1
|
Sun S, Arif Aslam M, Ma EB, Lee G, Muhammad Ahmad Javaid H, Yoon S, Huh JY. Activation of CXCR7 exerts an inhibitory effect on adipogenesis through regulation of β-arrestin2/Wnt and AKT signalling. Adipocyte 2025; 14:2490258. [PMID: 40302245 PMCID: PMC12045560 DOI: 10.1080/21623945.2025.2490258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/22/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
CXCR7, an alternative receptor for the inflammatory chemokine SDF-1, is involved in cell proliferation and migration. Recent studies have reported that CXCR7 also plays a role in adipose tissue. However, evidence regarding the role of CXCR7 and its ligands in adipocyte differentiation is limited. In this study, we aimed to elucidate changes in CXCR7 expression during adipocyte differentiation and the role of the SDF-1/CXCR7/CXCR4 axis in adipogenesis using recombinant SDF-1, the CXCR7 ligand CCX771, and small interfering RNAs. The results indicated that the levels of SDF-1 and its receptors, CXCR7 and CXCR4, decreased during the early stages of adipogenesis. Treatment with recombinant SDF-1 and CCX771 inhibited adipogenesis and lipid accumulation by inducing β-arrestin2, Wnt expression, and AKT phosphorylation and downregulating C/EBPα, PPARγ, and FABP4 expression. In contrast, knockdown of SDF-1 and CXCR7 in preadipocytes downregulated the β-arrestin2/Wnt and AKT pathway, leading to the induction of adipogenesis. Meanwhile, knockdown of CXCR4 had no significant effect. In mice, basal gene expression levels of SDF-1 and CXCR7 were higher in the stromal vascular fraction compared to mature adipocytes and were significantly upregulated by a high-fat diet. Our results provide new insights into the local role of the SDF-1-CXCR7 axis in adipocytes and offer additional benefits for the prevention of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Shiyue Sun
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Gahui Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hafiz Muhammad Ahmad Javaid
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Somy Yoon
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Xiaoyang C, Yijun C, Chenguang Z, Wanying D, Zijun C, Jun W, Xuegong X, Wei W, Chun L. Resibufogenin protects against atherosclerosis in ApoE -/- mice through blocking NLRP3 inflammasome assembly. J Adv Res 2025:S2090-1232(25)00272-3. [PMID: 40258472 DOI: 10.1016/j.jare.2025.04.029] [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: 01/11/2025] [Revised: 04/01/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025] Open
Abstract
INTRODUCTION Atherosclerosis (AS), a major cause of cardiovascular diseases, is characterized by lipid accumulation and chronic inflammation within arterial walls. Traditional treatments, such as statins, are often ineffective for many patients, highlighting the need for novel therapeutic strategies. OBJECTIVE This study explores the potential of Resibufogenin (RBG) as an NLRP3 inflammasome inhibitor for treating AS in ApoE-/- mice. METHODS We performed experiments encompassing cellular studies, animal model assessments, molecular simulations, and binding assays to assess RBG's impact on the NLRP3 inflammasome, inflammatory cytokine release, and foam cell formation. RESULTS RBG treatment alleviated AS in ApoE-/- mice, evidenced by reduced body weight, smaller atherosclerotic plaques, and improved serum lipid profiles. Transcriptomics and molecular biology demonstrated that RBG suppressed the expression of key inflammatory markers such as NLRP3. RBG also reduced macrophage infiltration and promoted polarization toward the anti-inflammatory M2 phenotype. Molecular docking, SPR, Pull-down studies identified a non-covalent interaction between RBG and the CYS-279 residue of NLRP3, confirming its role as a potent NLRP3 inhibitor. CONCLUSION RBG effectively inhibits NLRP3 inflammasome activation, reduces pro-inflammatory cytokine release, and decreases formation of foamy macrophages, thereby slowing the progression of AS. Although these findings highlight RBG as a promising therapeutic approach for cardiovascular diseases, further research is necessary to assess its safety and effectiveness in humans and to investigate possible synergistic effects with other treatments.
Collapse
Affiliation(s)
- Chen Xiaoyang
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chen Yijun
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhai Chenguang
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Du Wanying
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chen Zijun
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Wang Jun
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xu Xuegong
- Zhengzhou Hospital of TCM Affiliated to Henan University of Chinese Medicine(Zhengzhou Hospital of Traditional Chinese Medicine), Zhengzhou 450007, China; Institute of Geriatric Diseases, Henan Academy of Chinese Medical Sciences, Zhengzhou 451100, China.
| | - Wang Wei
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Li Chun
- State Key Laboratory of Traditional Chinese Medicine Syndrome; School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Formula and Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
3
|
Poulsen AS, Stisen ZR, Skougaard M, Christensen R, Overgaard A, Gudbergsen H, Jacobsen S, Balslev-Clausen AP, Henriksen M, Kristensen LE, Bliddal H. Effect of weight loss and liraglutide on neutrophil gelatinase-associated lipocalin levels among individuals with overweight and knee osteoarthritis: Exploratory analyses of a randomized controlled trial. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100562. [PMID: 39877802 PMCID: PMC11773487 DOI: 10.1016/j.ocarto.2024.100562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Objective Obesity is a major risk factor for osteoarthritis (OA). Adipose tissues may be linked to OA development through secretion of potential proinflammatory cytokines including neutrophil gelatinase-associated lipocalin (NGAL). Our objective was to assess changes in serum NGAL after a low-calorie diet (LCD) and subsequent glucagon-like peptide 1 receptor agonist (GLP-1 RA) treatment. Design A secondary analysis of a randomized, double-blinded, placebo-controlled trial in adults with overweight (BMI≥27 kg/m2) and symptomatic, early-to-moderate radiographic knee OA. Prior to randomization, participants underwent an 8-week LCD (week -8 to 0). Participants who lost min. 5 % of initial bodyweight were randomized 1:1 to liraglutide 3 mg/d or placebo for 52 weeks. Main outcome was change in serum NGAL from enrollment (week -8) to randomization (week 0). Other outcome was change in serum NGAL from week 0 to week 52 comparing liraglutide and placebo. Results 168 participants were enrolled to the initial intensive diet intervention; 127 participants, with NGAL samples, were randomized. Following the 8-week diet intervention, NGAL concentrations rose by 93.0 ng/mL (95 % CI: 18.9 to 167.1, P = 0.015), with no correlation to weight loss magnitude. 52 weeks of treatment with either liraglutide or placebo, liraglutide did not cause a greater decrease in serum NGAL (14.9 ng/ml, 95%CI: -92.1 to 121.7 ng/mL, P = 0.78). Conclusion An intensive 8-week calorie restriction was associated with a rise in serum NGAL. Compared to placebo, 52 weeks of liraglutide did not cause additional changes in NGAL. This indicates a complex pattern of proinflammatory cytokine-release during hypocaloric diet interventions. Trial registration Clinicaltrials.gov: NCT02905864.
Collapse
Affiliation(s)
- Asbjørn Seenithamby Poulsen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Zara Rebecca Stisen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
| | - Marie Skougaard
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Copenhagen Center of Translational Research, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Denmark
| | - Robin Christensen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Anders Overgaard
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
| | - Henrik Gudbergsen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Center for General Practice, Department of Public Health, University of Copenhagen, Denmark
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Denmark
| | | | - Marius Henriksen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Lars Erik Kristensen
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
| | - Henning Bliddal
- The Parker Institute, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
4
|
Yan L, Guo L. The role and mechanism of m6A methylation in diabetic nephropathy. Life Sci 2025; 363:123355. [PMID: 39778764 DOI: 10.1016/j.lfs.2024.123355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/19/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications of diabetes mellitus, characterized by progressive deterioration of renal structure and function, which may eventually lead to end-stage kidney disease (ESKD). The N6-methyladenosine (m6A) methylation, an important modality of RNA modification, involves three classes of key regulators, writers (e.g., METTL3), erasers (e.g., FTO, ALKBH5) and readers (e.g., YTHDF2), which play important roles in DN. Writers are responsible for introducing m6A modifications on RNAs, erasers remove m6A modifications and readers recognize and bind m6A-modified RNAs to regulate RNAs functions, such as mRNA stability, translation and localization. In DN, abnormal m6A modification may promote kidney injury and proteinuria by regulating key pathways involved in multiple processes, including lipid metabolism and inflammatory response, in kidney cells such as podocytes. Therefore, an in-depth study of the role and mechanism of m6A methylation that are regulated by "writers", "erasers" and "readers" in DN is expected to provide new targets and strategies for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Linjing Yan
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, PR China
| | - Liang Guo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, PR China.
| |
Collapse
|
5
|
Zhang B, Zeng M, Tie Q, Wang R, Wang M, Wu Y, Zheng X, Feng W. (-)-Epigallocatechin-3-gallate (EGCG) ameliorates ovalbumin-induced asthma by inhibiting inflammation via the TNF-α/TNF-R1/NLRP3 signaling pathway. Int Immunopharmacol 2025; 144:113708. [PMID: 39626539 DOI: 10.1016/j.intimp.2024.113708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024]
Abstract
(-)-Epigallocatechin-3-gallate (EGCG) is a polyphenol in green tea with potential lung-protective effects. However, the effects of EGCG on airway inflammation in asthma remain unclear. The aim of this study was to investigate the effect and mechanism of EGCG on asthmatic airway inflammation. In this study, the therapeutic effects of EGCG on ovalbumin (OVA)-induced asthmatic mice were tested first. Second, the effects of EGCG on airway inflammation, airway hyperresponsiveness (AHR), airway mucus hypersecretion, cell apoptosis and differential genes were investigated. Finally, the relationships between the effects of EGCG on airway inflammation and the TNF-α/TNF-R1/NLRP3 signaling pathway in asthmatic mice were explored. The results showed that EGCG could attenuate AHR, alleviate the symptoms of alveolar wall thickening and inflammatory cell infiltration, decrease the levels of inflammatory cytokines and airway mucus markers, reduce apoptosis and reactive oxygen species (ROS) and increase the mitochondrial membrane potential (MMP) in primary lung cells in asthmatic mice. Additionally, EGCG significantly inhibited the activation of the TNF-α/TNF-R1/NLRP3 signaling pathway in the lung tissues of asthmatic mice. The lowest binding free energies of EGCG with TNF-α, TNF-R1 and NLRP3 were -11.6, -11.6 and -8.2 kcal/mol, respectively. Moreover, the equilibrium dissociation constant (KD) of EGCG and TNF-R1was 26.05 μmol/L. EGCG-mediated inhibition of TNF-α/TNF-R1/NLRP3 signaling pathway activation was blocked in LPS-induced BEAS-2B and RAW264.7 cells overexpressing TNF-α. Consequently, EGCG effectively attenuated AHR and inhibited airway inflammation and airway mucus hypersecretion in asthmatic mice, and these effects may be closely related to the TNF-α/TNF-R1/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Beibei Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Mengnan Zeng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qimei Tie
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ru Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengya Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yuanyuan Wu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Weisheng Feng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
6
|
Zhang ZX, Peng J, Ding WW. Lipocalin-2 and intestinal diseases. World J Gastroenterol 2024; 30:4864-4879. [PMID: 39679305 PMCID: PMC11612708 DOI: 10.3748/wjg.v30.i46.4864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Dysfunction of the intestinal barrier is a prevalent phenomenon observed across a spectrum of diseases, encompassing conditions such as mesenteric artery dissection, inflammatory bowel disease, cirrhosis, and sepsis. In these pathological states, the integrity of the intestinal barrier, which normally serves to regulate the selective passage of substances between the gut lumen and the bloodstream, becomes compromised. This compromised barrier function can lead to a range of adverse consequences, including increased permeability to harmful substances, the translocation of bacteria and their products into systemic circulation, and heightened inflammatory responses within the gut and beyond. Understanding the mechanisms underlying intestinal barrier dysfunction in these diverse disease contexts is crucial for the development of targeted therapeutic interventions aimed at restoring barrier integrity and ameliorating disease progression. Lipocalin-2 (LCN2) expression is significantly upregulated during episodes of intestinal inflammation, making it a pivotal indicator for gauging the extent of such inflammatory processes. Notably, however, LCN2 derived from distinct cellular sources, whether intestinal epithelial cells or immune cells, exhibits notably divergent functional characteristics. Furthermore, the multifaceted nature of LCN2 is underscored by its varying roles across different diseases, sometimes even demonstrating contradictory effects.
Collapse
Affiliation(s)
- Zhong-Xu Zhang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Jian Peng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei-Wei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
7
|
Wang G, Liu S, Kong X, Jiao H, Tong F, Guo Z, Zhang M, Guan X, Ren N, Li W, Qi L, Wei Y. Lipocalin-2 induced LDHA expression promotes vascular remodelling in pulmonary hypertension. Cell Prolif 2024; 57:e13717. [PMID: 39021353 PMCID: PMC11628741 DOI: 10.1111/cpr.13717] [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: 05/13/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
Aerobic glycolysis is involved in the pathogenesis of pulmonary hypertension (PH). The mechanisms by which glycolysis is increased and how it contributes to pulmonary vascular remodelling are not yet fully understood. In this study, we demonstrated that elevated lipocalin-2 (LCN2) in PH significantly enhances aerobic glycolysis in human pulmonary artery smooth muscle cells (PASMCs) by up-regulating LDHA expression. Knockout of Lcn2 or having heterozygous LDHA deficiency in mice significantly inhibits the progression of hypoxic PH. Our study reveals that LCN2 stimulates LDHA expression by activating Akt-HIF-1α signalling pathway. Inhibition of Akt or HIF-1α reduces LDHA expression and proliferation of PASMCs. Both Akt and HIF-1α play critical roles in the development of PH and are suppressed in the pulmonary vessels of hypoxic PH mice lacking LCN2. These findings shed light on the LCN2-Akt-HIF1α-LDHA axis in aerobic glycolysis in PH.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaohui Kong
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Hong Jiao
- Department of Tumor and Immunology, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Feng Tong
- Department of Cardiac Surgery, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Zhangke Guo
- Department of Cardiac Surgery, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Meng Zhang
- Department of Pathology, Beijing Children's Hospital, Capital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Xiaoxing Guan
- Department of Pathology, Beijing Children's Hospital, Capital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Na Ren
- Department of Clinical Laboratory Center, Beijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Wanzhen Li
- Department of Lipidomics Experimental Platform, State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental BiologyChinese Academy of SciencesBeijingChina
| | - Lihua Qi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
8
|
Ren Y, Huang P, Zhang L, Tang Y, He S, Li H, Huang X, Ding Y, Liu L, Liu L, He X. Multi-omics landscape of childhood simple obesity: novel insights into pathogenesis and biomarkers discovery. Cell Biosci 2024; 14:145. [PMID: 39609876 PMCID: PMC11606102 DOI: 10.1186/s13578-024-01322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The increasing incidence of childhood obesity annually has led to a surge in physical and mental health risks, making it a significant global public health concern. This study aimed to discover novel biomarkers of childhood simple obesity through integrative multi-omics analysis, uncovering their potential connections and providing fresh research directions for the complex pathogenesis and treatment strategies. METHODS Transcriptome, untargeted metabolome, and 16 S rDNA sequencing were conducted on subjects to examine transcripts, metabolites in blood, and gut microflora in stool. RESULTS Transcriptomic analysis identified 599 differentially expressed genes (DEGs), of which 25 were immune-related genes, and participated in immune pathways such as antimicrobial peptides, neutrophil degranulation, and interferons. The optimal random forest model based on these genes exhibited an AUC of 0.844. The metabolomic analysis examined 71 differentially expressed metabolites (DEMs), including 12 immune-related metabolites. Notably, lauric acid showed an extremely strong positive correlation with BMI and showed a good discriminative power for obesity (AUC = 0.82). DEMs were found to be significantly enriched in four metabolic pathways, namely "Aminoacyl-tRNA biosynthesis", "Valine leucine and isoleucine biosynthesis, and Glycine", "Serine and threonine metabolism", and "Biosynthesis of unsaturated fatty acids". Microbiome analysis revealed 12 differential gut microbiotas (DGMs) at the phylum and genus levels, with p_Firmicutes dominating in the obese group and g_Escherichia-Shigella in the normal group. Subsequently, a Random Forest model was developed based on the DEMs, immune-related DEGs, and metabolites with an AUC value of 0.912. The 14 indicators identified by this model could potentially serve as a set of biomarkers for obesity. The analysis of the inter-omics correlation network found 233 pairs of significant correlations. DEGs BPIFA1, BPI, and SAA1, DEMs Dimethy(tetradecyl)amine, Deoxycholic acid, Pathalic anhydride, and DL-Alanine, and DGMs g_Intestinimonas and g_Turicibacter showed strong connectivity within the network, constituting a large proportion of interactions. CONCLUSION This study presents the first comprehensive description of the multi-omics characteristics of childhood simple obesity, recognizing promising biomarkers. Immune-related markers offer a new perspective for researching the immunological mechanisms underlying obesity and its associated complications. The revealed interactions among these biomarkers contribute to a deeper understanding the intricate biological regulatory networks associated with obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, 570100, China
- Department of Pediatrics, Hainan Modern Women and Children's. Medical, Haikou, 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan, 570311, China
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - HaiDan Li
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - XiaoYan Huang
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - Yan Ding
- Department of Dermatology, Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Laboratory of Pediatric Nephrology, Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
9
|
Lai M, Lin K, Chen X, Cheng Y. Diverse Cytokines Secreted by Adipocyte in Linking Cardio-Metabolic Disorder and SLE. FRONT BIOSCI-LANDMRK 2024; 29:373. [PMID: 39614444 DOI: 10.31083/j.fbl2911373] [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: 07/25/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 12/01/2024]
Abstract
Systemic lupus erythematosus (SLE) is a multi-factorial autoimmune-mediated disease with hyper-stimulation of immune cells especially the T lymphocytes. By this method, it might facilitate the systematic damages in multiple tissues and organs. Otherwise, SLE is also correlated with diverse cardio-metabolic comorbidities, including dyslipidemia, insulin resistance, and hypertension. It is worth-noting that the risk of cardio-metabolic disorders is significantly higher compared with the healthy patients which was reported as approximately one-third of SLE patients were proved as obesity. Notably, current focus is shifting to implementing cardio-metabolic protective strategies as well as elucidating underlying mechanisms of lupus-mediated obese status. On the other hand, adipocyte, as the most abundant endocrine cell in fat tissue, are dysfunctional in obese individuals with aberrant secretion of adipokines. It is proposing that the adipokine might link the pathology of cardio-metabolic disorders and SLE, whereas the related mechanism is complicated. In the current review, the functions of adipokine and the potential mechanisms by which the adipokine link cardio-metabolic disorders and SLE was well listed. Furthermore, the recommendations, which identify the adipokine as the potential therapeutic targets for the treatment of cardio-metabolic disorders and SLE, were also summarized.
Collapse
Affiliation(s)
- Min Lai
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, 361000 Xiamen, Fujian, China
| | - Kai Lin
- Department of Interventional Clinic, The Xiamen Cardiovascular Hospital of Xiamen University, 361000 Xiamen, Fujian, China
| | - Xiaofang Chen
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, 361000 Xiamen, Fujian, China
| | - Ye Cheng
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, 361000 Xiamen, Fujian, China
| |
Collapse
|
10
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
11
|
Meng X, Li W, Qian Y, Cai X, Wei J, Zhang L. Mechanisms of colon toxicity induced by long-term perfluorooctanoic acid exposure in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116762. [PMID: 39047366 DOI: 10.1016/j.ecoenv.2024.116762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Perfluorooctanoic acid (PFOA), a persistent organic pollutant known for its chemical stability, is widely dispersed in the environment, posing significant health risks to mammals through various exposure routes such as ingestion, inhalation, and dermal contact. In this study, mice were exposed to PFOA (0, 0.2, 2 mg/L) through drinking water for 180 days to investigate its toxic effects on the colon. We identified differentially expressed genes through RNA sequencing and validated the impact of PFOA on the expression of these genes in colon tissue. Our findings revealed that long-term exposure to PFOA caused inflammatory bowel disease (IBD)-like damage to the mouse colon. We found PFOA could induce damage to the intestinal barrier. Inhibition of the Wnt signaling pathway following PFOA exposure results in impaired stem cell function in the colon of mice. Furthermore, PFOA activated the PPAR signaling pathway, disrupting cellular lipid metabolism in colon tissues. Additionally, PFOA induced inflammatory responses in colon tissue, facilitating NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation and cell apoptosis. This study offers a thorough understanding of the mechanisms responsible for the damage to mouse colon tissue resulting from long-term exposure to PFOA.
Collapse
Affiliation(s)
- Xiannan Meng
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Wei Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Yongjing Qian
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Xiaojing Cai
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Jianfeng Wei
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Ling Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
12
|
Ma C, Gou C, Sun S, Wang J, Wei X, Xing F, Xing N, Yuan J, Wang Z. Unraveling the molecular complexity: Wtap/Ythdf1 and Lcn2 in novel traumatic brain injury secondary injury mechanisms. Cell Biol Toxicol 2024; 40:65. [PMID: 39110292 PMCID: PMC11306654 DOI: 10.1007/s10565-024-09909-x] [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: 01/24/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024]
Abstract
The primary aim of this research was to explore the functions of Wtap and Ythdf1 in regulating neuronal Lipocalin-2 (Lcn2) through m6A modification in traumatic brain injury (TBI). By employing transcriptome sequencing and enrichment analysis, we identified the Wtap/Ythdf1-mediated Lcn2 m6A modification pathway as crucial in TBI. In our in vitro experiments using primary cortical neurons, knockout of Wtap and Ythdf1 led to the inhibition of Lcn2 m6A modification, resulting in reduced neuronal death and inflammation. Furthermore, overexpression of Lcn2 in cortical neurons induced the activation of reactive astrocytes and M1-like microglial cells, causing neuronal apoptosis. In vivo experiments confirmed the activation of reactive astrocytes and microglial cells in TBI and importantly demonstrated that Wtap knockdown improved neuroinflammation and functional impairment. These findings underscore the significance of Wtap/Ythdf1-mediated Lcn2 regulation in TBI secondary injury and suggest potential therapeutic implications for combating TBI-induced neuroinflammation and neuronal damage.
Collapse
Affiliation(s)
- Chaobang Ma
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China
| | - Caili Gou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Shiyu Sun
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China
| | - Junmin Wang
- Department of Human Anatomy Basic Medical College of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xin Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Jingjing Yuan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Zhongyu Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450052, Henan, China.
- Department of Human Anatomy Basic Medical College of Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
13
|
Lu F, Li E, Yang X. Proprotein convertase subtilisin/kexin type 9 deficiency in extrahepatic tissues: emerging considerations. Front Pharmacol 2024; 15:1413123. [PMID: 39139638 PMCID: PMC11319175 DOI: 10.3389/fphar.2024.1413123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily secreted by hepatocytes. PCSK9 is critical in liver low-density lipoprotein receptors (LDLRs) metabolism. In addition to its hepatocellular presence, PCSK9 has also been detected in cardiac, cerebral, islet, renal, adipose, and other tissues. Once perceived primarily as a "harmful factor," PCSK9 has been a focal point for the targeted inhibition of both systemic circulation and localized tissues to treat diseases. However, PCSK9 also contributes to the maintenance of normal physiological functions in numerous extrahepatic tissues, encompassing both LDLR-dependent and -independent pathways. Consequently, PCSK9 deficiency may harm extrahepatic tissues in close association with several pathophysiological processes, such as lipid accumulation, mitochondrial impairment, insulin resistance, and abnormal neural differentiation. This review encapsulates the beneficial effects of PCSK9 on the physiological processes and potential disorders arising from PCSK9 deficiency in extrahepatic tissues. This review also provides a comprehensive analysis of the disparities between experimental and clinical research findings regarding the potential harm associated with PCSK9 deficiency. The aim is to improve the current understanding of the diverse effects of PCSK9 inhibition.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Yu W, Zhang Y, Sun L, Huang W, Li X, Xia N, Chen X, Wikana LP, Xiao Y, Chen M, Han S, Wang Z, Pu L. Myeloid Trem2 ameliorates the progression of metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. Metabolism 2024; 155:155911. [PMID: 38609037 DOI: 10.1016/j.metabol.2024.155911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFβ1, thereby promoting tissue repair. CONCLUSIONS Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.
Collapse
Affiliation(s)
- Wenjie Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Yu Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Linfeng Sun
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Wei Huang
- Department of General Surgery, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Xiangdong Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Nan Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Xuejiao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Likalamu Pascalia Wikana
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Yuhao Xiao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Minhao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Sheng Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Ziyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
15
|
Rosendo-Silva D, Gomes PB, Rodrigues T, Viana S, da Costa AN, Scherer PE, Reis F, Pereira F, Seiça R, Matafome P. Clinical and molecular profiling of human visceral adipose tissue reveals impairment of vascular architecture and remodeling as an early hallmark of dysfunction. Metabolism 2024; 153:155788. [PMID: 38219974 DOI: 10.1016/j.metabol.2024.155788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Adipose tissue dysfunction is more related to insulin resistance than body mass index itself and an alteration in adipose tissue function is thought to underlie the shift from metabolically healthy to unhealthy obesity. Herein, we performed a clustering analysis that revealed distinct visceral adipose tissue gene expression patterns in patients with obesity at distinct stages of metabolic dysregulation. We have built a cross-sectional cohort that aims at reflecting the evolution of the metabolic sequelae of obesity with the main objective to map the sequential events that play a role in adipose tissue dysfunction from the metabolically healthy (insulin-sensitive) state to several incremental degrees of metabolic dysregulation, encompassing insulin resistance establishment, pre-diabetes, and type 2 diabetes. We found that insulin resistance is mainly marked by the downregulation of adipose tissue vasculature remodeling-associated gene expression, suggesting that processes like angiogenesis and adaptative expansion/retraction ability suffer early dysregulation. Prediabetes was characterized by compensatory growth factor-dependent signaling and increased response to hypoxia, while type 2 diabetes was associated with loss of cellular response to insulin and hypoxia and concomitant upregulation of inflammatory markers. Our findings suggest a putative sequence of dysregulation of biological processes that is not linear and has multiple distinct phases across the metabolic dysregulation process, ultimately culminating in the climax of adipose tissue dysfunction in type 2 diabetes. Several studies have addressed the transcriptomic changes in adipose tissue of patients with obesity. However, to the best of our knowledge, this is the first study unraveling the potential molecular mechanisms associated with the multi-step evolution of adipose tissue dysfunction along the metabolic sequelae of obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Pedro Bastos Gomes
- Department of Surgery, Universitary Hospital Center of Coimbra, Portugal
| | - Tiago Rodrigues
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - André Nogueira da Costa
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Translational Medicine, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Flávio Reis
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Francisco Pereira
- Polytechnic University of Coimbra, Coimbra Institute of Engineering, Coimbra, Portugal; Centre for Informatics and Systems of the University of Coimbra (CISUC), University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
16
|
Susca N, Leone P, Prete M, Cozzio S, Racanelli V. Adipose failure through adipocyte overload and autoimmunity. Autoimmun Rev 2024; 23:103502. [PMID: 38101692 DOI: 10.1016/j.autrev.2023.103502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
Metabolic syndrome poses a great worldwide threat to the health of the patients. Increased visceral adiposity is recognized as the main determinant of the detrimental clinical effects of insulin resistance. Inflammation and immune system activation in the adipose tissue (AT) have a central role in the pathophysiology of metabolic syndrome, but the mechanisms linking increased adiposity to immunity in the AT remain in part elusive. In this review, we support the central role of adipocyte overload and relative adipose failure as key determinants in triggering immune aggression to AT. This provides a mechanistic explanation of the relative metabolic wellness of metabolically normal obese people and the disruption in insulin signaling in metabolically obese lean people.
Collapse
Affiliation(s)
- Nicola Susca
- Department of Interdisciplinary Medicine, School of Medicine, 'Aldo Moro' University of Bari, 70124 Bari, Italy
| | - Patrizia Leone
- Department of Interdisciplinary Medicine, School of Medicine, 'Aldo Moro' University of Bari, 70124 Bari, Italy
| | - Marcella Prete
- Department of Interdisciplinary Medicine, School of Medicine, 'Aldo Moro' University of Bari, 70124 Bari, Italy
| | - Susanna Cozzio
- U.O. di Medicina Interna, Ospedale di Rovereto, Azienda Sanitaria per i Servizi Provinciali di Trento, Trento, Italy
| | - Vito Racanelli
- Centre for Medical Sciences - CISMed, University of Trento and Department of Internal Medicine, Santa Chiara Hospital, Trento, Italy.
| |
Collapse
|
17
|
Cao J, Li L, Zhang R, Shu Z, Zhang Y, Sun W, Zhang Y, Hu Z. Libertellenone C attenuates oxidative stress and neuroinflammation with the capacity of NLRP3 inhibition. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:17. [PMID: 38407685 PMCID: PMC10897105 DOI: 10.1007/s13659-024-00438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/01/2024] [Indexed: 02/27/2024]
Abstract
Neurodegenerative diseases (NDs) are common chronic diseases arising from progressive damage to the nervous system. Here, in-house natural product database screening revealed that libertellenone C (LC) obtained from the fermentation products of Arthrinium arundinis separated from the gut of a centipede collected in our Tongji campus, showed a remarkable neuroprotective effect. Further investigation was conducted to clarify the specific mechanism. LC dose-dependently reversed glutamate-induced decreased viability, accumulated reactive oxygen species, mitochondrial membrane potential loss, and apoptosis in SH-SY5Y cells. Network pharmacology analysis predicted that the targets of LC were most likely directly related to oxidative stress and the regulation of inflammatory factor-associated signaling pathways. Further study demonstrated that LC attenuated nitrite, TNF-α, and IL-1β production and decreased inducible nitric oxide synthase and cyclooxygenase expression in lipopolysaccharide-induced BV-2 cells. LC could directly inhibit NLRP3 inflammasome activation by decreasing the expression levels of NLRP3, ASC, cleaved Caspase-1, and NF-κB p65. Our results provide a new understanding of how LC inhibits the NLRP3 inflammasome in microglia, providing neuroprotection. These findings might guide the development of effective LC-based therapeutic strategies for NDs.
Collapse
Affiliation(s)
- Jie Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lanqin Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Runge Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhou Shu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yaxin Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Weiguang Sun
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhengxi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
18
|
Margalit Grigg L, Abu Shrkihe B, Efimova I, Solodeev I, Shteingard Y, Shani N, Zvibel I, Varol C. NLRP3 Deficiency in Nonimmune Cells Averts Obesity-Induced Fatty Liver Disease. J Transl Med 2024; 104:100308. [PMID: 38135154 DOI: 10.1016/j.labinv.2023.100308] [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: 08/03/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Obesity predisposes to metabolic dysfunction-associated fatty liver disease (MAFLD), cardiovascular disease, and type 2 diabetes. Accumulating evidence suggests a complex role of NLR family pyrin domain containing 3 (NLRP3) inflammasome function in multiple manifestations of the metabolic syndrome, with contradictory results. Its broad expression and pleiotropic functions during obesity led us to investigate the contribution of its expression in nonimmune versus immune cells to the development of obesity and MAFLD. Bone marrow chimerism was used to target NLRP3 deficiency to immune (ImmuneΔNlrp3) versus nonimmune (NonimmuneΔNlrp3) cells. Irradiated WT mice reconstituted with WT bone marrow served as controls. Mice were fed a 60% high-fat diet for 16 weeks. NonimmuneΔNlrp3 mice gained less weight and displayed reduced liver and epididymal white adipose tissue (epiWAT) mass. They also exhibited reduced adipocyte hypertrophy and increased epiWAT adipogenesis and lipolysis. Notable was the diminished hepatic steatosis in NonimmuneΔNlrp3 livers, which persisted even following equilibration of their body weight to that of the control. This was accompanied by a decline in liver triglycerides and in expression of transcriptional modules involved with lipid uptake, storage, and de novo lipogenesis. Thermogenic pathways in brown adipose tissue were comparable to control mice, but an elevation was observed in the genes encoding for lipid transporters and fatty acid oxidation. In contrast, deletion of NLRP3 in the immune cell compartment had limited effects on obesity and hepatic steatosis. Collectively, our results outline a prominent role for NLRP3 in nonimmune cells in facilitating MAFLD during constant energy surplus.
Collapse
Affiliation(s)
- Lilah Margalit Grigg
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel; The Microsurgery and Plastic Surgery Lab, Department of Plastic and Reconstructive Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Bander Abu Shrkihe
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel; Department of Clinical Microbiology and Immunology, School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Irina Efimova
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel; Department of Clinical Microbiology and Immunology, School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Inna Solodeev
- The Microsurgery and Plastic Surgery Lab, Department of Plastic and Reconstructive Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Yuval Shteingard
- Department of Pathology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Nir Shani
- The Microsurgery and Plastic Surgery Lab, Department of Plastic and Reconstructive Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Isabel Zvibel
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel; Department of Clinical Microbiology and Immunology, School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
19
|
Pries R, Kosyna FK, Depping R, Plötze-Martin K, Lange C, Meyhöfer S, Meyhöfer SM, Marquardt JU, Bruchhage KL, Steffen A. Distinguishing the impact of distinct obstructive sleep apnea syndrome (OSAS) and obesity related factors on human monocyte subsets. Sci Rep 2024; 14:340. [PMID: 38172514 PMCID: PMC10764945 DOI: 10.1038/s41598-023-49921-5] [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: 04/18/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) and obesity go hand in hand in the majority of patients and both are associated with a systemic inflammation, immune disturbance and comorbidities such as cardiovascular disease. However, the unambiguous impact of OSAS and obesity on the individual inflammatory microenvironment and the immunological consequences of human monocytes has not been distinguished yet. Therefore, aim of this study was to investigate the impact of OSAS and obesity related factors on the inflammatory microenvironment by performing flow cytometric whole blood measurements of CD14/CD16 monocyte subsets in normal weight OSAS patients, patients with obesity but without OSAS, and patients with OSAS and obesity, compared to healthy donors. Moreover, explicitly OSAS and obesity related plasma levels of inflammatory mediators adiponectin, leptin, lipocalin and metalloproteinase-9 were determined and the influence of different OSAS and obesity related factors on cytokine secretion and expression of different adhesion molecules by THP-1 monocytes was analysed. Our data revealed a significant redistribution of circulating classical and intermediate monocytes in all three patient cohorts, but differential effects in terms of monocytic adhesion molecules CD11a, CD11b, CD11c, CX3CR1, CD29, CD49d, and plasma cytokine levels. These data were reflected by differential effects of OSAS and obesity related factors leptin, TNFα and hypoxia on THP-1 cytokine secretion patterns and expression of adhesion molecules CD11b and CD49d. In summary, our data revealed differential effects of OSAS and obesity, which underlines the need for a customized therapeutic regimen with respect to the individual weighting of these overlapping diseases.
Collapse
Affiliation(s)
- Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| | - Friederike Katharina Kosyna
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Reinhard Depping
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Christian Lange
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Svenja Meyhöfer
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sebastian M Meyhöfer
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jens U Marquardt
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Armin Steffen
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| |
Collapse
|
20
|
Pathak MP, Patowary P, Chattopadhyay P, Barbhuiyan PA, Islam J, Gogoi J, Wankhar W. Obesity-associated Airway Hyperresponsiveness: Mechanisms Underlying Inflammatory Markers and Possible Pharmacological Interventions. Endocr Metab Immune Disord Drug Targets 2024; 24:1053-1068. [PMID: 37957906 DOI: 10.2174/0118715303256440231028072049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 11/15/2023]
Abstract
Obesity is rapidly becoming a global health problem affecting about 13% of the world's population affecting women and children the most. Recent studies have stated that obese asthmatic subjects suffer from an increased risk of asthma, encounter severe symptoms, respond poorly to anti-asthmatic drugs, and ultimately their quality-of-life decreases. Although, the association between airway hyperresponsiveness (AHR) and obesity is a growing concern among the public due to lifestyle and environmental etiologies, however, the precise mechanism underlying this association is yet to establish. Apart from aiming at the conventional antiasthmatic targets, treatment should be directed towards ameliorating obesity pathogenesis too. Understanding the pathogenesis underlying the association between obesity and AHR is limited, however, a plethora of obesity pathologies have been reported viz., increased pro-inflammatory and decreased anti-inflammatory adipokines, depletion of ROS controller Nrf2/HO-1 axis, NLRP3 associated macrophage polarization, hypertrophy of WAT, and down-regulation of UCP1 in BAT following down-regulated AMPKα and melanocortin pathway that may be correlated with AHR. Increased waist circumference (WC) or central obesity was thought to be related to severe AHR, however, some recent reports suggest body mass index (BMI), not WC tends to exaggerate airway closure in AHR due to some unknown mechanisms. This review aims to co-relate the above-mentioned mechanisms that may explain the copious relation underlying obesity and AHR with the help of published reports. A proper understanding of these mechanisms discussed in this review will ensure an appropriate treatment plan for patients through advanced pharmacological interventions.
Collapse
Affiliation(s)
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India
| | | | | | - Johirul Islam
- Department of Pharmaceutical Sciences, School of Health Sciences, Assam Kaziranga University, Jorhat, India
| | - Jyotchna Gogoi
- Department of Biochemistry, Faculty of Science, Assam Down Town University, Guwahati, India
| | - Wankupar Wankhar
- Department of Dialysis, Faculty of Paramedical Science, Assam Down Town University, Guwahati, India
| |
Collapse
|
21
|
Hua Y, Xie D, Zhang Y, Wang M, Wen W, Sun J. Identification and analysis of key genes in adipose tissue for human obesity based on bioinformatics. Gene 2023; 888:147755. [PMID: 37659596 DOI: 10.1016/j.gene.2023.147755] [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: 07/09/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Obesity is a complex condition that is affected by a variety of factors, including the environment, behavior, and genetics. However, the genetic mechanisms underlying obesity remains poorly elucidated. Therefore, our study aimed at identifying key genes for human obesity using bioinformatics analysis. METHODS The microarray datasets of adipose tissue in humans were downloaded from the Gene Expression Omnibus (GEO) database. After the selection of differentially expressed genes (DEGs), we used Lasso regression and Support Vector Machine (SVM) algorithm to further identify the feature genes. Moreover, immune cell infiltration analysis, gene set variation analysis (GSVA), GeneCards database and transcriptional regulation analysis were conducted to study the potential mechanisms by which the feature genes may impact obesity. We utilized receiver operating characteristic (ROC) curve to analysis the diagnostic efficacy of feature genes. Finally, we verified the feature genes in cell experiments and animal experiments. The statistical analyses in validation experiments were conducted using SPSS version 28.0, and the graph were generated using GraphPad Prism 9.0 software. The bioinformatics analyses were conducted using R language (version 4.2.2), with a significance threshold of p < 0.05 used. RESULTS 199 DEGs were selected using Limma package, and subsequently, 5 feature genes (EGR2, NPY1R, GREM1, BMP3 and COL8A1) were selected through Lasso regression and SVM algorithm. Through various bioinformatics analyses, we found some signaling pathways by which feature genes influence obesity and also revealed the crucial role of these genes in the immune microenvironment, as well as their strong correlations with obesity-related genes. Additionally, ROC curve showed that all the feature genes had good predictive and diagnostic efficiency in obesity. Finally, after validation through in vitro experiments, EGR2, NPY1R and GREM1 were identified as the key genes. CONCLUSIONS This study identified EGR2, GREM1 and NPY1R as the potential key genes and potential diagnostic biomarkers for obesity in humans. Moreover, EGR2 was discovered as a key gene for obesity in human adipose tissue for the first time, which may provide novel targets for diagnosing and treating obesity.
Collapse
Affiliation(s)
- Yuchen Hua
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Danyingzhu Xie
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong Province 510282, China
| | - Yugang Zhang
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong Province 510282, China
| | - Ming Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong Province 510282, China.
| | - Weiheng Wen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong Province 510282, China.
| | - Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong Province 510282, China.
| |
Collapse
|
22
|
Hu T, Xu Y, Shen Y, Li X, Xiao Y, Wang Y, Bao Y, Ma X. Interaction between serum neutrophil gelatinase associated lipocalin and visceral fat area on cardiovascular health in a cohort of community-based individuals. Clin Chim Acta 2023; 551:117606. [PMID: 37844679 DOI: 10.1016/j.cca.2023.117606] [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: 04/06/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND We assessed the predictive values of neutrophil gelatinase-associated lipocalin (NGAL), fat distribution, and their interaction on the development of major adverse cardiovascular events (MACE) in a community-based cohort of middle-aged and older individuals. METHODS This prospective study involved 1349 adults (43.2% men) aged 50-80 y, without baseline cardiovascular diseases, from communities in 2013-2014. All participants were followed up for a mean of 7.6 y via phone calls and medical records. Serum NGAL concentrations were analyzed at baseline. Fat distribution, including subcutaneous fat area and visceral fat area (VFA), was assessed by magnetic resonance imaging. RESULTS In fully-adjusted Cox regression models, baseline high NGAL concentrations were related to an increased risk of MACE in women [HR 1.75, 95% CI 1.03-2.99], compared with low NGAL concentrations. After stratification by VFA concentrations, the observed association was more predominant in women with baseline low VFA (HR 1.24, 95% CI 1.11-1.38). Moreover, the association between NGAL and MACE was interacted by VFA, strengthening the association at low VFA concentrations (Pinteraction < 0.05). CONCLUSIONS Serum NGAL determined at baseline predicts the development of MACE, and the association is modified by VFA in women.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Yiting Xu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Yun Shen
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Xiaoya Li
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Yunfeng Xiao
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yufei Wang
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China.
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China.
| |
Collapse
|
23
|
Sadeghi A, Niknam M, Momeni-Moghaddam MA, Shabani M, Aria H, Bastin A, Teimouri M, Meshkani R, Akbari H. Crosstalk between autophagy and insulin resistance: evidence from different tissues. Eur J Med Res 2023; 28:456. [PMID: 37876013 PMCID: PMC10599071 DOI: 10.1186/s40001-023-01424-9] [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] [Received: 04/04/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Insulin is a critical hormone that promotes energy storage in various tissues, as well as anabolic functions. Insulin resistance significantly reduces these responses, resulting in pathological conditions, such as obesity and type 2 diabetes mellitus (T2DM). The management of insulin resistance requires better knowledge of its pathophysiological mechanisms to prevent secondary complications, such as cardiovascular diseases (CVDs). Recent evidence regarding the etiological mechanisms behind insulin resistance emphasizes the role of energy imbalance and neurohormonal dysregulation, both of which are closely regulated by autophagy. Autophagy is a conserved process that maintains homeostasis in cells. Accordingly, autophagy abnormalities have been linked to a variety of metabolic disorders, including insulin resistance, T2DM, obesity, and CVDs. Thus, there may be a link between autophagy and insulin resistance. Therefore, the interaction between autophagy and insulin function will be examined in this review, particularly in insulin-responsive tissues, such as adipose tissue, liver, and skeletal muscle.
Collapse
Affiliation(s)
- Asie Sadeghi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Niknam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Bastin
- Clinical Research Development Center "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Teimouri
- Department of Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Akbari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
24
|
Li J, Xu P, Hong Y, Xie Y, Peng M, Sun R, Guo H, Zhang X, Zhu W, Wang J, Liu X. Lipocalin-2-mediated astrocyte pyroptosis promotes neuroinflammatory injury via NLRP3 inflammasome activation in cerebral ischemia/reperfusion injury. J Neuroinflammation 2023; 20:148. [PMID: 37353794 DOI: 10.1186/s12974-023-02819-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/26/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Neuroinflammation is a vital pathophysiological process during ischemic stroke. Activated astrocytes play a major role in inflammation. Lipocalin-2 (LCN2), secreted by activated astrocytes, promotes neuroinflammation. Pyroptosis is a pro-inflammatory form of programmed cell death that has emerged as a new area of research in stroke. Nevertheless, the potential role of LCN2 in astrocyte pyroptosis remains unclear. METHODS An ischemic stroke model was established by middle cerebral artery occlusion (MCAO) in vivo. In this study, in vitro, oxygen-glucose deprivation and reoxygenation (O/R) were applied to cultured astrocytes. 24p3R (the LCN2 receptor) was inhibited by astrocyte-specific adeno-associated virus (AAV-GFAP-24p3Ri). MCC950 and Nigericin sodium salt (Nig) were used to inhibit or promote the activation of NLRP3 inflammasome pharmacologically, respectively. Histological and biochemical analyses were performed to assess astrocyte and neuron death. Additionally, the neurological deficits of mice were evaluated. RESULTS LCN2 expression was significantly induced in astrocytes 24 h after stroke onset in the mouse MCAO model. Lcn2 knockout (Lcn2-/-) mice exhibited reduced infarct volume and improved neurological and cognitive functions after MCAO. LCN2 and its receptor 24p3R were colocalized in astrocytes. Mechanistically, suppression of 24p3R by AAV-GFAP-24p3Ri alleviated pyroptosis-related pore formation and the secretion of pro-inflammatory cytokines via LCN2, which was then reversed by Nig-induced NLRP3 inflammasome activation. Astrocyte pyroptosis was exacerbated in Lcn2-/- mice by intracerebroventricular administration of recombinant LCN2 (rLCN2), while this aggravation was restricted by blocking 24p3R or inhibiting NLRP3 inflammasome activation with MCC950. CONCLUSION LCN2/24p3R mediates astrocyte pyroptosis via NLRP3 inflammasome activation following cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Juanji Li
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Pengfei Xu
- Division of Life Sciences and Medicine, Department of Neurology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Yi Xie
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Mengna Peng
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Rui Sun
- Department of Neurology, Shanghai Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Hongquan Guo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiaohao Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Junjun Wang
- Department of Clinical Laboratory, Affiliated Jinling Hospital, Medical School of Nanjing University, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Xinfeng Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China.
- Division of Life Sciences and Medicine, Department of Neurology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
25
|
Baragetti A, Da Dalt L, Moregola A, Svecla M, Terenghi O, Mattavelli E, De Gaetano LN, Uboldi P, Catapano AL, Norata GD. Neutrophil aging exacerbates high fat diet induced metabolic alterations. Metabolism 2023; 144:155576. [PMID: 37116643 DOI: 10.1016/j.metabol.2023.155576] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND High fat diet (HFD) chronically hyper-activate the myeloid cell precursors, but whether it affects the neutrophil aging is unknown. PURPOSE We characterized how HFD impacts neutrophil aging, infiltration in metabolic tissues and if this aging, in turn, modulates the development of metabolic alterations. We immunophenotyped neutrophils and characterized the metabolic responses in physiology (wild-type mice, WT) and in mice with constitutively aged neutrophils (MRP8 driven conditional deletion of CXCR4; herein CXCR4fl/flCre+) or with constitutively fresh neutrophils (MRP8 driven conditional deletion of CXCR2; CXCR2fl/flCre+), following 20 weeks of HFD feeding (45 % kcal from fat). FINDINGS After 20 weeks HFD, the gluco-metabolic profile of CXCR4fl/flCre+ mice was comparable to that of WT mice, while CXCR2fl/flCre+ mice were protected from metabolic alterations. CXCR4fl/flCre+ infiltrated more, but CXCR2fl/flCre+ neutrophils infiltrated less, in liver and visceral adipose tissue (VAT). As consequence, while CXCR4fl/flCre+ resulted into hepatic "suicidal" neutrophils extracellular traps (NETs) and altered immune cell architecture in VAT, CXCR2fl/flCre+ promoted proresolutive hepatic NETs and reduced accumulation of pro-inflammatory macrophages in VAT. In humans, higher Cxcl12 (CXCR4 ligand) plasma levels correlated with visceral adiposity while higher levels of Cxcl1, the ligand of CXCR2, correlated with indexes of hepatic steatosis, adiposity and metabolic syndrome. CONCLUSIONS Neutrophil aging might contribute to the development of HFD induced metabolic disorders.
Collapse
Affiliation(s)
- Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Moregola
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monika Svecla
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Ottavia Terenghi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elisa Mattavelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Lucia Nicolini De Gaetano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS Multimedica Hospital, Milan, Italy
| | - Patrizia Uboldi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alberico Luigi Catapano
- SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy; IRCCS Multimedica Hospital, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; SISA Centre for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.
| |
Collapse
|