabolism via mRNA processing. However, whether and how HNRNPA1 alters adipocyte function in obesity remain obscure. Here, we found that the obese state downregulated HNRNPA1 expression in white adipose tissue (WAT). The depletion of adipocyte HNRNPA1 promoted markedly increased macrophage infiltration and expression of proinflammatory and fibrosis genes in WAT of obese mice, eventually leading to exacerbated insulin sensitivity, glucose tolerance, and hepatic steatosis. Mechanistically, HNRNPA1 interacted with Ccl2 and regulated its mRNA stability. Intraperitoneal injection of CCL2-CCR2 signaling antagonist improved adipose tissue inflammation and systemic glucose homeostasis. Furthermore, HNRNPA1 expression in human WAT was negatively correlated with BMI, fat percentage, and subcutaneous fat area. Among individuals with 1-year metabolic surgery follow-up, HNRNPA1 expression was positively related to percentage of total weight loss. These findings identify adipocyte HNRNPA1 as a link between adipose tissue inflammation and systemic metabolic homeostasis, which might be a promising therapeutic target for obesity-related disorders. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xiaoya Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yingying Su
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yiting Xu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Tingting Hu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xuhong Lu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jingjing Sun
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Wenfei Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
17
|
Bays HE, Kirkpatrick CF, Maki KC, Toth PP, Morgan RT, Tondt J, Christensen SM, Dixon DL, Jacobson TA. Obesity, dyslipidemia, and cardiovascular disease: A joint expert review from the Obesity Medicine Association and the National Lipid Association 2024. J Clin Lipidol 2024; 18:e320-e350. [PMID: 38664184 DOI: 10.1016/j.jacl.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
BACKGROUND This joint expert review by the Obesity Medicine Association (OMA) and National Lipid Association (NLA) provides clinicians an overview of the pathophysiologic and clinical considerations regarding obesity, dyslipidemia, and cardiovascular disease (CVD) risk. METHODS This joint expert review is based upon scientific evidence, clinical perspectives of the authors, and peer review by the OMA and NLA leadership. RESULTS Among individuals with obesity, adipose tissue may store over 50% of the total body free cholesterol. Triglycerides may represent up to 99% of lipid species in adipose tissue. The potential for adipose tissue expansion accounts for the greatest weight variance among most individuals, with percent body fat ranging from less than 5% to over 60%. While population studies suggest a modest increase in blood low-density lipoprotein cholesterol (LDL-C) levels with excess adiposity, the adiposopathic dyslipidemia pattern most often described with an increase in adiposity includes elevated triglycerides, reduced high-density lipoprotein cholesterol (HDL-C), increased non-HDL-C, elevated apolipoprotein B, increased LDL particle concentration, and increased small, dense LDL particles. CONCLUSIONS Obesity increases CVD risk, at least partially due to promotion of an adiposopathic, atherogenic lipid profile. Obesity also worsens other cardiometabolic risk factors. Among patients with obesity, interventions that reduce body weight and improve CVD outcomes are generally associated with improved lipid levels. Given the modest improvement in blood LDL-C with weight reduction in patients with overweight or obesity, early interventions to treat both excess adiposity and elevated atherogenic cholesterol (LDL-C and/or non-HDL-C) levels represent priorities in reducing the risk of CVD.
Collapse
Affiliation(s)
- Harold Edward Bays
- Louisville Metabolic and Atherosclerosis Research Center, Clinical Associate Professor, University of Louisville School of Medicine, 3288 Illinois Avenue, Louisville KY 40213 (Dr Bays).
| | - Carol F Kirkpatrick
- Kasiska Division of Health Sciences, Idaho State University, Pocatello, ID (Dr Kirkpatrick).
| | - Kevin C Maki
- Indiana University School of Public Health, Bloomington, IN (Dr Maki).
| | - Peter P Toth
- CGH Medical Center, Department of Clinical Family and Community Medicine, University of Illinois School of Medicine, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine (Dr Toth).
| | - Ryan T Morgan
- Oklahoma State University Center for Health Sciences, Principal Investigator at Lynn Health Science Institute, 3555 NW 58th St., STE 910-W, Oklahoma City, OK 73112 (Dr Morgan).
| | - Justin Tondt
- Department of Family and Community Medicine, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center (Dr Tondt)
| | | | - Dave L Dixon
- Deptartment of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy 410 N 12th Street, Box 980533, Richmond, VA 23298-0533 (Dr Dixon).
| | - Terry A Jacobson
- Lipid Clinic and Cardiovascular Risk Reduction Program, Emory University Department of Medicine, Atlanta, GA (Dr Jacobson).
| |
Collapse
|
18
|
Huai Y, Wang X, Mao W, Wang X, Zhao Y, Chu X, Huang Q, Ru K, Zhang L, Li Y, Chen Z, Qian A. HuR-positive stress granules: Potential targets for age-related osteoporosis. Aging Cell 2024; 23:e14053. [PMID: 38375951 PMCID: PMC10928564 DOI: 10.1111/acel.14053] [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: 07/17/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 02/21/2024] Open
Abstract
Aging impairs osteoblast function and bone turnover, resulting in age-related bone degeneration. Stress granules (SGs) are membrane-less organelles that assemble in response to stress via the recruitment of RNA-binding proteins (RBPs), and have emerged as a novel mechanism in age-related diseases. Here, we identified HuR as a bone-related RBP that aggregated into SGs and facilitated osteogenesis during aging. HuR-positive SG formation increased during osteoblast differentiation, and HuR overexpression mitigated the reduction in SG formation observed in senescent osteoblasts. Moreover, HuR positively regulated the mRNA stability and expression of its target β-catenin by binding and recruiting β-catenin into SGs. As a potential therapeutic target, HuR activator apigenin (API) enhanced its expression and thus aided osteoblasts differentiation. API treatment increased HuR nuclear export, enhanced the recruitment of β-catenin into HuR-positive SGs, facilitated β-catenin nuclear translocation, and contributed osteogenesis. Our findings highlight the roles of HuR and its SGs in promoting osteogenesis during skeletal aging and lay the groundwork for novel therapeutic strategies against age-related skeletal disorders.
Collapse
Affiliation(s)
- Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Department of OrthopedicsTangdu Hospital, Air Force Military Medical UniversityXi'anChina
| | - Xue Wang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Wenjing Mao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Xuehao Wang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yipu Zhao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Xiaohua Chu
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Qian Huang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Kang Ru
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Ling Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yu Li
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Zhihao Chen
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health EngineeringNorthwestern Polytechnical UniversityXi'anChina
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems EngineeringNorthwestern Polytechnical UniversityXi'anChina
- NPU‐UAB Joint Laboratory for Bone Metabolism, School of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
19
|
Adesanya O, Das D, Kalsotra A. Emerging roles of RNA-binding proteins in fatty liver disease. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1840. [PMID: 38613185 PMCID: PMC11018357 DOI: 10.1002/wrna.1840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 04/14/2024]
Abstract
A rampant and urgent global health issue of the 21st century is the emergence and progression of fatty liver disease (FLD), including alcoholic fatty liver disease and the more heterogenous metabolism-associated (or non-alcoholic) fatty liver disease (MAFLD/NAFLD) phenotypes. These conditions manifest as disease spectra, progressing from benign hepatic steatosis to symptomatic steatohepatitis, cirrhosis, and, ultimately, hepatocellular carcinoma. With numerous intricately regulated molecular pathways implicated in its pathophysiology, recent data have emphasized the critical roles of RNA-binding proteins (RBPs) in the onset and development of FLD. They regulate gene transcription and post-transcriptional processes, including pre-mRNA splicing, capping, and polyadenylation, as well as mature mRNA transport, stability, and translation. RBP dysfunction at every point along the mRNA life cycle has been associated with altered lipid metabolism and cellular stress response, resulting in hepatic inflammation and fibrosis. Here, we discuss the current understanding of the role of RBPs in the post-transcriptional processes associated with FLD and highlight the possible and emerging therapeutic strategies leveraging RBP function for FLD treatment. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
| | - Diptatanu Das
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center @ Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute of Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
20
|
Wang H, Yu L, Wang J, Zhang Y, Xu M, Lv C, Cui B, Yuan M, Zhang Y, Yan Y, Hui R, Wang Y. SLC35D3 promotes white adipose tissue browning to ameliorate obesity by NOTCH signaling. Nat Commun 2023; 14:7643. [PMID: 37996411 PMCID: PMC10667520 DOI: 10.1038/s41467-023-43418-5] [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: 02/20/2022] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
White adipose tissue browning can promote lipid burning to increase energy expenditure and improve adiposity. Here, we show that Slc35d3 expression is significantly lower in adipose tissues of obese mice. While adipocyte-specific Slc35d3 knockin is protected against diet-induced obesity, adipocyte-specific Slc35d3 knockout inhibits white adipose tissue browning and causes decreased energy expenditure and impaired insulin sensitivity in mice. Mechanistically, we confirm that SLC35D3 interacts with the NOTCH1 extracellular domain, which leads to the accumulation of NOTCH1 in the endoplasmic reticulum and thus inhibits the NOTCH1 signaling pathway. In addition, knockdown of Notch1 in mouse inguinal white adipose tissue mediated by orthotopic injection of AAV8-adiponectin-shNotch1 shows considerable improvement in obesity and glucolipid metabolism, which is more pronounced in adipocyte-specific Slc35d3 knockout mice than in knockin mice. Overall, in this study, we reveal that SLC35D3 is involved in obesity via NOTCH1 signaling, and low adipose SLC35D3 expression in obesity might be a therapeutic target for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Hongrui Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin'e Wang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Yaqing Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Mengchen Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Lv
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupeng Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
21
|
Guo YF, Sun JY, Liu Y, Liu ZY, Huang Y, Xiao Y, Su T. lncRNA Hnscr Regulates Lipid Metabolism by Mediating Adipocyte Lipolysis. Endocrinology 2023; 164:bqad147. [PMID: 37788569 PMCID: PMC10628467 DOI: 10.1210/endocr/bqad147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Obesity is a process of fat accumulation due to the imbalance between energy intake and consumption. Long noncoding RNA (lncRNA) Hnscr is crucial for metabolic regulation, but its roles in lipid metabolism during obesity are still unknown. In this article, we found that the expression of Hnscr gradually decreased in adipose tissues of diet-induced obese mice. Furthermore, the deletion of Hnscr promoted an increase in body weight and adipose tissue weight by upregulating the expression of lipogenesis genes and downregulating lipolysis genes in inguinal white adipose tissue (iWAT) and brown adipose tissue. In vitro knockdown of Hnscr in adipocytes resulted in reduced lipolysis of adipocytes. Overexpression of Hnscr by adenovirus or drug mimics showed the opposite. Mechanistically, Hnscr regulated adipose lipid metabolism by mediating the cyclic adenosine monophosphate/protein kinase A signaling pathway. This study identifies the initial characterization of Hnscr as a critical modifier that regulates lipid metabolism, suggesting that lncRNA Hnscr is a potential target for treating obesity.
Collapse
Affiliation(s)
- Yi-Fan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Jing-Yi Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Zhe-Yu Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yuan Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
22
|
Aibara D, Sakaguchi A, Matsusue K. Transcriptional regulation of adipogenin expression in liver steatosis by hepatic peroxisome proliferator-activated receptor gamma. Genes Cells 2023; 28:585-594. [PMID: 37249025 DOI: 10.1111/gtc.13052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023]
Abstract
The nuclear receptors peroxisome proliferator-activated receptor gamma (PPARγ) and adipogenin (ADIG) play vital roles in lipid metabolism. However, the interaction between PPARγ and ADIG during liver steatosis remains unclear. In this study, we aimed to investigate the role of PPARγ in the transcriptional regulation of hepatic ADIG expression. Adig was found to be highly expressed in various fatty liver mouse models. Although hepatic Adig was expressed at high levels in the fatty liver of type 2 diabetic ob/ob mice and was upregulated by PPARγ agonist treatment, it was expressed at significantly low levels in liver-specific Pparg-knockout mice. Moreover, hepatic Adig expression was observed in other mouse models of liver steatosis, such as the leptin receptor mutant db/db and alcohol-fed mice. Adig was also highly expressed in the white and brown adipose tissues, skeletal muscles, and heart of ob/ob mice. Reporter and electromobility shift assays showed that PPARγ positively regulates Adig transcriptional activity by directly binding to a functional PPARγ-responsive element in the promoter region. Our results indicate that Adig is a novel target gene of hepatic PPARγ in liver steatosis.
Collapse
Affiliation(s)
- Daisuke Aibara
- Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka, Japan
| | - Ai Sakaguchi
- Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka, Japan
| | - Kimihiko Matsusue
- Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
23
|
Imi Y, Amano R, Kasahara N, Obana Y, Hosooka T. Nicotinamide mononucleotide induces lipolysis by regulating ATGL expression via the SIRT1-AMPK axis in adipocytes. Biochem Biophys Rep 2023; 34:101476. [PMID: 37144119 PMCID: PMC10151261 DOI: 10.1016/j.bbrep.2023.101476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) -dependent protein deacetylase SIRT1 plays an important role in the regulation of metabolism. Although the administration of nicotinamide mononucleotide (NMN), a key NAD+ intermediate, has been shown to ameliorate metabolic disorders, such as insulin resistance and glucose intolerance, the direct effect of NMN on the regulation of lipid metabolism in adipocytes remains unclear. We here investigated the effect of NMN on lipid storage in 3T3-L1 differentiated adipocytes. Oil-red O staining showed that NMN treatment reduced lipid accumulation in these cells. NMN was found to enhance lipolysis in adipocytes since the concentration of glycerol in the media was increased by NMN treatment. Western blotting and real-time RT-PCR analysis revealed that adipose triglyceride lipase (ATGL) expression at both protein and mRNA level was increased with NMN treatment in 3T3-L1 adipocytes. Whereas NMN increased SIRT1 expression and AMPK activation, an AMPK inhibitor compound C restored the NMN-dependent upregulation of ATGL expression in these cells, suggesting that NMN upregulates ATGL expression through the SIRT1-AMPK axis. NMN administration significantly decreased subcutaneous fat mass in mice on a high-fat diet. We also found that adipocyte size in subcutaneous fat was decreased with NMN treatment. Consistent with the alteration of fat mass and adipocyte size, the ATGL expression in subcutaneous fat was slightly, albeit significantly, increased with NMN treatment. These results indicate that NMN suppresses subcutaneous fat mass in diet-induced obese mice, potentially in part via the upregulation of ATGL. Unexpectedly, the reduction in fat mass as well as ATGL upregulation with NMN treatment were not observed in epididymal fat, implying that the effects of NMN are site-specific in adipose tissue. Thus, these findings provide important insights into the mechanism of NMN/NAD+ in the regulation of metabolism.
Collapse
|
24
|
Merat R. The human antigen R as an actionable super-hub within the network of cancer cell persistency and plasticity. Transl Oncol 2023; 35:101722. [PMID: 37352624 DOI: 10.1016/j.tranon.2023.101722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023] Open
Abstract
In this perspective article, a clinically inspired phenotype-driven experimental approach is put forward to address the challenge of the adaptive response of solid cancers to small-molecule targeted therapies. A list of conditions is derived, including an experimental quantitative assessment of cell plasticity and an information theory-based detection of in vivo dependencies, for the discovery of post-transcriptional druggable mechanisms capable of preventing at multiple levels the emergence of plastic dedifferentiated slow-proliferating cells. The approach is illustrated by the author's own work in the example case of the adaptive response of BRAFV600-melanoma to BRAF inhibition. A bench-to-bedside and back to bench effort leads to a therapeutic strategy in which the inhibition of the baseline activity of the interferon-γ-activated inhibitor of translation (GAIT) complex, incriminated in the expression insufficiency of the RNA-binding protein HuR in a minority of cells, results in the suppression of the plastic, intermittently slow-proliferating cells involved in the adaptive response. A similar approach is recommended for the validation of other classes of mechanisms that we seek to modulate to overcome this complex challenge of modern cancer therapy.
Collapse
Affiliation(s)
- Rastine Merat
- Dermato-Oncology Unit, Division of Dermatology, Geneva University Hospitals, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
25
|
Ferrigno A, Campagnoli LIM, Barbieri A, Marchesi N, Pascale A, Croce AC, Vairetti M, Di Pasqua LG. MCD Diet Modulates HuR and Oxidative Stress-Related HuR Targets in Rats. Int J Mol Sci 2023; 24:9808. [PMID: 37372956 DOI: 10.3390/ijms24129808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The endogenous antioxidant defense plays a big part in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), a common metabolic disorder that can lead to serious complications such as cirrhosis and cancer. HuR, an RNA-binding protein of the ELAV family, controls, among others, the stability of MnSOD and HO-1 mRNA. These two enzymes protect the liver cells from oxidative damage caused by excessive fat accumulation. Our aim was to investigate the expression of HuR and its targets in a methionine-choline deficient (MCD) model of NAFLD. To this aim, we fed male Wistar rats with an MCD diet for 3 and 6 weeks to induce NAFLD; then, we evaluated the expression of HuR, MnSOD, and HO-1. The MCD diet induced fat accumulation, hepatic injury, oxidative stress, and mitochondrial dysfunction. A HuR downregulation was also observed in association with a reduced expression of MnSOD and HO-1. Moreover, the changes in the expression of HuR and its targets were significantly correlated with oxidative stress and mitochondrial injury. Since HuR plays a protective role against oxidative stress, targeting this protein could be a therapeutic strategy to both prevent and counteract NAFLD.
Collapse
Affiliation(s)
- Andrea Ferrigno
- Unit of Cellular and Molecular Pharmacology and Toxicology, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | | | - Annalisa Barbieri
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta Marchesi
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Alessia Pascale
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Anna Cleta Croce
- IGM-CNR, Unit of Histochemistry and Cytometry, University of Pavia, 27100 Pavia, Italy
| | - Mariapia Vairetti
- Unit of Cellular and Molecular Pharmacology and Toxicology, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Laura Giuseppina Di Pasqua
- Unit of Cellular and Molecular Pharmacology and Toxicology, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
26
|
Sachse M, Tual-Chalot S, Ciliberti G, Amponsah-Offeh M, Stamatelopoulos K, Gatsiou A, Stellos K. RNA-binding proteins in vascular inflammation and atherosclerosis. Atherosclerosis 2023; 374:55-73. [PMID: 36759270 DOI: 10.1016/j.atherosclerosis.2023.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/01/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the major cause of premature death and disability worldwide, even when patients with an established manifestation of atherosclerotic heart disease are optimally treated according to the clinical guidelines. Apart from the epigenetic control of transcription of the genetic information to messenger RNAs (mRNAs), gene expression is tightly controlled at the post-transcriptional level before the initiation of translation. Although mRNAs are traditionally perceived as the messenger molecules that bring genetic information from the nuclear DNA to the cytoplasmic ribosomes for protein synthesis, emerging evidence suggests that processes controlling RNA metabolism, driven by RNA-binding proteins (RBPs), affect cellular function in health and disease. Over the recent years, vascular endothelial cell, smooth muscle cell and immune cell RBPs have emerged as key co- or post-transcriptional regulators of several genes related to vascular inflammation and atherosclerosis. In this review, we provide an overview of cell-specific function of RNA-binding proteins involved in all stages of ASCVD and how this knowledge may be used for the development of novel precision medicine therapeutics.
Collapse
Affiliation(s)
- Marco Sachse
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Cardiovascular Surgery, University Heart Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Michael Amponsah-Offeh
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany; Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany.
| |
Collapse
|
27
|
Michel LYM. Extracellular Vesicles in Adipose Tissue Communication with the Healthy and Pathological Heart. Int J Mol Sci 2023; 24:ijms24097745. [PMID: 37175451 PMCID: PMC10177965 DOI: 10.3390/ijms24097745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023] Open
Abstract
Adipose tissue and its diverse cell types constitute one of the largest endocrine organs. With multiple depot locations, adipose tissue plays an important regulatory role through paracrine and endocrine communication, particularly through the secretion of a wide range of bioactive molecules, such as nucleic acids, proteins, lipids or adipocytokines. Over the past several years, research has uncovered a myriad of interorgan communication signals mediated by small lipid-derived nanovesicles known as extracellular vesicles (EVs), in which secreted bioactive molecules are stably transported as cargo molecules and delivered to adjacent cells or remote organs. EVs constitute an essential part of the human adipose secretome, and there is a growing body of evidence showing the crucial implications of adipose-derived EVs in the regulation of heart function and its adaptative capacity. The adipose tissue modifications and dysfunction observed in obesity and aging tremendously affect the adipose-EV secretome, with important consequences for the myocardium. The present review presents a comprehensive analysis of the findings in this novel area of research, reports the key roles played by adipose-derived EVs in interorgan cross-talk with the heart and discusses their implications in physiological and pathological conditions affecting adipose tissue and/or the heart (pressure overload, ischemia, diabetic cardiomyopathy, etc.).
Collapse
Affiliation(s)
- Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 57 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
28
|
Aragón-Herrera A, Moraña-Fernández S, Otero-Santiago M, Anido-Varela L, Campos-Toimil M, García-Seara J, Román A, Seijas J, García-Caballero L, Rodríguez J, Tarazón E, Roselló-Lletí E, Portolés M, Lage R, Gualillo O, González-Juanatey JR, Feijóo-Bandín S, Lago F. The lipidomic and inflammatory profiles of visceral and subcutaneous adipose tissues are distinctly regulated by the SGLT2 inhibitor empagliflozin in Zucker diabetic fatty rats. Biomed Pharmacother 2023; 161:114535. [PMID: 36931025 DOI: 10.1016/j.biopha.2023.114535] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The pharmacological inhibition of sodium-glucose cotransporter 2 (SGLT2) has emerged as a treatment for patients with type 2 diabetes mellitus (T2DM), cardiovascular disease and/or other metabolic disturbances, although some of the mechanisms implicated in their beneficial effects are unknown. The SGLT2 inhibitor (SGLT2i) empagliflozin has been suggested as a regulator of adiposity, energy metabolism, and systemic inflammation in adipose tissue. The aim of our study was to evaluate the impact of a 6-week-empagliflozin treatment on the lipidome of visceral (VAT) and subcutaneous adipose tissue (SAT) from diabetic obese Zucker Diabetic Fatty (ZDF) rats using an untargeted metabolomics approach. We found that empagliflozin increases the content of diglycerides and oxidized fatty acids (FA) in VAT, while in SAT, it decreases the levels of several lysophospholipids and increases 2 phosphatidylcholines. Empagliflozin also reduces the expression of the cytokines interleukin-1 beta (IL-1β), IL-6, tumor necrosis factor-alpha (TNFα), monocyte-chemotactic protein-1 (MCP-1) and IL-10, and of Cd86 and Cd163 M1 and M2 macrophage markers in VAT, with no changes in SAT, except for a decrease in IL-1β. Empagliflozin treatment also shows an effect on lipolysis increasing the expression of hormone-sensitive lipase (HSL) in SAT and VAT and of adipose triglyceride lipase (ATGL) in VAT, together with a decrease in the adipose content of the FA transporter cluster of differentiation 36 (CD36). In conclusion, our data highlighted differences in the VAT and SAT lipidomes, inflammatory profiles and lipolytic function, which suggest a distinct metabolism of these two white adipose tissue depots after the empagliflozin treatment.
Collapse
Affiliation(s)
- Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Sandra Moraña-Fernández
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) and Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS). Av. Barcelona, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Manuel Otero-Santiago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Laura Anido-Varela
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Manuel Campos-Toimil
- Group of Pharmacology of Chronic Diseases (CD Pharma), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Spain
| | - Javier García-Seara
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Arrhytmia Unit, Clinical University Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ana Román
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Cardiology Department, Clinical University Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - José Seijas
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Cardiology Department, Clinical University Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Lucía García-Caballero
- Department of Morphological Sciences, School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Javier Rodríguez
- Clinical Biochemistry Laboratory, Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Estefanía Tarazón
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Esther Roselló-Lletí
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Manuel Portolés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Ricardo Lage
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) and Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS). Av. Barcelona, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Oreste Gualillo
- Laboratory of Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Cardiology Department, Clinical University Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain.
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Xu J, Liu X, Wu S, Zhang D, Liu X, Xia P, Ling J, Zheng K, Xu M, Shen Y, Zhang J, Yu P. RNA-binding proteins in metabolic-associated fatty liver disease (MAFLD): From mechanism to therapy. Biosci Trends 2023; 17:21-37. [PMID: 36682800 DOI: 10.5582/bst.2022.01473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease globally and seriously increases the public health burden, affecting approximately one quarter of the world population. Recently, RNA binding proteins (RBPs)-related pathogenesis of MAFLD has received increasing attention. RBPs, vividly called the gate keepers of MAFLD, play an important role in the development of MAFLD through transcription regulation, alternative splicing, alternative polyadenylation, stability and subcellular localization. In this review, we describe the mechanisms of different RBPs in the occurrence and development of MAFLD, as well as list some drugs that can improve MAFLD by targeting RBPs. Considering the important role of RBPs in the development of MAFLD, elucidating the RNA regulatory networks involved in RBPs will facilitate the design of new drugs and biomarkers discovery.
Collapse
Affiliation(s)
- Jiawei Xu
- The Second Clinical Medical College / The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xingyu Liu
- The Second Clinical Medical College / The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shuqin Wu
- The Second Clinical Medical College / The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Xiao Liu
- Department of Cardiology, The Second Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kai Zheng
- Medical Care Strategic Customer Department, China Merchants Bank Shenzhen Branch, Shenzhen, Guangdong, Guangdong, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Zhang
- The Second Clinical Medical College / The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- The Second Clinical Medical College / The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
30
|
Yamamoto K, Yamashita M, Oda M, Tjendana Tjhin V, Inagawa H, Soma GI. Oral Administration of Lipopolysaccharide Enhances Insulin Signaling-Related Factors in the KK/Ay Mouse Model of Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054619. [PMID: 36902049 PMCID: PMC10003108 DOI: 10.3390/ijms24054619] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Lipopolysaccharide (LPS), an endotoxin, induces systemic inflammation by injection and is thought to be a causative agent of chronic inflammatory diseases, including type 2 diabetes mellitus (T2DM). However, our previous studies found that oral LPS administration does not exacerbate T2DM conditions in KK/Ay mice, which is the opposite of the response from LPS injection. Therefore, this study aims to confirm that oral LPS administration does not aggravate T2DM and to investigate the possible mechanisms. In this study, KK/Ay mice with T2DM were orally administered LPS (1 mg/kg BW/day) for 8 weeks, and blood glucose parameters before and after oral administration were compared. Abnormal glucose tolerance, insulin resistance progression, and progression of T2DM symptoms were suppressed by oral LPS administration. Furthermore, the expressions of factors involved in insulin signaling, such as insulin receptor, insulin receptor substrate 1, thymoma viral proto-oncogene, and glucose transporter type 4, were upregulated in the adipose tissues of KK/Ay mice, where this effect was observed. For the first time, oral LPS administration induces the expression of adiponectin in adipose tissues, which is involved in the increased expression of these molecules. Briefly, oral LPS administration may prevent T2DM by inducing an increase in the expressions of insulin signaling-related factors based on adiponectin production in adipose tissues.
Collapse
Affiliation(s)
- Kazushi Yamamoto
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Masashi Yamashita
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Masataka Oda
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Vindy Tjendana Tjhin
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Hiroyuki Inagawa
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-0841, Japan
| | - Gen-Ichiro Soma
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-0841, Japan
- Correspondence: ; Tel.: +81-87-813-9201
| |
Collapse
|
31
|
Wang Z, Wang H, Guo C, Yu F, Zhang Y, Qiao L, Zhang H, Zhang C. Role of hsa_circ_0000280 in regulating vascular smooth muscle cell function and attenuating neointimal hyperplasia via ELAVL1. Cell Mol Life Sci 2023; 80:3. [PMID: 36477660 PMCID: PMC9729135 DOI: 10.1007/s00018-022-04602-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/25/2022] [Accepted: 10/07/2022] [Indexed: 12/12/2022]
Abstract
The pathological proliferation of cells in vascular smooth muscle underlies neointimal hyperplasia (NIH) development during atherosclerosis. Circular RNAs (circRNAs), which represent novel functional biomarkers and RNA-binding proteins, contribute to multiple cardiovascular diseases; however, their roles in regulating the vascular smooth muscle cell cycle remain unknown. Thus, we aimed to identify the roles of circRNAs in vascular smooth muscle during coronary heart disease (CHD). Through circRNA sequencing of CHD samples and human antigen R (ELAVL1) immunoprecipitation, we identified circRNAs that are associated with CHD and interact with ELAVL1. Our results suggested that the hsa_circ_0000280 associated with CHD inhibits cell proliferation and induces ELAVL1-dependent cell cycle arrest. Gain/loss-of-function experiments and assays in vivo indicated that hsa_circ_0000280 facilitates interactions between ELAVL1 and cyclin-dependent kinase suppressor 1 (CDKN1A) mRNA and stabilization of this complex and leads to cell cycle arrest at the G1/S checkpoint, inhibiting cell proliferation of vascular smooth muscle cells in vitro and NIH in vivo. Importantly, hsa_circ_0000280 reduced neointimal thickness and smooth muscle cell proliferation in vivo. Taken together, these findings reveal a novel pathway in which hsa_circ_0000280 facilitates the regulation of ELAVL1 on CDKN1A mRNA to inhibit NIH. Therefore, measuring and modulating their expression might represent a potential diagnostic or therapeutic strategy for CHD.
Collapse
Affiliation(s)
- Zunzhe Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Huating Wang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, China
| | - Chenghu Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Fangpu Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Ya Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Lei Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China
| | - Haijun Zhang
- Institute of Vascular Intervention, Medical College of Tongji University, Shanghai, 200072, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan City, 250012, Shandong, China.
| |
Collapse
|
32
|
Wang Y, Tai YL, Way G, Zeng J, Zhao D, Su L, Jiang X, Jackson KG, Wang X, Gurley EC, Liu J, Liu J, Chen W, Wang XY, Sanyal AJ, Hylemon PB, Zhou H. RNA binding protein HuR protects against NAFLD by suppressing long noncoding RNA H19 expression. Cell Biosci 2022; 12:172. [PMID: 36224648 PMCID: PMC9558407 DOI: 10.1186/s13578-022-00910-7] [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: 07/26/2022] [Accepted: 10/06/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND NAFLD has become the most common chronic liver disease worldwide. Human antigen R (HuR), an RNA-binding protein, is an important post-transcriptional regulator. HuR has been reported as a key player in regulating lipid homeostasis in the liver and adipose tissues by using tissue-specific HuR knockout mice. However, the underlying mechanism by which hepatocyte-specific HuR regulates hepatic lipid metabolism under metabolic stress remains unclear and is the focus of this study. METHODS Hepatocyte-specific HuR deficient mice (HuRhKO) and age-/gender-matched control mice, as well as long-noncoding RNA H19 knockout mice (H19-/-), were fed a Western Diet plus sugar water (WDSW). Hepatic lipid accumulation, inflammation and fibrosis were examined by histology, RNA transcriptome analysis, qRT-PCR, and Western blot analysis. Bile acid composition was measured using LC-MS/MS. RESULTS Hepatocyte-specific deletion of HuR not only significantly increased hepatic lipid accumulation by modulating fatty acid synthesis and metabolism but also markedly induced inflammation by increasing immune cell infiltration and neutrophil activation under metabolic stress. In addition, hepatic deficiency of HuR disrupted bile acid homeostasis and enhanced liver fibrosis. Mechanistically, HuR is a repressor of H19 expression. Analysis of a recently published dataset (GSE143358) identified H19 as the top-upregulated gene in liver-specific HuR knockout mice. Similarly, hepatocyte-specific deficiency of HuR dramatically induced the expression of H19 and sphingosine-1 phosphate receptor 2 (S1PR2), but reduced the expression of sphingosine kinase 2 (SphK2). WDSW-induced hepatic lipid accumulation was alleviated in H19-/- mice. Furthermore, the downregulation of H19 alleviated WDSW-induced NAFLD in HuRhKO mice. CONCLUSIONS HuR not only functions as an RNA binding protein to modulate post-transcriptional gene expression but also regulates H19 promoter activity. Hepatic HuR is an important regulator of hepatic lipid metabolism via modulating H19 expression.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, China
| | - Yun-Ling Tai
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
| | - Grayson Way
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Jing Zeng
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
| | - Derrick Zhao
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Lianyong Su
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Xixian Jiang
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Kaitlyn G. Jackson
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Xuan Wang
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Emily C. Gurley
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Jinze Liu
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA USA
| | - Jinpeng Liu
- Department of Computer Science, University of Kentucky, Lexington, KY USA
| | - Weidong Chen
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, China
| | - Xiang-Yang Wang
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA
- Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA USA
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Arun J. Sanyal
- Department of Internal Medicine/GI Division, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Phillip B. Hylemon
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| | - Huiping Zhou
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678 USA
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, VA USA
| |
Collapse
|
33
|
Guo YY, Li BY, Xiao G, Liu Y, Guo L, Tang QQ. Cdo1 promotes PPARγ-mediated adipose tissue lipolysis in male mice. Nat Metab 2022; 4:1352-1368. [PMID: 36253617 DOI: 10.1038/s42255-022-00644-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/22/2022] [Indexed: 01/20/2023]
Abstract
Cysteine dioxygenase 1 (Cdo1) is a key enzyme in taurine synthesis. Here we show that Cdo1 promotes lipolysis in adipose tissue. Adipose-specific knockout of Cdo1 in mice impairs energy expenditure, cold tolerance and lipolysis, exacerbates diet-induced obesity (DIO) and decreases adipose expression of the key lipolytic genes encoding ATGL and HSL, with little effect on adipose taurine levels. White-adipose-specific overexpression of ATGL and HSL blunts the role of adipose Cdo1 deficiency in promoting DIO. Mechanistically, Cdo1 interacts with PPARγ and facilitates the recruitment of Med24, the core subunit of mediator complex, to ATGL and HSL gene promoters, thereby transactivating their expression. Further, mice with transgenic overexpression of Cdo1 show better cold tolerance, ameliorated DIO and higher lipolysis capacity. Thus, we uncover an unexpected and important role of Cdo1 in regulating adipose lipolysis.
Collapse
Affiliation(s)
- Ying-Ying Guo
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 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, Shanghai, China
| | - Gang Xiao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 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, Shanghai, China
| | - Liang Guo
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, and School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
34
|
Majumder M, Chakraborty P, Mohan S, Mehrotra S, Palanisamy V. HuR as a molecular target for cancer therapeutics and immune-related disorders. Adv Drug Deliv Rev 2022; 188:114442. [PMID: 35817212 DOI: 10.1016/j.addr.2022.114442] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/12/2022] [Accepted: 07/05/2022] [Indexed: 11/19/2022]
Abstract
The control of eukaryotic gene expression occurs at multiple levels, from transcription to messenger RNA processing, transport, localization, turnover, and translation. RNA-binding proteins control gene expression and are involved in different stages of mRNA processing, including splicing, maturation, turnover, and translation. A ubiquitously expressed RBP Human antigen R is engaged in the RNA processes mentioned above but, most importantly, controls mRNA stability and turnover. Dysregulation of HuR is linked to many diseases, including cancer and other immune-related disorders. HuR targets mRNAs containing AU-rich elements at their 3'untranslated region, which encodes proteins involved in cell growth, proliferation, tumor formation, angiogenesis, immune evasion, inflammation, invasion, and metastasis. HuR overexpression has been reported in many tumor types, which led to a poor prognosis for patients. Hence, HuR is considered an appealing drug target for cancer treatment. Therefore, multiple attempts have been made to identify small molecule inhibitors for blocking HuR functions. This article reviews the current prospects of drugs that target HuR in numerous cancer types, their mode of action, and off-target effects. Furthermore, we will summarize drugs that interfered with HuR-RNA interactions and established themselves as novel therapeutics. We will also highlight the significance of HuR overexpression in multiple cancers and discuss its role in immune functions. This review provides evidence of a new era of HuR-targeted small molecules that can be used for cancer therapeutics either as a monotherapy or in combination with other cancer treatment modalities.
Collapse
Affiliation(s)
- Mrinmoyee Majumder
- Department of Biochemistry and Molecular Biology, Charleston, SC 29425, USA
| | - Paramita Chakraborty
- Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sarumathi Mohan
- Department of Biochemistry and Molecular Biology, Charleston, SC 29425, USA
| | - Shikhar Mehrotra
- Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | | |
Collapse
|
35
|
Inhibition of the DAPKs-L13a axis prevents a GAIT-like motif-mediated HuR insufficiency in melanoma cells. Biochem Biophys Res Commun 2022; 626:21-29. [DOI: 10.1016/j.bbrc.2022.07.086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022]
|
36
|
RNA-Binding Proteins in the Regulation of Adipogenesis and Adipose Function. Cells 2022; 11:cells11152357. [PMID: 35954201 PMCID: PMC9367552 DOI: 10.3390/cells11152357] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
The obesity epidemic represents a critical public health issue worldwide, as it is a vital risk factor for many diseases, including type 2 diabetes (T2D) and cardiovascular disease. Obesity is a complex disease involving excessive fat accumulation. Proper adipose tissue accumulation and function are highly transcriptional and regulated by many genes. Recent studies have discovered that post-transcriptional regulation, mainly mediated by RNA-binding proteins (RBPs), also plays a crucial role. In the lifetime of RNA, it is bound by various RBPs that determine every step of RNA metabolism, from RNA processing to alternative splicing, nucleus export, rate of translation, and finally decay. In humans, it is predicted that RBPs account for more than 10% of proteins based on the presence of RNA-binding domains. However, only very few RBPs have been studied in adipose tissue. The primary aim of this paper is to provide an overview of RBPs in adipogenesis and adipose function. Specifically, the following best-characterized RBPs will be discussed, including HuR, PSPC1, Sam68, RBM4, Ybx1, Ybx2, IGF2BP2, and KSRP. Characterization of these proteins will increase our understanding of the regulatory mechanisms of RBPs in adipogenesis and provide clues for the etiology and pathology of adipose-tissue-related diseases.
Collapse
|
37
|
Li R, Li G, Hai Y, Li T, Bian Y, Ma T. The effect of aerobic exercise on the lipophagy of adipose tissue in obese male mice. Chem Phys Lipids 2022; 247:105225. [PMID: 35810833 DOI: 10.1016/j.chemphyslip.2022.105225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/27/2022] [Accepted: 07/06/2022] [Indexed: 01/18/2023]
Abstract
This article explores the obesity state and the changes in the level of lipophagy in adipose tissue after exercise to lose weight, so as to provide direction and basis for theoretical research on obesity prevention and control. We established a high-fat diet model of obese mice, and applied exercise intervention and intraperitoneal injection of chloroquine to inhibit autophagy. Long-term high-fat diet can cause obesity in mice, and the process of lipophagy is inhibited, which may be one of the reasons for fat accumulation. Eight weeks of aerobic exercise can effectively reduce the weight of obese mice and promote lipolysis; this process is mainly completed by lipase decomposition, in addition to require the participation of the lipophagy process.
Collapse
Affiliation(s)
- Rendong Li
- Physical Education Department, Shenyang University of Chemical Technology, Shenyang Economic and Technological Development Zone, Shenyang 110142, PR China.
| | - Guangkuan Li
- Department of Postgraduate, Shenyang Sport University, Shenyang 110102, PR China.
| | - Yan Hai
- Department of Postgraduate, Shenyang Sport University, Shenyang 110102, PR China.
| | - Tao Li
- Department of Postgraduate, Shenyang Sport University, Shenyang 110102, PR China.
| | - Yuanyuan Bian
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China.
| | - Tie Ma
- The College of Kinesiology, Shenyang Sport University, Shenyang 110102, PR China.
| |
Collapse
|
38
|
Latorre J, Aroca A, Fernández-Real JM, Romero LC, Moreno-Navarrete JM. The Combined Partial Knockdown of CBS and MPST Genes Induces Inflammation, Impairs Adipocyte Function-Related Gene Expression and Disrupts Protein Persulfidation in Human Adipocytes. Antioxidants (Basel) 2022; 11:antiox11061095. [PMID: 35739994 PMCID: PMC9220337 DOI: 10.3390/antiox11061095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Recent studies in mice and humans demonstrated the relevance of H2S synthesising enzymes, such as CTH, CBS, and MPST, in the physiology of adipose tissue and the differentiation of preadipocyte into adipocytes. Here, our objective was to investigate the combined role of CTH, CBS, and MPST in the preservation of adipocyte protein persulfidation and adipogenesis. Combined partial CTH, CBS, and MPST gene knockdown was achieved treating fully human adipocytes with siRNAs against these transcripts (siRNA_MIX). Adipocyte protein persulfidation was analyzed using label-free quantitative mass spectrometry coupled with a dimedone-switch method for protein labeling and purification. Proteomic analysis quantified 216 proteins with statistically different levels of persulfidation in KD cells compared to control adipocytes. In fully differentiated adipocytes, CBS and MPST mRNA and protein levels were abundant, while CTH expression was very low. It is noteworthy that siRNA_MIX administration resulted in a significant decrease in CBS and MPST expression, without impacting on CTH. The combined partial knockdown of the CBS and MPST genes resulted in reduced cellular sulfide levels in parallel to decreased expression of relevant genes for adipocyte biology, including adipogenesis, mitochondrial biogenesis, and lipogenesis, but increased proinflammatory- and senescence-related genes. It should be noted that the combined partial knockdown of CBS and MPST genes also led to a significant disruption in the persulfidation pattern of the adipocyte proteins. Although among the less persulfidated proteins, we identified several relevant proteins for adipocyte adipogenesis and function, among the most persulfidated, key mediators of adipocyte inflammation and dysfunction as well as some proteins that might play a positive role in adipogenesis were found. In conclusion, the current study indicates that the combined partial elimination of CBS and MPST (but not CTH) in adipocytes affects the expression of genes related to the maintenance of adipocyte function and promotes inflammation, possibly by altering the pattern of protein persulfidation in these cells, suggesting that these enzymes were required for the functional maintenance of adipocytes.
Collapse
Affiliation(s)
- Jessica Latorre
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), 17190 Salt, Spain; (J.L.); (J.M.F.-R.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/010), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Angeles Aroca
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones and Universidad de Sevilla, 41092 Seville, Spain; (A.A.); (L.C.R.)
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), 17190 Salt, Spain; (J.L.); (J.M.F.-R.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/010), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, Universitat de Girona, 17003 Girona, Spain
| | - Luis C. Romero
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones and Universidad de Sevilla, 41092 Seville, Spain; (A.A.); (L.C.R.)
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), 17190 Salt, Spain; (J.L.); (J.M.F.-R.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/010), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-872-987087 (ext. 70)
| |
Collapse
|
39
|
Wu X, Xu L. The RNA-binding protein HuR in human cancer: A friend or foe? Adv Drug Deliv Rev 2022; 184:114179. [PMID: 35248670 PMCID: PMC9035123 DOI: 10.1016/j.addr.2022.114179] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/26/2022] [Accepted: 02/27/2022] [Indexed: 12/12/2022]
Abstract
The RNA-binding proteins (RBPs) are critical trans factors that associate with specific cis elements present in mRNAs whose stability and translation are subject to regulation. The RBP Hu antigen R (HuR) is overexpressed in a wide variety of human cancers and serves as a prognostic factor of poor clinical outcome. HuR promotes tumorigenesis by interacting with a subset of oncogenic mRNAs implicated in different cancer hallmarks, and resistance to therapy. Reduction of HuR levels in cancer cells leads to tumor regression in mouse xenograft models. These findings prompt a working model whereby cancer cells use HuR, a master switch of multiple oncogenic mRNAs, to drive drug resistance and promote cell survival and metastasis, thus rendering the tumor cells with high cytoplasmic HuR more progressive and resistant to therapy. This review summarizes the roles of HuR in cancer and other diseases, therapeutic potential of HuR inhibition, and the current status of drug discovery on HuR.
Collapse
Affiliation(s)
- Xiaoqing Wu
- Higuchi Biosciences Center, The University of Kansas, Lawrence, KS, USA; The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS, USA.
| | - Liang Xu
- The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS, USA; Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA; Department of Radiation Oncology, The University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
40
|
Assoni G, La Pietra V, Digilio R, Ciani C, Licata NV, Micaelli M, Facen E, Tomaszewska W, Cerofolini L, Pérez-Ràfols A, Varela Rey M, Fragai M, Woodhoo A, Marinelli L, Arosio D, Bonomo I, Provenzani A, Seneci P. HuR-targeted agents: An insight into medicinal chemistry, biophysical, computational studies and pharmacological effects on cancer models. Adv Drug Deliv Rev 2022; 181:114088. [PMID: 34942276 DOI: 10.1016/j.addr.2021.114088] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/07/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Abstract
The Human antigen R (HuR) protein is an RNA-binding protein, ubiquitously expressed in human tissues, that orchestrates target RNA maturation and processing both in the nucleus and in the cytoplasm. A survey of known modulators of the RNA-HuR interactions is followed by a description of its structure and molecular mechanism of action - RRM domains, interactions with RNA, dimerization, binding modes with naturally occurring and synthetic HuR inhibitors. Then, the review focuses on HuR as a validated molecular target in oncology and briefly describes its role in inflammation. Namely, we show ample evidence for the involvement of HuR in the hallmarks and enabling characteristics of cancer, reporting findings from in vitro and in vivo studies; and we provide abundant experimental proofs of a beneficial role for the inhibition of HuR-mRNA interactions through silencing (CRISPR, siRNA) or pharmacological inhibition (small molecule HuR inhibitors).
Collapse
Affiliation(s)
- Giulia Assoni
- Chemistry Department, University of Milan, Via Golgi 19, I-20133 Milan, Italy; Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Valeria La Pietra
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Rosangela Digilio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Caterina Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Nausicaa Valentina Licata
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Mariachiara Micaelli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Elisa Facen
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Weronika Tomaszewska
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence and Interuniversity Consortium for Magnetic Resonance of Metalloproteins (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Anna Pérez-Ràfols
- Giotto Biotech S.R.L., Via Madonna del Piano 6, 50019 Sesto Fiorentino (FI), Italy
| | - Marta Varela Rey
- Gene Regulatory Control in Disease Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence and Interuniversity Consortium for Magnetic Resonance of Metalloproteins (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, Spain; Department of Functional Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain; Center for Cooperative Research in Biosciences (CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Luciana Marinelli
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Chimiche "G. Natta" (SCITEC), National Research Council (CNR), Via C. Golgi 19, I-20133 Milan, Italy
| | - Isabelle Bonomo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy.
| | - Pierfausto Seneci
- Chemistry Department, University of Milan, Via Golgi 19, I-20133 Milan, Italy.
| |
Collapse
|
41
|
Pérez-García A, Torrecilla-Parra M, Fernández-de Frutos M, Martín-Martín Y, Pardo-Marqués V, Ramírez CM. Posttranscriptional Regulation of Insulin Resistance: Implications for Metabolic Diseases. Biomolecules 2022; 12:biom12020208. [PMID: 35204710 PMCID: PMC8961590 DOI: 10.3390/biom12020208] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance defines an impairment in the biologic response to insulin action in target tissues, primarily the liver, muscle, adipose tissue, and brain. Insulin resistance affects physiology in many ways, causing hyperglycemia, hypertension, dyslipidemia, visceral adiposity, hyperinsulinemia, elevated inflammatory markers, and endothelial dysfunction, and its persistence leads to the development metabolic disease, including diabetes, obesity, cardiovascular disease, or nonalcoholic fatty liver disease (NAFLD), as well as neurological disorders such as Alzheimer’s disease. In addition to classical transcriptional factors, posttranscriptional control of gene expression exerted by microRNAs and RNA-binding proteins constitutes a new level of regulation with important implications in metabolic homeostasis. In this review, we describe miRNAs and RBPs that control key genes involved in the insulin signaling pathway and related regulatory networks, and their impact on human metabolic diseases at the molecular level, as well as their potential use for diagnosis and future therapeutics.
Collapse
|
42
|
Deficiency of Cathelicidin Attenuates High-Fat Diet Plus Alcohol-Induced Liver Injury through FGF21/Adiponectin Regulation. Cells 2021; 10:cells10123333. [PMID: 34943840 PMCID: PMC8699208 DOI: 10.3390/cells10123333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol consumption and obesity are known risk factors of steatohepatitis. Here, we report that the deficiency of CRAMP (cathelicidin-related antimicrobial peptide—gene name: Camp) is protective against a high-fat diet (HFD) plus acute alcohol (HFDE)-induced liver injury. HFDE markedly induced liver injury and steatosis in WT mice, which were attenuated in Camp–/– mice. Neutrophil infiltration was lessened in the liver of Camp–/– mice. HFDE feeding dramatically increased epididymal white adipose tissue (eWAT) mass and induced adipocyte hypertrophy in WT mice, whereas these effects were attenuated by the deletion of Camp. Furthermore, Camp–/– mice had significantly increased eWAT lipolysis, evidenced by up-regulated expression of lipolytic enzymes, adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL). The depletion of Camp also increased uncoupling protein 1 (UCP1)-dependent thermogenesis in the brown adipose tissue (BAT) of mice. HFDE fed Camp–/– mice had elevated protein levels of fibroblast growth factor 21 (FGF21) in the eWAT, with an increased adiponectin production, which had been shown to alleviate hepatic fat deposition and inflammation. Collectively, we have demonstrated that Camp–/– mice are protected against HFD plus alcohol-induced liver injury and steatosis through FGF21/adiponectin regulation. Targeting CRAMP could be an effective approach for prevention/treatment of high-fat diet plus alcohol consumption-induced steatohepatitis.
Collapse
|
43
|
Rajan S, de Guzman HC, Palaia T, Goldberg IJ, Hussain MM. A simple, rapid, and sensitive fluorescence-based method to assess triacylglycerol hydrolase activity. J Lipid Res 2021; 62:100115. [PMID: 34508728 PMCID: PMC8488599 DOI: 10.1016/j.jlr.2021.100115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 01/22/2023] Open
Abstract
Lipases constitute an important class of water-soluble enzymes that catalyze the hydrolysis of hydrophobic triacylglycerol (TAG). Their enzymatic activity is typically measured using multistep procedures involving isolation and quantification of the hydrolyzed products. We report here a new fluorescence method to measure lipase activity in real time that does not require the separation of substrates from products. We developed this method using adipose triglyceride lipase (ATGL) and lipoprotein lipase (LpL) as model lipases. We first incubated a source of ATGL or LpL with substrate vesicles containing nitrobenzoxadiazole (NBD)-labeled TAG, then measured increases in NBD fluorescence, and calculated enzyme activities. Incorporation of NBD-TAG into phosphatidylcholine (PC) vesicles resulted in some hydrolysis; however, incorporation of phosphatidylinositol into these NBD-TAG/PC vesicles and increasing the ratio of NBD-TAG to PC greatly enhanced substrate hydrolysis. This assay was also useful in measuring the activity of pancreatic lipase and hormone-sensitive lipase. Next, we tested several small-molecule lipase inhibitors and found that orlistat inhibits all lipases, indicating that it is a pan-lipase inhibitor. In short, we describe a simple, rapid, fluorescence-based triacylglycerol hydrolysis assay to assess four major TAG hydrolases: intracellular ATGL and hormone-sensitive lipase, LpL localized at the extracellular endothelium, and pancreatic lipase present in the intestinal lumen. The major advantages of this method are its speed, simplicity, and elimination of product isolation. This assay is potentially applicable to a wide range of lipases, is amenable to high-throughput screening to discover novel modulators of triacylglycerol hydrolases, and can be used for diagnostic purposes.
Collapse
Affiliation(s)
- Sujith Rajan
- Department of Foundations of Medicine, NYU Long Island School of Medicine, and Diabetes and Obesity Research Center, NYU Langone Hospitals - Long Island, Mineola, NY, USA
| | - Hazel C de Guzman
- Department of Foundations of Medicine, NYU Long Island School of Medicine, and Diabetes and Obesity Research Center, NYU Langone Hospitals - Long Island, Mineola, NY, USA; Department of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas Palaia
- Department of Foundations of Medicine, NYU Long Island School of Medicine, and Diabetes and Obesity Research Center, NYU Langone Hospitals - Long Island, Mineola, NY, USA
| | - Ira J Goldberg
- Division of Endocrinology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, and Diabetes and Obesity Research Center, NYU Langone Hospitals - Long Island, Mineola, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
44
|
Kong C, Lyu D, He C, Li R, Lu Q. Dioscin elevates lncRNA MANTIS in therapeutic angiogenesis for heart diseases. Aging Cell 2021; 20:e13392. [PMID: 34081836 PMCID: PMC8282240 DOI: 10.1111/acel.13392] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/13/2021] [Accepted: 05/08/2021] [Indexed: 01/16/2023] Open
Abstract
Dioscin has been widely used in clinics for coronary artery disease (CAD) treatment for years in China. However, the underlying mechanism for Dioscin‐mediated cardioprotective effect has not been elucidated. Here, we showed that Dioscin significantly rescues the cardiac function in mouse model of myocardial infarction (MI), accompanied by the reduction of cardiac fibrosis and apoptosis, resulting from elevated angiogenesis. Mechanistically, Dioscin promotes the proliferation and migration of hypoxic endothelial cells via the up‐regulation of lncRNA MANTIS, which serves as a scaffolding lncRNA within a chromatin remodeling complex. Meanwhile, it enables pol II binding to the transcription start sites, which leads to induced expression of angiogenesis‐related genes, including SOX18, SMAD6, and COUP‐TFII. Conversely, IncRNA MANTIS silencing prevents Dioscin‐induced migration and angiogenesis in hypoxic endothelial cells. Taken together, these data provide new insights that clarifies the cardioprotective effects of Dioscin against myocardial infarcted injury and confirms the effect on angiogenic activity of endothelial cells. This will build a solid theoretical basis for clinical therapeutic strategies.
Collapse
Affiliation(s)
- Chuiyu Kong
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing China
| | - Dayin Lyu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing China
| | - Chang He
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing China
| | - Rui Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing China
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing China
| |
Collapse
|
45
|
Female Mice Are Protected from Metabolic Decline Associated with Lack of Skeletal Muscle HuR. BIOLOGY 2021; 10:biology10060543. [PMID: 34204316 PMCID: PMC8233974 DOI: 10.3390/biology10060543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 11/24/2022]
Abstract
Simple Summary Metabolic flexibility describes the ability to adapt to utilization of metabolic fuels such as carbohydrates, lipids, and proteins as they become available. The RNA binding protein HuR controls this flexibility in mouse and human skeletal muscle, but the molecular mechanisms governing this process remain poorly characterized. Additionally, studies from mice indicate that HuR control of metabolic flexibility may be more essential for males than females. This is because males lacking HuR in skeletal muscle develop hallmarks of insulin sensitivity, while females have not been shown to do so. Here we examine this sexual dimorphism in mice lacking HuR in skeletal muscle. Our results reveal that lack of HuR in skeletal muscle drives increased adiposity regardless of sex, but that this increase in adiposity drives the development of insulin resistance in male animals only. Additionally, relative to male mice, the detrimental metabolic phenotype associated with HuR inhibition in skeletal muscle can be corrected by feeding of a diet heavily composed of either lipids or carbohydrates. Abstract Male mice lacking HuR in skeletal muscle (HuRm−/−) have been shown to have decreased gastrocnemius lipid oxidation and increased adiposity and insulin resistance. The same consequences have not been documented in female HuRm−/− mice. Here we examine this sexually dimorphic phenotype. HuRm−/− mice have an increased fat mass to lean mass ratio (FM/LM) relative to controls where food intake is similar. Increased body weight for male mice correlates with increased blood glucose during glucose tolerance tests (GTT), suggesting increased fat mass in male HuRm−/− mice as a driver of decreased glucose clearance. However, HuRm−/− female mice show decreased blood glucose levels during GTT relative to controls. HuRm−/− mice display decreased palmitate oxidation in skeletal muscle relative to controls. This difference is more robust for male HuRm−/− mice and more exaggerated for both sexes at high dietary fat. A high-fat diet stimulates expression of Pgc1α in HuRm−/− male skeletal muscle, but not in females. However, the lipid oxidation Pparα pathway remains decreased in HuRm−/− male mice relative to controls regardless of diet. This pathway is only decreased in female HuRm−/− mice fed high fat diet. A decreased capacity for lipid oxidation in skeletal muscle in the absence of HuR may thus be linked to decreased glucose clearance in male but not female mice.
Collapse
|
46
|
Ishihara A, Park I, Suzuki Y, Yajima K, Cui H, Yanagisawa M, Sano T, Kido J, Tokuyama K. Metabolic responses to polychromatic LED and OLED light at night. Sci Rep 2021; 11:12402. [PMID: 34117328 PMCID: PMC8196130 DOI: 10.1038/s41598-021-91828-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023] Open
Abstract
Light exposure at night has various implications for human health, but little is known about its effects on energy metabolism during subsequent sleep. We investigated the effects of polychromatic white light using conventional light-emitting diodes (LED) and an alternative light source, organic light-emitting diodes (OLED), producing reduced spectral content in the short wavelength of blue light (455 nm). Ten male participants were exposed to either LED, OLED (1000 lx), or dim (< 10 lx) light for 4 h before sleep in a metabolic chamber. Following OLED exposure, energy expenditure and core body temperature during sleep were significantly decreased (p < 0.001). Fat oxidation during sleep was significantly reduced (p = 0.001) after the exposure to LED compared with OLED. Following exposure to OLED, fat oxidation positively correlated with the 6-sulfatoxymelatonin levels, suggesting that the role of melatonin in lipolysis differs depending on the light. These findings advance our knowledge regarding the role of light in energy metabolism during sleep and provide a potential alternative to mitigate the negative consequences of light exposure at night.
Collapse
Affiliation(s)
- Asuka Ishihara
- grid.20515.330000 0001 2369 4728International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki Japan ,grid.20515.330000 0001 2369 4728Ph.D. Program in Human Biology, School of Integrative Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Insung Park
- grid.20515.330000 0001 2369 4728International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Yoko Suzuki
- grid.20515.330000 0001 2369 4728International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Katsuhiko Yajima
- grid.411949.00000 0004 1770 2033Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Huiyun Cui
- grid.20515.330000 0001 2369 4728Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Masashi Yanagisawa
- grid.20515.330000 0001 2369 4728International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Takeshi Sano
- grid.268394.20000 0001 0674 7277Innovation Center for Organic Electronics, Yamagata University, Yamagata, Japan
| | - Junji Kido
- grid.268394.20000 0001 0674 7277Graduate School of Organic Materials Science, Yamagata University, Yamagata, Japan
| | - Kumpei Tokuyama
- grid.20515.330000 0001 2369 4728International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki Japan
| |
Collapse
|
47
|
Jain S, Pydi SP, Jung YH, Scortichini M, Kesner EL, Karcz TP, Cook DN, Gavrilova O, Wess J, Jacobson KA. Adipocyte P2Y14 receptors play a key role in regulating whole-body glucose and lipid homeostasis. JCI Insight 2021; 6:146577. [PMID: 34027896 PMCID: PMC8262345 DOI: 10.1172/jci.insight.146577] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/09/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity is the major driver of the worldwide epidemic in type 2 diabetes (T2D). In the obese state, chronically elevated plasma free fatty acid levels contribute to peripheral insulin resistance, which can ultimately lead to the development of T2D. For this reason, drugs that are able to regulate lipolytic processes in adipocytes are predicted to have considerable therapeutic potential. Gi-coupled P2Y14 receptor (P2Y14R; endogenous agonist, UDP-glucose) is abundantly expressed in both mouse and human adipocytes. Because activated Gi-type G proteins exert an antilipolytic effect, we explored the potential physiological relevance of adipocyte P2Y14Rs in regulating lipid and glucose homeostasis. Metabolic studies indicate that the lack of adipocyte P2Y14R enhanced lipolysis only in the fasting state, decreased body weight, and improved glucose tolerance and insulin sensitivity. Mechanistic studies suggested that adipocyte P2Y14R inhibits lipolysis by reducing lipolytic enzyme activity, including ATGL and HSL. In agreement with these findings, agonist treatment of control mice with a P2Y14R agonist decreased lipolysis, an effect that was sensitive to inhibition by a P2Y14R antagonist. In conclusion, we demonstrate that adipose P2Y14Rs were critical regulators of whole-body glucose and lipid homeostasis, suggesting that P2Y14R antagonists might be beneficial for the therapy of obesity and T2D.
Collapse
Affiliation(s)
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | | | | | | | - Tadeusz P Karcz
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Donald N Cook
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | | |
Collapse
|
48
|
Guarnieri AR, Anthony SR, Gozdiff A, Green LC, Fleifil SM, Slone S, Nieman ML, Alam P, Benoit JB, Owens AP, Kanisicak O, Tranter M. Adipocyte-specific deletion of HuR induces spontaneous cardiac hypertrophy and fibrosis. Am J Physiol Heart Circ Physiol 2021; 321:H228-H241. [PMID: 34018851 DOI: 10.1152/ajpheart.00957.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adipose tissue homeostasis plays a central role in cardiovascular physiology, and the presence of thermogenically active brown adipose tissue (BAT) has recently been associated with cardiometabolic health. We have previously shown that adipose tissue-specific deletion of HuR (Adipo-HuR-/-) reduces BAT-mediated adaptive thermogenesis, and the goal of this work was to identify the cardiovascular impacts of Adipo-HuR-/-. We found that Adipo-HuR-/- mice exhibit a hypercontractile phenotype that is accompanied by increased left ventricle wall thickness and hypertrophic gene expression. Furthermore, hearts from Adipo-HuR-/- mice display increased fibrosis via picrosirius red staining and periostin expression. To identify underlying mechanisms, we applied both RNA-seq and weighted gene coexpression network analysis (WGCNA) across both cardiac and adipose tissue to define HuR-dependent changes in gene expression as well as significant relationships between adipose tissue gene expression and cardiac fibrosis. RNA-seq results demonstrated a significant increase in proinflammatory gene expression in both cardiac and subcutaneous white adipose tissue (scWAT) from Adipo-HuR-/- mice that is accompanied by an increase in serum levels of both TNF-α and IL-6. In addition to inflammation-related genes, WGCNA identified a significant enrichment in extracellular vesicle-mediated transport and exosome-associated genes in scWAT, whose expression most significantly associated with the degree of cardiac fibrosis observed in Adipo-HuR-/- mice, implicating these processes as a likely adipose-to-cardiac paracrine mechanism. These results are significant in that they demonstrate the spontaneous onset of cardiovascular pathology in an adipose tissue-specific gene deletion model and contribute to our understanding of how disruptions in adipose tissue homeostasis may mediate cardiovascular disease.NEW & NOTEWORTHY The presence of functional brown adipose tissue in humans is known to be associated with cardiovascular health. Here, we show that adipocyte-specific deletion of the RNA binding protein HuR, which we have previously shown to reduce BAT-mediated thermogenesis, is sufficient to mediate a spontaneous development of cardiac hypertrophy and fibrosis. These results may have implications on the mechanisms by which BAT function and adipose tissue homeostasis directly mediate cardiovascular disease.
Collapse
Affiliation(s)
- Adrienne R Guarnieri
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Sarah R Anthony
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Anamarie Gozdiff
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa C Green
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Salma M Fleifil
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Sam Slone
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michelle L Nieman
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Perwez Alam
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio
| | - A Phillip Owens
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael Tranter
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
49
|
Smooth muscle-specific HuR knockout induces defective autophagy and atherosclerosis. Cell Death Dis 2021; 12:385. [PMID: 33837179 PMCID: PMC8035143 DOI: 10.1038/s41419-021-03671-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 01/19/2023]
Abstract
Human antigen R (HuR) is a widespread RNA-binding protein involved in homeostatic regulation and pathological processes in many diseases. Atherosclerosis is the leading cause of cardiovascular disease and acute cardiovascular events. However, the role of HuR in atherosclerosis remains unknown. In this study, mice with smooth muscle-specific HuR knockout (HuRSMKO) were generated to investigate the role of HuR in atherosclerosis. HuR expression was reduced in atherosclerotic plaques. As compared with controls, HuRSMKO mice showed increased plaque burden in the atherosclerotic model. Mechanically, HuR could bind to the mRNAs of adenosine 5′-monophosphate-activated protein kinase (AMPK) α1 and AMPKα2, thus increasing their stability and translation. HuR deficiency reduced p-AMPK and LC3II levels and increased p62 level, thereby resulting in defective autophagy. Finally, pharmacological AMPK activation induced autophagy and suppressed atherosclerosis in HuRSMKO mice. Our findings suggest that smooth muscle HuR has a protective effect against atherosclerosis by increasing AMPK-mediated autophagy.
Collapse
|
50
|
Tian M, Wang J, Liu S, Li X, Li J, Yang J, Zhang C, Zhang W. Hepatic HuR protects against the pathogenesis of non-alcoholic fatty liver disease by targeting PTEN. Cell Death Dis 2021; 12:236. [PMID: 33664225 PMCID: PMC7933173 DOI: 10.1038/s41419-021-03514-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/22/2022]
Abstract
The liver plays an important role in lipid and glucose metabolism. Here, we show the role of human antigen R (HuR), an RNA regulator protein, in hepatocyte steatosis and glucose metabolism. We investigated the level of HuR in the liver of mice fed a normal chow diet (NCD) and a high-fat diet (HFD). HuR was downregulated in the livers of HFD-fed mice. Liver-specific HuR knockout (HuRLKO) mice showed exacerbated HFD-induced hepatic steatosis along with enhanced glucose tolerance as compared with control mice. Mechanistically, HuR could bind to the adenylate uridylate-rich elements of phosphatase and tensin homolog deleted on the chromosome 10 (PTEN) mRNA 3' untranslated region, resulting in the increased stability of Pten mRNA; genetic knockdown of HuR decreased the expression of PTEN. Finally, lentiviral overexpression of PTEN alleviated the development of hepatic steatosis in HuRLKO mice in vivo. Overall, HuR regulates lipid and glucose metabolism by targeting PTEN.
Collapse
Affiliation(s)
- Mi Tian
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Jingjing Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Shandong, China
| | - Shangming Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Shandong, China
| | - Xinyun Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingyuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wencheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Shandong, China.
| |
Collapse
|