1
|
Gong Y, Tian X, Ma X, Xing J, Ji Z, Gao X, Chen Z, Hu F, Yin D, Li F, Sheng C, Liu Y, Shi S. New insights into the pharmacological mechanisms of Jinqi Jiangtang Tablets in the treatment of type 2 diabetes mellitus: A multi-omics approach combined with experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025:120020. [PMID: 40449695 DOI: 10.1016/j.jep.2025.120020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) is a prevalent chronic metabolic disorder characterized by impaired glucose homeostasis and insulin dysfunction. Despite ongoing efforts, the management of T2DM remains challenging due to its complex pathogenesis and the limited effectiveness of current treatments. Jinqi Jiangtang Tablets (JQJTT), a well-established Traditional Chinese Medicine (TCM) formula, have shown promising clinical benefits in regulating blood glucose levels and improving metabolic health in T2DM patients. However, the specific pharmacological mechanisms behind its efficacy are still not fully understood, prompting the need for deeper exploration into its therapeutic actions. AIM OF THE STUDY This study aims to elucidate the pharmacologically active ingredients and mechanisms of JQJTT in T2DM using multi-omics methods combined with experimental verification. MATERIALS AND METHODS Liquid chromatography-mass spectrometry (LC-MS) identified the chemical profile of JQJTT, and targets were screened via SwissTargetPrediction database. T2DM-related targets were acquired from GeneCards and Gene Expression Omnibus (GEO) databases. Therapeutic targets were ascertained by intersecting JQJTT and T2DM targets, followed by Protein-Protein Interaction (PPI) network analysis. GO and KEGG pathway enrichment was performed, and the compound-target-pathway network was built using Cytoscape. Mendelian randomization, single-sample Gene Set Enrichment Analysis (ssGSEA), and CIBERSORT analyses were executed to explore immune cell involvement. Single-cell RNA sequencing (scRNA-seq) identified affected cell types, while molecular docking and molecular dynamics (MD) simulation assessed compound-target affinities. Metabolomics and 16S rDNA sequencing analyzed JQJTT's effects on metabolites and gut microbiota. RESULTS LC-MS identified 161 compounds in JQJTT. Through network pharmacology, 6 core targets (AKT1, MMP9, HSP90AA1, CASP3, IL-6, PTGS2) were screened, and PI3K/AKT, NFκB, and lipid metabolism were pinpointed as key regulatory pathways. Mendelian randomization linked 39 immune cell types to T2DM, while ssGSEA and CIBERSORT suggested JQJTT potentially modulated the proportion of immune cells. scRNA-seq indicated JQJTT may affect macrophage polarization. Tangeritin, isorhamnetin, and kaempferol were determined as the main active compounds with glucose-lowering effects confirmed in vitro. In vivo experiments validated the molecular mechanisms of JQJTT against T2DM. Metabolomics unearthed 56 differentially abundant metabolites, primarily in linoleic acid, arachidonic acid, and pyruvaldehyde metabolisms. JQJTT also improved gut microbiota diversity in T2DM. CONCLUSION JQJTT treated T2DM through a multi-target, multi-pathway approach, including attenuating inflammation, reshaping macrophage polarization, enhancing insulin signaling, regulating metabolites, and restoring intestinal flora balance.
Collapse
Affiliation(s)
- Yining Gong
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| | - Xiaoqing Ma
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Jin Xing
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Zongwen Ji
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Xueying Gao
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Zhi Chen
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Fangzhi Hu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Dandan Yin
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Feng Li
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Can Sheng
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining 272000, China.
| | - Yaping Liu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Shulong Shi
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China; Cisen Pharmaceutical Co., Ltd, Jining 272000, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
2
|
Vallazhath A, Thimmappa PY, Joshi HB, Hebbar KR, Nayak A, Umakanth S, Saoji AA, Manjunath NK, Hadapad BS, Joshi MB. A comprehensive review on the implications of Yogic/Sattvic diet in reducing inflammation in type 2 diabetes. Nutr Diabetes 2025; 15:14. [PMID: 40216734 PMCID: PMC11992243 DOI: 10.1038/s41387-025-00371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic inflammation in type 2 diabetes (T2D), characterized by constitutively activated immune cells and elevated pro-inflammatory mediators along with hyperglycaemia and increased free fatty acids and branched chain amino acid levels, significantly alters the immuno-metabolic axis. Over the years, dietary intervention has been explored as an effective strategy for managing T2D. Evidence from experimental and clinical studies indicates that various diets, including Mediterranean, Nordic, Palaeolithic and ketogenic diets, increase insulin sensitivity, decrease gluconeogenesis, and adiposity, and exert anti-inflammatory effects, thus preserving immuno-metabolic homeostasis in individuals with T2D. Indian dietary sources are categorized as Sattvic, Rajasic, and Tamasic, depending on their impact on health and behaviour. The Yogic diet, commonly recommended during yoga practice, is predominantly Sattvic, emphasizing plant-based whole foods while limiting processed and high-glycaemic-index items. Yogic diet is also recommended for Mitahara, emphasizing mindful eating, which is attributed to calorie restriction. Adopting a Yogic diet, featuring low-fat vegetarian principles, strongly reduces inflammatory mediator levels. This diet not only ameliorates insulin resistance and maintains a healthy body weight but also regulates immunomodulation, enhances gut microbiome diversity and provides essential phytonutrients, collectively preventing inflammation. Although, preliminary studies show aforementioned beneficial role of Yogic diet in improving diabetes associated metabolic and inflammatory changes, precise cellular and molecular mechanisms are not yet understood. Hence, further studies are warranted to decipher the mechanisms. This review summarizes the multiple roles of Yogic diet and related dietary components in mitigating inflammation and enhancing glycaemic control in T2D.
Collapse
Affiliation(s)
- Anupama Vallazhath
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Harshit B Joshi
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Krishna Raghava Hebbar
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anupama Nayak
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | | | - Apar Avinash Saoji
- Swami Vivekananda Yoga Anusandhana Samsthana, Bangalore, 560105, Karnataka, India
| | | | - Basavaraj S Hadapad
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Centre for Ayurveda Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
3
|
Chen D, Tang H, Liu J, Zhang H, Rao K, Teng X, Yang F, Liu H. Luteolin-mediated phosphoproteomic changes in chicken splenic lymphocytes: Unraveling the detoxification mechanisms against ammonia-induced stress. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136931. [PMID: 39709809 DOI: 10.1016/j.jhazmat.2024.136931] [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: 06/19/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Long-term exposure to high ammonia concentrations could severely impact chicken health. On the other hand, luteolin has been shown to protect against ammonia poisoning. Although phosphorylation is critically involved in toxicity induction, the specific role of phosphorylated proteins in ammonia poisoning remains unclear. Herein, we constructed an in vitro model to study chicken ammonia poisoning and also analyzed the protective effects of luteolin. Specifically, a combined series of organic techniques such as protein extraction, enzyme digestion, modified peptide enrichment, Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) analysis, and bioinformatics analysis were employed for a quantitative omics study of phosphorylation modification in three groups of samples. Our findings revealed thousands of Differentially Expressed Proteins (DEPs). The differentially expressed modified proteins were subjected to GO classification, KEGG pathway analysis, cluster analysis, and protein interaction analysis, revealing the detoxification mechanism encompassed mitochondrial maintenance, signal transduction, transcriptional regulation, and cytoskeleton regulation. In the process, mitochondria and Golgi apparatus were the key organelles. Furthermore, the AKT1/FOXO signaling pathway and Heat Shock Proteins (HSPs) were the key core modifiers of the proteins. We hope that our findings will provide a theoretical basis and experimental support for future research on luteolin's detoxification mechanism against ammonia poisoning.
Collapse
Affiliation(s)
- Dechun Chen
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Haojinming Tang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Jiahao Liu
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Huanrong Zhang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Kaijing Rao
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Falong Yang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China.
| | - Haifeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
4
|
Visvanathan R, Houghton MJ, Williamson G. Impact of Glucose, Inflammation and Phytochemicals on ACE2, TMPRSS2 and Glucose Transporter Gene Expression in Human Intestinal Cells. Antioxidants (Basel) 2025; 14:253. [PMID: 40227199 PMCID: PMC11939507 DOI: 10.3390/antiox14030253] [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: 01/15/2025] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 04/15/2025] Open
Abstract
Inflammation is associated with the pathophysiology of type 2 diabetes and COVID-19. Phytochemicals have the potential to modulate inflammation, expression of SARS-CoV-2 viral entry receptors (angiotensin-converting enzyme 2 (ACE2) and transmembrane protease, serine 2 (TMPRSS2)) and glucose transport in the gut. This study assessed the impact of phytochemicals on these processes. We screened 12 phytochemicals alongside 10 pharmaceuticals and three plant extracts, selected for known or hypothesised effects on the SARS-CoV-2 receptors and COVID-19 risk, for their effects on the expression of ACE2 or TMPRSS2 in differentiated Caco-2/TC7 human intestinal epithelial cells. Genistein, apigenin, artemisinin and sulforaphane were the most promising ones, as assessed by the downregulation of TMPRSS2, and thus they were used in subsequent experiments. The cells were then co-stimulated with pro-inflammatory cytokines interleukin-1 beta (IL-1β) and tumour necrosis factor-alpha (TNF-α) for ≤168 h to induce inflammation, which are known to induce multiple pathways, including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. Target gene expression (ACE2, TMPRSS2, SGLT1 (sodium-dependent glucose transporter 1) and GLUT2 (glucose transporter 2)) was measured by droplet digital PCR, while interleukin-1 (IL-6), interleukin-1 (IL-8) and ACE2 proteins were assessed using ELISA in both normal and inflamed cells. IL-1β and TNF-α treatment upregulated ACE2, TMPRSS2 and SGLT1 gene expression. ACE2 increased with the duration of cytokine exposure, coupled with a significant decrease in IL-8, SGLT1 and TMPRSS2 over time. Pearson correlation analysis revealed that the increase in ACE2 was strongly associated with a decrease in IL-8 (r = -0.77, p < 0.01). The regulation of SGLT1 gene expression followed the same pattern as TMPRSS2, implying a common mechanism. Although none of the phytochemicals decreased inflammation-induced IL-8 secretion, genistein normalised inflammation-induced increases in SGLT1 and TMPRSS2. The association between TMPRSS2 and SGLT1 gene expression, which is particularly evident in inflammatory conditions, suggests a common regulatory pathway. Genistein downregulated the inflammation-induced increase in SGLT1 and TMPRSS2, which may help lower the postprandial glycaemic response and COVID-19 risk or severity in healthy individuals and those with metabolic disorders.
Collapse
Affiliation(s)
- Rizliya Visvanathan
- Department of Nutrition, Dietetics and Food, BASE Facility, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia
- Victorian Heart Institute, Monash University, Level 2, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Michael J. Houghton
- Department of Nutrition, Dietetics and Food, BASE Facility, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia
- Victorian Heart Institute, Monash University, Level 2, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, BASE Facility, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC 3168, Australia
- Victorian Heart Institute, Monash University, Level 2, Victorian Heart Hospital, 631 Blackburn Road, Clayton, VIC 3168, Australia
| |
Collapse
|
5
|
Wu Q, Zeng Y, Geng K, Guo M, Teng FY, Yan PJ, Lei Y, Long Y, Jiang ZZ, Law BYK, Xu Y. The role of IL-1 family cytokines in diabetic cardiomyopathy. Metabolism 2025; 163:156083. [PMID: 39603339 DOI: 10.1016/j.metabol.2024.156083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with diabetes and is characterised by contractile dysfunction and left ventricular hypertrophy. The complex pathological and physiological mechanisms underlying DCM have contributed to a limited number of available treatment options. A substantial body of evidence has established that DCM is a low-grade inflammatory cardiovascular disorder, with the interleukin-1 (IL-1) family of cytokines playing crucial roles in initiating inflammatory responses and shaping innate and adaptive immunity. In this review, we aim to provide an overview of the underlying mechanisms of the IL-1 family and their relevance in DCM of various aetiologies. Furthermore, we highlighted potential therapeutic targets within the IL-1 family for the management of DCM.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Pathology, and Luzhou Key Laboratory of Precision Pathology Diagnosis for Serious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Long
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China.
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
6
|
Guo C, Xu P, Luo W, Zhang J, Sun X, Hoang H, Ma D, Wu D, Zhong J, Miao C. The Role of Dectin-1-Akt-RNF146 Pathway in β-Glucan Induced Immune Trained State of Monocyte in Sepsis. J Inflamm Res 2025; 18:1147-1165. [PMID: 39881796 PMCID: PMC11775823 DOI: 10.2147/jir.s482213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025] Open
Abstract
Background Sepsis is regarded as a dysregulated immune response to infections. Recent study showed partially reversal of immunosuppression by trained immunity, which fosters an enhanced immune response towards a secondary challenge. However, the role of trained immunity in sepsis has not been fully understood. Methods We profiled the characteristics of peripheral blood mononuclear cells from septic patients using single-cell RNA sequencing (scRNA-seq) analyses. Murine double-hit models (pretreatment or post-treatment of β-glucan in septic mice) and murine monocyte/macrophage cell line RAW264.7 were used then. Results scRNA-seq revealed that Ring finger protein 146 (RNF146) and protein kinase B (Akt) were downregulated in the immunosuppression period of septic patients and were verified to be decreased in bone marrow and spleen monocytes from septic mice. While β-glucan pretreatment improved the immunosuppressed state in septic mice and increased dectin-1/Akt/RNF146 expressions in monocytes, along with the increased survival rate, inflammatory factors and aerobic glycolysis, indicating a change from immunosuppression to immune training. Moreover, RNF146 regulated dectin-1-Akt-mTOR signaling in the trained immune state of murine monocyte/macrophage RAW264.7 cell line and the expression of RNF146 was dependent on dectin-1-Akt activation. The inhibition of dectin-1 by its antagonist laminarin downregulated Akt-RNF146 signaling and partially reversed β-glucan induced trained immunity in septic mice. Conclusion RNF146 and Akt are downregulated in the immunosuppression period of sepsis, while increased after β-glucan pretreatment induced trained immunity in septic mice. Moreover, RNF146 regulates the immune trained state of monocyte through dectin-1-Akt-mTOR pathway, suggesting a possible target in reversal of immunosuppression in sepsis.
Collapse
Affiliation(s)
- Chenyue Guo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Peiyao Xu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Jinlin Zhang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Xingfeng Sun
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Harry Hoang
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Dehua Wu
- Department of Anesthesiology, Shanghai Songjiang District Central Hospital, Shanghai, People’s Republic of China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Meier DT, de Paula Souza J, Donath MY. Targeting the NLRP3 inflammasome-IL-1β pathway in type 2 diabetes and obesity. Diabetologia 2025; 68:3-16. [PMID: 39496966 DOI: 10.1007/s00125-024-06306-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 11/06/2024]
Abstract
Increased activity of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome-IL-1β pathway is observed in obesity and contributes to the development of type 2 diabetes and its complications. In this review, we describe the pathological activation of IL-1β by metabolic stress, ageing and the microbiome and present data on the role of IL-1β in metabolism. We explore the physiological role of the IL-1β pathway in insulin secretion and the relationship between circulating levels of IL-1β and the development of diabetes and associated diseases. We highlight the paradoxical nature of IL-1β as both a friend and a foe in glucose regulation and provide details on clinical translation, including the glucose-lowering effects of IL-1 antagonism and its impact on disease modification. We also discuss the potential role of IL-1β in obesity, Alzheimer's disease, fatigue, gonadal dysfunction and related disorders such as rheumatoid arthritis and gout. Finally, we address the safety of NLRP3 inhibition and IL-1 antagonists and the prospect of using this therapeutic approach for the treatment of type 2 diabetes and its comorbidities.
Collapse
Affiliation(s)
- Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Joyce de Paula Souza
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
8
|
Huard A, Fauteux-Daniel S, Goldstein J, Martin P, Jarlborg M, Andries J, Caruso A, Díaz-Barreiro A, Rodriguez E, Vaillant L, Savvides SN, Gabay C. Development of anti-murine IL-18 binding protein antibodies to stimulate IL-18 bioactivity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:180-191. [PMID: 40018678 PMCID: PMC7617445 DOI: 10.1093/jimmun/vkae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Interleukin (IL)-18 is an immunoregulatory cytokine that acts as a potent inducer of T helper 1 and cytotoxic responses. IL-18 activity is regulated by its decoy receptor IL-18 binding protein (IL-18BP) which forms a high affinity complex with IL-18 to block binding of the cognate receptors. A disbalance between IL-18 and IL-18BP associated with excessive IL-18 signaling can lead to systemic inflammation. Indeed, the severity of CpG-induced macrophage activation syndrome (MAS) is exacerbated in IL-18BP KO mice. On the contrary, targeting IL-18BP can have promising effects to enhance immune responses against pathogens and cancer. We generated monoclonal rabbit anti-mouse IL-18BP antibodies labeled from 441 to 450. All antibodies, except from antibody 443, captured mIL-18BP when used in a sandwich ELISA. Using an IL-18 bioassay, we showed that antibody 441 did not interfere with the regulatory effect of mIL-18BP, whereas all other antibodies displayed different levels of antagonism. Further experiments were performed using antibody 445 endowed with potent neutralizing activity and antibody 441. Despite binding to IL-18BP with the same affinity, antibody 445, but not antibody 441, was able to release IL-18 from preformed IL-18-IL-18BP complexes. Administration of antibody 445 significantly aggravated the severity of CpG-induced MAS as compared to antibody 441. Additional experiments using naïve WT, IL-18BP KO, and IL-18 KO mice confirmed the specificity of the neutralizing effect of antibody 445 towards IL-18BP. Our studies led to the development of a monoclonal anti-IL-18BP antibody with neutralizing activity that results in the promotion of IL-18 activities.
Collapse
Affiliation(s)
- Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Sébastien Fauteux-Daniel
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Jérémie Goldstein
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Praxedis Martin
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Matthias Jarlborg
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Julie Andries
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, 9052, Ghent, Belgium
| | - Assunta Caruso
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Alejandro Díaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Emiliana Rodriguez
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Laurie Vaillant
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Savvas N. Savvides
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, 9052, Ghent, Belgium
| | - Cem Gabay
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| |
Collapse
|
9
|
Abstract
Inflammation is an essential physiological defence mechanism, but prolonged or excessive inflammation can cause disease. Indeed, unresolved systemic and adipose tissue inflammation drives obesity-related cardiovascular disease and type 2 diabetes mellitus. Drugs targeting pro-inflammatory cytokine pathways or inflammasome activation have been approved for clinical use for the past two decades. However, potentially serious adverse effects, such as drug-induced weight gain and increased susceptibility to infections, prevented their wider clinical implementation. Furthermore, these drugs do not modulate the resolution phase of inflammation. This phase is an active process orchestrated by specialized pro-resolving mediators, such as lipoxins, and other endogenous resolution mechanisms. Pro-resolving mediators mitigate inflammation and development of obesity-related disease, for instance, alleviating insulin resistance and atherosclerosis in experimental disease models, so mechanisms to modulate their activity are, therefore, of great therapeutic interest. Here, we review current clinical attempts to either target pro-inflammatory mediators (IL-1β, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, tumour necrosis factor (TNF) and IL-6) or utilize endogenous resolution pathways to reduce obesity-related inflammation and improve cardiometabolic outcomes. A remaining challenge in the field is to establish more precise biomarkers that can differentiate between acute and chronic inflammation and to assess the functionality of individual leukocyte populations. Such advancements would improve the monitoring of drug effects and support personalized treatment strategies that battle obesity-related inflammation and cardiometabolic disease.
Collapse
Affiliation(s)
- Matúš Soták
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Madison Clark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bianca E Suur
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emma Börgeson
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Velloso LA, Donato J. Growth Hormone, Hypothalamic Inflammation, and Aging. J Obes Metab Syndr 2024; 33:302-313. [PMID: 39639711 PMCID: PMC11704225 DOI: 10.7570/jomes24032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
While inflammation is a crucial response in injury repair and tissue regeneration, chronic inflammation is a prevalent feature in various chronic, non-communicable diseases such as obesity, diabetes, and cancer and in cardiovascular and neurodegenerative diseases. Long-term inflammation considerably affects disease prevalence, quality of life, and longevity. Our research indicates that the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis is a pivotal regulator of inflammation in some tissues, including the hypothalamus, which is a key player in systemic metabolism regulation. Moreover, the GH/IGF-1 axis is strongly linked to longevity, as GH- or GH receptor-deficient mice live approximately twice as long as wild-type animals and exhibit protection against aging-induced inflammation. Conversely, GH excess leads to increased neuroinflammation and reduced longevity. Our review studies the associations between the GH/IGF-1 axis, inflammation, and aging, with a particular focus on evidence suggesting that GH receptor signaling directly induces hypothalamic inflammation. This finding underscores the significant impact of changes in the GH axis on metabolism and on the predisposition to chronic, non-communicable diseases.
Collapse
Affiliation(s)
- Licio A. Velloso
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Campinas, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
11
|
Zhang ZY, Guo XL, Liu JTY, Gu YJ, Ji XW, Zhu S, Xie JY, Guo F. Conjugated bile acids alleviate acute pancreatitis through inhibition of TGR5 and NLRP3 mediated inflammation. J Transl Med 2024; 22:1124. [PMID: 39707318 DOI: 10.1186/s12967-024-05922-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Severe acute pancreatitis (SAP) is a crucial gastrointestinal disease characterized by systemic inflammatory responses and persistent multiple organ failure. The role of bile acids (BAs) in diverse inflammatory diseases is increasingly recognized as crucial, but the underlying role of BA conjugation remains elusive. OBJECTIVES Our study aim to investigate the potential role of conjugated bile acids in SAP and reveal the molecular mechanisms underlying its regulatory effects. We hypothesized that taurochenodeoxycholic acid (TCDCA) and glycochenodeoxycholic acid (GCDCA) could protect SAP through inhibiting the activation of NLRP3 inflammasomes via the TGR5 pathway in macrophages. METHODS To test our hypothesis, we used BA-CoA: amino acid N-acyltransferase knockout (Baat-/-) mice and established SAP mouse models using caerulein- and sodium taurocholate- induced. We utilized a range of methods, including pathology sections, qRT-PCR, immunofluorescence, Western blotting, and ELISA, to identify the mechanisms of regulation. RESULTS BA-CoA: Amino acid N-acyltransferase knockout (Baat-/-) mice significantly exacerbated pancreatitis by increasing pancreatic and systemic inflammatory responses and pancreatic damage in SAP mouse models. Moreover, the serum TCDCA levels in Baat-/- mice were lower than those in wild-type (WT) mice with or without SAP, and GCDCA and TCDCA showed stronger anti-inflammatory effects than chenodeoxycholic acid (CDCA) in vitro. TCDCA treatment alleviated SAP in a Takeda G protein-coupled receptor 5 and NOD-like receptor family, pyrin domain containing 3-dependent manner in vivo. Reinforcing our conclusions from the mouse study, clinical SAP patients exhibited decreased serum content of conjugated BAs, especially GCDCA, which was inversely correlated with the severity of systemic inflammatory responses. CONCLUSION Conjugated bile acids significantly inhibit NLRP3 inflammasome activation by activating TGR5 pathway, thereby alleviating pancreatic immunopathology. The results provide new insights into the variability of clinical outcomes and paves the way for developing more effective therapeutic interventions for AP.
Collapse
Affiliation(s)
- Zi-Yi Zhang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiu-Liu Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing-Tian-Yi Liu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi-Jie Gu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Xing-Wei Ji
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Shu Zhu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jin-Yan Xie
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, People's Republic of China.
| | - Feng Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, People's Republic of China.
| |
Collapse
|
12
|
Bojarczuk A, Garbacz A, Żekanowski C, Borzemska B, Cięszczyk P, Maculewicz E. Systematic Review of IL-1, IL-4, IL-6, IL-10, IL-15, and IL-18 Gene Polymorphisms and Meta-Analysis of IL-6 Variant and Its Association with Overweight and Obesity Risk in Men. Int J Mol Sci 2024; 25:13501. [PMID: 39769263 PMCID: PMC11679641 DOI: 10.3390/ijms252413501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Obesity is a complex health risk influenced by genetic, environmental, and lifestyle factors. This review systematically assessed the association between interleukin gene polymorphisms (rs16944, rs17561, rs1143623, rs1143633, rs1143634, rs1800587, rs2234677, and rs4848306), IL-4 (rs180275, rs1805010, IL-6 rs13306435, rs1800795, rs1800796, rs1800797, rs2228145, rs2228145, rs2229238, and rs4845623), IL-10 (rs1518110, rs1518111, rs1800871, rs1800872, rs1800896, rs1878672, rs2834167, rs3024491, rs3024496, rs3024498, and rs3024505), IL-15 (rs3136617, rs3136618, and rs2296135), and IL-18 (rs187238, rs1946518, rs2272127, rs2293225, and rs7559479) and the risk of overweight and obesity in adults, focusing on IL-6 rs1800795 through a meta-analysis. The focus on IL-6 in this review arises from its pleiotropic nature and unclear effect on obesity risk. The review included studies published from 1998 to 2023, sourced from Science Direct, EBSCOhost, Web of Science, and Google Scholar. Bias was assessed with the Cochrane Collaboration tool, and funnel plots were used for publication bias. Results were synthesized into pooled odds ratios (ORs) and confidence intervals (CIs). Thirty studies comprising approximately 29,998 participants were included. The selection criteria required that the articles include participants who were overweight or obese, and this condition needed to be linked to IL polymorphisms. In a meta-analysis, in the dominant model, the pooled OR was 1.26 (95% CI 1.08 to 1.47), indicating those with the GC/CC genotype for IL-6 rs1800795 are 1.26 times more likely to be overweight/obese than GG genotype carriers. For the recessive model, the OR was 1.25 (95% CI 1.04 to 1.51). The overdominant model showed no significant association (OR 1.08, 95% CI 0.94 to 1.25). Interleukin gene variation, particularly the IL-6 rs1800795 variant, is modestly associated with obesity risk. This suggests that other factors, such as the environment, also play a role in obesity. Thus, individuals with this particular IL-6 variant may have a slightly higher likelihood of being overweight or obese compared to those without it, but this is just one of many factors influencing obesity risk.
Collapse
Affiliation(s)
- Aleksandra Bojarczuk
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (C.Ż.); (B.B.); (P.C.)
| | - Aleksandra Garbacz
- Faculty of Animal Genetics and Conservation, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Cezary Żekanowski
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (C.Ż.); (B.B.); (P.C.)
| | - Beata Borzemska
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (C.Ż.); (B.B.); (P.C.)
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Paweł Cięszczyk
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (C.Ż.); (B.B.); (P.C.)
| | - Ewelina Maculewicz
- Faculty of Physical Education, Jozef Pilsudski University of Physical Education in Warsaw, 00-968 Warsaw, Poland;
- Department of Laboratory Diagnostics, Military Institute of Aviation Medicine, 01-755 Warsaw, Poland
| |
Collapse
|
13
|
Geng C, Chen C. Association between inflammatory cytokines and disease severity in restless legs syndrome. Neurol Sci 2024; 45:5765-5770. [PMID: 39198357 DOI: 10.1007/s10072-024-07736-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND This study aims to evaluate serum cytokine levels in individuals with primary Restless Legs Syndrome (RLS) and analyze their correlation with RLS symptom severity, sleep disturbances, anxiety, and depression. METHODS A case-control study was conducted involving 87 primary RLS patients and 100 healthy individuals undergoing health check-ups. Serum cytokine levels were assessed, and RLS severity was evaluated using the International Restless Legs Syndrome Study Group (IRLSSG) rating scale. Additionally, the Pittsburgh Sleep Quality Index (PSQI), 24-item Hamilton Depression Rating Scale (HAMD24), and 14-item Hamilton Anxiety Rating Scale (HAMA14) were utilized to assess symptom severity, sleep disturbances, and emotional states in RLS patients. RESULTS Serum cytokine levels, including IL-6 and TNF-α, were significantly higher in RLS patients compared to controls (P < 0.05). Adjustment for disease duration, sex, age, BMI, SBP, and DBP revealed an independent association between serum IL-6 and RLS (OR = 1.46; 95% CI = 1.02-1.93; P = 0.003). Linear regression analysis demonstrated a significant correlation between IL-6 levels and IRLS scores (β = 0.123, P = 0.008). CONCLUSION Elevated circulating levels of inflammatory cytokines in RLS patients suggest a potential role of inflammation in the pathogenesis of primary RLS. Additionally, serum cytokine levels correlate closely with disease severity.
Collapse
Affiliation(s)
- Chaofan Geng
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Chen Chen
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China.
- Department of Neurology, Zhengzhou University people's hospital, Henan provincial people's hospital, 7 Weiwu Street, 450000, Zhengzhou, China.
| |
Collapse
|
14
|
Li C, Wang K, Wang C, Li J, Zhang Q, Song L, Wu Z, Zhang S. A glucose-rich heteropolysaccharide from Marsdenia tenacissima (Roxb.) Wight et Arn. and its zinc-modified complex enhance immunoregulation by regulating TLR4-Myd88-NF-κB pathway. Int J Biol Macromol 2024; 283:137529. [PMID: 39537046 DOI: 10.1016/j.ijbiomac.2024.137529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/15/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
A previously unreported immunological polysaccharide (MTP70-1) was obtained from Marsdenia tenacissima (Roxb.) Wight et Arn. MTP70-1 (2738 Da) is a heteropolysaccharide that mainly consists of (1 → 5)-linked-L-Araf, t-D-Glcp, (1 → 3,5)-linked-L-Araf, (1 → 4)-linked-D-Galp, (1 → 6)-linked-D-Glcp, and (1 → 3,6)-linked-D-Manp. In vitro cell assays revealed that MTP70-1 exhibits moderate immunomodulatory effects at the cellular level, and MTP70-1 was further modified with zinc to improve these effects. These modifications enhanced the immunomodulatory effects of MTP70-1, as phagocytosis was enhanced, the secretion of cytokines (TNF-α, IL-6, IL-1β, and IL-18) was increased, and the generation of chemokines (NO and ROS) in macrophages was enhanced. The intracellular mechanism by which MTP70-1 and MTP70-Zn activate macrophages was further revealed to be closely related to the TLR4-Myd88-NF-κB signaling pathway. In addition, a microscale thermophoresis binding (MST) assay confirmed that Zn modification can effectively enhance the binding affinity of MTP70-1 for TLR4. Ultimately, better immune-enhancing activity was attained with MTP70-Zn than MTP70-1. The immune-enhancing activity of MTP70-Zn was further demonstrated through zebrafish assays, which revealed that MTP70-Zn can effectively enhance the proliferation of macrophages and neutrophils.
Collapse
Affiliation(s)
- Chong Li
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, Tianjin 300350, China
| | - Cancan Wang
- Medical College, Guangxi University, Nanning 530004, China
| | - Junhao Li
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Qian Zhang
- School of Pharmacy, Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijun Song
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Zhongnan Wu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Shaojie Zhang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
15
|
Dani C, Tarchi L, Cassioli E, Rossi E, Merola GP, Ficola A, Cordasco VZ, Ricca V, Castellini G. A transdiagnostic and diagnostic-specific approach on inflammatory biomarkers in eating disorders: A meta-analysis and systematic review. Psychiatry Res 2024; 340:116115. [PMID: 39128168 DOI: 10.1016/j.psychres.2024.116115] [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: 04/16/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024]
Abstract
Eating disorders (EDs) are severe mental illnesses with a multifactorial etiology and a chronic course. Among the biological factors related to pathogenesis and maintenance of EDs, inflammation acquired growing scientific interest. This study aimed to assess the inflammatory profile of EDs, focusing on anorexia nervosa, bulimia nervosa, and including for the first time binge eating disorder. A comprehensive research of existing literature identified 51 eligible studies for meta-analysis, comparing levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), C-reactive protein (CRP), osteoprotegerin (OPG), soluble receptor activator of nuclear factor kappa-B ligand (sRANKL), interleukin-1β (IL-1β), and interleukin-10 (IL-10) between patients with EDs and healthy controls (HCs). The systematic review explored other inflammatory biomarkers of interest, which did not meet the meta-analysis criteria. Results revealed significantly elevated levels of TNF-α, OPG, sRANKL, and IL-1β in patients with EDs compared to HCs. Additionally, the results highlighted the heterogeneity of inflammatory state among patients with EDs, emphasizing the need for further research into the association between inflammatory biomarkers and psychopathological correlates. This approach should transcend categorical diagnoses, enabling more precise subcategorizations of patients. Overall, this study contributed to the understanding of the inflammatory pathways involved in EDs, emphasizing potential implications for diagnosis, staging, and targeted interventions.
Collapse
Affiliation(s)
- Cristiano Dani
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Livio Tarchi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Emanuele Cassioli
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Eleonora Rossi
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Arianna Ficola
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Valdo Ricca
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
16
|
Bernardoni F, Tam F, Poitz DM, Hellerhoff I, Arold D, Geisler D, Lemme F, Keeler J, Weidner K, Pariante C, Roessner V, King JA, Ehrlich S. Effect of serum concentrations of IL-6 and TNF-α on brain structure in anorexia nervosa: a combined cross-sectional and longitudinal study. Neuropsychopharmacology 2024; 49:1509-1517. [PMID: 38461330 PMCID: PMC11319803 DOI: 10.1038/s41386-024-01836-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/11/2024]
Abstract
Previous studies of brain structure in anorexia nervosa (AN) have reported reduced gray matter in underweight patients, which largely normalizes upon weight gain. One underlying biological mechanism may be glial cell alterations related to low-grade inflammation. Here, we investigated relationships between brain structure as measured by magnetic resonance imaging and serum concentrations of two pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor alpha) cross-sectionally in 82 underweight adolescent and young adult female patients (mean age 16.8 years; 59 of whom were observed longitudinally after short-term weight restoration; mean duration 2.8 months), 20 individuals long-term weight-recovered from AN (mean age 22.7 years) and 105 healthy control (HC) participants (mean age 17.2 years). We measured cortical thickness, subcortical volumes and local gyrification index, a measure of cortical folding. In contrast to most previous studies of cytokine concentrations in AN, we found no cross-sectional group differences (interleukin-6: p = 0.193, tumor necrosis factor alpha: p = 0.057) or longitudinal changes following weight restoration (interleukin-6: p = 0.201, tumor necrosis factor alpha: p = 0.772). As expected, widespread gray matter reductions (cortical thickness, subcortical volumes, cortical folding) were observed in underweight patients with AN compared to HC. However, we found no evidence of associations between cytokine concentrations and structural brain measures in any participant group. Furthermore, longitudinal changes in cytokine concentrations were unrelated to changes in gray matter. In conclusion, we did not identify any association between (sub-)inflammatory processes and structural brain changes in AN. Future studies are needed to elucidate which other factors besides nutritional status may contribute to brain morphological alterations.
Collapse
Affiliation(s)
- Fabio Bernardoni
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Friederike Tam
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - David M Poitz
- University Hospital Carl Gustav Carus, Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Inger Hellerhoff
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Dominic Arold
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Daniel Geisler
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Frances Lemme
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Johanna Keeler
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Kerstin Weidner
- Department of Psychotherapy and Psychosomatic Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Joseph A King
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
- Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
17
|
Miaskowski C, Conley YP, Levine JD, Cooper BA, Paul SM, Hammer MJ, Oppegaard K, Harris C, Shin J, Abrams G, Asakitogum D, Fu MR, Alismal S. Chronic Decrements in Energy in Women with Breast Cancer are Associated with Cytokine Gene Polymorphisms. Semin Oncol Nurs 2024; 40:151652. [PMID: 38834449 DOI: 10.1016/j.soncn.2024.151652] [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: 01/31/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 06/06/2024]
Abstract
OBJECTIVES Decrements in energy were found in 67% of women who underwent breast cancer surgery. However, no information is available on chronic decrements in energy and associations with inflammation. Purposes were to identify latent classes of patients with distinct average energy profiles from prior to through 12 months after breast cancer surgery; evaluate for differences in demographic and clinical characteristics between the two extreme average energy classes; and evaluate for polymorphisms for cytokine genes associated with membership in the Low energy class. METHODS Women (n = 397) completed assessments of energy prior to and for 12 months following breast cancer surgery. Growth mixture modeling was used to identify classes of patients with distinct average energy profiles. Eighty-two single nucleotide polymorphisms (SNPs) among 15 cytokine genes were evaluated. RESULTS Three distinct energy profiles were identified (ie, Low [27.0%], Moderate [54.4%], Changing [18.6%]). Data from patients in the Low and Moderate energy classes were used in the candidate gene analyses. Five SNPs and one haplotype in six different genes remained significant in logistic regression analyses (ie, interleukin [IL]-1β rs1143623, IL1 receptor 1 rs3917332 IL4 rs2243263, IL6 HapA1 [that consisted of rs1800795, rs2069830, rs2069840, rs1554606, rs2069845, rs2069849, and rs2069861], nuclear factor kappa beta subunit 1 rs170731, tumor necrosis factor rs1799964). For several SNPs for IL6, expression quantitative trait locis were identified in subcutaneous and visceral adipose tissue and thyroid tissue. In addition, skeletal muscle was identified as an expression quantitative trait loci for nuclear factor kappa beta subunit 1. CONCLUSIONS Findings suggest that cytokine genes are involved in the mechanisms that underlie chronic decrements in energy in women following breast cancer surgery. Given the roles of subcutaneous and visceral adipose and thyroid tissues in metabolism and energy balance, the findings related to IL6 suggest that these polymorphisms may have a functional role in the development and maintenance of chronic decrements in energy.
Collapse
Affiliation(s)
- Christine Miaskowski
- School of Nursing, University of California, San Francisco; School of Medicine, University of California, San Francisco.
| | | | - Jon D Levine
- School of Medicine, University of California, San Francisco
| | - Bruce A Cooper
- School of Nursing, University of California, San Francisco
| | - Steven M Paul
- School of Nursing, University of California, San Francisco
| | | | | | - Carolyn Harris
- School of Nursing, University of Pittsburgh, Pittsburgh, PA
| | | | - Gary Abrams
- School of Medicine, University of California, San Francisco
| | | | - Mei R Fu
- University of Missouri, Kansas City
| | - Sarah Alismal
- Beckman Research Institute, City of Hope, Duarte, CA
| |
Collapse
|
18
|
Fisher A, Fisher L, Srikusalanukul W. Prediction of Osteoporotic Hip Fracture Outcome: Comparative Accuracy of 27 Immune-Inflammatory-Metabolic Markers and Related Conceptual Issues. J Clin Med 2024; 13:3969. [PMID: 38999533 PMCID: PMC11242639 DOI: 10.3390/jcm13133969] [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: 06/11/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Objectives: This study, based on the concept of immuno-inflammatory-metabolic (IIM) dysregulation, investigated and compared the prognostic impact of 27 indices at admission for prediction of postoperative myocardial injury (PMI) and/or hospital death in hip fracture (HF) patients. Methods: In consecutive HF patient (n = 1273, mean age 82.9 ± 8.7 years, 73.5% females) demographics, medical history, laboratory parameters, and outcomes were recorded prospectively. Multiple logistic regression and receiver-operating characteristic analyses (the area under the curve, AUC) were used to establish the predictive role for each biomarker. Results: Among 27 IIM biomarkers, 10 indices were significantly associated with development of PMI and 16 were indicative of a fatal outcome; in the subset of patients aged >80 years with ischaemic heart disease (IHD, the highest risk group: 90.2% of all deaths), the corresponding figures were 26 and 20. In the latter group, the five strongest preoperative predictors for PMI were anaemia (AUC 0.7879), monocyte/eosinophil ratio > 13.0 (AUC 0.7814), neutrophil/lymphocyte ratio > 7.5 (AUC 0.7784), eosinophil count < 1.1 × 109/L (AUC 0.7780), and neutrophil/albumin × 10 > 2.4 (AUC 0.7732); additionally, sensitivity was 83.1-75.4% and specificity was 82.1-75.0%. The highest predictors of in-hospital death were platelet/lymphocyte ratio > 280.0 (AUC 0.8390), lymphocyte/monocyte ratio < 1.1 (AUC 0.8375), albumin < 33 g/L (AUC 0.7889), red cell distribution width > 14.5% (AUC 0.7739), and anaemia (AUC 0.7604), sensitivity 88.2% and above, and specificity 85.1-79.3%. Internal validation confirmed the predictive value of the models. Conclusions: Comparison of 27 IIM indices in HF patients identified several simple, widely available, and inexpensive parameters highly predictive for PMI and/or in-hospital death. The applicability of IIM biomarkers to diagnose and predict risks for chronic diseases, including OP/OF, in the preclinical stages is discussed.
Collapse
Affiliation(s)
- Alexander Fisher
- Department of Geriatric Medicine, The Canberra Hospital, ACT Health, Canberra 2605, Australia
- Department of Orthopaedic Surgery, The Canberra Hospital, ACT Health, Canberra 2605, Australia
- Medical School, Australian National University, Canberra 2601, Australia
| | - Leon Fisher
- Frankston Hospital, Peninsula Health, Melbourne 3199, Australia
| | - Wichat Srikusalanukul
- Department of Geriatric Medicine, The Canberra Hospital, ACT Health, Canberra 2605, Australia
| |
Collapse
|
19
|
Wang L, Li Y, Li R, Luan J, Cao K, Liu T, Hu H, Chen S, Bu L, Liu L, Wang H, Lu Q. Diverse associations between pancreatic intra-, inter-lobular fat and the development of type 2 diabetes in overweight or obese patients. Front Nutr 2024; 11:1421032. [PMID: 39021593 PMCID: PMC11252058 DOI: 10.3389/fnut.2024.1421032] [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: 04/21/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Pancreatic fat is associated with obesity and type 2 diabetes mellitus (T2DM); however, the relationship between different types of pancreatic fat and diabetes status remains unclear. Therefore, we aimed to determine the potential of different types of pancreatic fat accumulation as a risk factor for T2DM in overweight or obese patients. In total, 104 overweight or obese patients were recruited from January 2020 to December 2022. The patients were divided into three groups: normal glucose tolerance (NGT), impaired fasting glucose or glucose tolerance (IFG/IGT), and T2DM. mDixon magnetic resonance imaging (MRI) was used to detect pancreatic fat in all three groups of patients. The pancreatic head fat (PHF), body fat (PBF), and tail fat (PTF) in the IFG/IGT group were 21, 20, and 31% more than those in the NGT group, respectively. PHF, PBF, and PTF were positively associated with glucose metabolic dysfunction markers in the NGT group, and inter-lobular fat volume (IFV) was positively associated with these markers in the IFG/IGT group. The areas under the receiver operating characteristic curves for PHF, PBF, and PTF (used to evaluate their diagnostic potential for glucose metabolic dysfunction) were 0.73, 0.73, and 0.78, respectively, while those for total pancreatic volume (TPV), pancreatic parenchymal volume, IFV, and IFV/TPV were 0.67, 0.67, 0.66, and 0.66, respectively. These results indicate that intra-lobular pancreatic fat, including PHF, PTF, and PBF, may be a potential independent risk factor for the development of T2DM. Additionally, IFV exacerbates glucose metabolic dysfunction. Intra-lobular pancreatic fat indices were better than IFV for the diagnosis of glucose metabolic dysfunction.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yinghao Li
- Physics Department & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Renfeng Li
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Jinwen Luan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kaiming Cao
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Tiancheng Liu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Haiyang Hu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Shanshan Chen
- College of Medical Imaging, Shanghai University of Medicine and Health Science, Shanghai, China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Medicine School of Tongji University, Shanghai, China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hongzhi Wang
- Physics Department & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Qing Lu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Yang L, Liang Y, Pu J, Cai L, Gao R, Han F, Chang K, Pan S, Wu Z, Zhang Y, Wang Y, Song J, Wu H, Tang J, Wang X. Dysregulated serum lipid profile is associated with inflammation and disease activity in primary Sjögren's syndrome: a retrospective study in China. Immunol Lett 2024; 267:106865. [PMID: 38705483 DOI: 10.1016/j.imlet.2024.106865] [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] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE To investigate the relationship between the lipid profiles of patients with primary Sjögren's syndrome (pSS) and other clinical characteristics, laboratory examination, disease activity, and inflammatory factors. In addition, the risk factors for hyperlipidemia-related complications of pSS and the effect of hydroxychloroquine (HCQ) usage on the lipid profile were incorporated into this study. METHODS This is a single-center, retrospective study that included 367 patients who were diagnosed with pSS at Tongji Hospital, School of Medicine, Tongji University, China from January 2010 to March 2022. Initially, demographic information, clinical characteristics, medication records, and complications of the patients were gathered. A case-control analysis compared the 12 systems involvement (ESSDAI domain), clinical symptoms, and laboratory tests between pSS patients with and without dyslipidemia. A simple linear regression model was employed to investigate the relationship between serum lipid profile and inflammatory factors. Logistics regression analysis was performed to assess variables for hyperlipidemia-related complications of pSS. The paired t-test was then used to evaluate the improvement in lipid profile among pSS patients. RESULTS 48.7 % of all pSS patients had dyslipidemia, and alterations in lipid levels were related to gender, age, and smoking status but not body mass index (BMI). Dyslipidemia is more prevalent in pSS patients who exhibit heightened autoimmunity and elevated levels of inflammation. Higher concentrations of multiple highly inflammatory factors correlate with a more severe form of dyslipidemia. Non-traditional cardiovascular risk factors may contribute to hyperlipidemia-related complications of pSS, such as increased, low complement 3 (C3) and low C4. According to our study, HCQ usage may protect against lipid-related disease in pSS. CONCLUSION Attention should be paid to the dyslipidemia of pSS. This research aims to clarify the population portrait of pSS patients with abnormal lipid profiles and provides insights into the correlation between metabolism and inflammation in individuals with pSS and the potential role they play in the advancement of the disease. These findings provide novel avenues for further understanding the underlying mechanisms of pSS pathogenesis.
Collapse
Affiliation(s)
- Lufei Yang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yuanyuan Liang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jincheng Pu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Li Cai
- Department of Science and Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ronglin Gao
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Fang Han
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Keni Chang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shengnan Pan
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Youwei Zhang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yanqing Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jiamin Song
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Huihong Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jianping Tang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China..
| | - Xuan Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China..
| |
Collapse
|
21
|
Brooks SJ, Dahl K, Dudley-Jones R, Schiöth HB. A neuroinflammatory compulsivity model of anorexia nervosa (NICAN). Neurosci Biobehav Rev 2024; 159:105580. [PMID: 38417395 DOI: 10.1016/j.neubiorev.2024.105580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024]
Affiliation(s)
- S J Brooks
- Department of Surgical Sciences, Uppsala University, Sweden; School of Psychology, Liverpool John Moores University, UK; Neuroscience Research Laboratory (NeuRL), Department of Psychology, School of Human and Community Development, University of the Witwatersrand, Johannesburg, South Africa.
| | - K Dahl
- Department of Surgical Sciences, Uppsala University, Sweden
| | - R Dudley-Jones
- School of Psychology, Liverpool John Moores University, UK
| | - H B Schiöth
- Department of Surgical Sciences, Uppsala University, Sweden
| |
Collapse
|
22
|
Zhao L, Chang F, Tong Y, Yin J, Xu J, Li H, Du L, Jiang Y. A Multifunctional Bimetallic Nanoplatform for Synergic Local Hyperthermia and Chemotherapy Targeting HER2-Positive Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308316. [PMID: 38380506 PMCID: PMC11040336 DOI: 10.1002/advs.202308316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/19/2024] [Indexed: 02/22/2024]
Abstract
Anti-HER2 (human epidermal growth factor receptor 2) therapies significantly increase the overall survival of patients with HER2-positive breast cancer. Unfortunately, a large fraction of patients may develop primary or acquired resistance. Further, a multidrug combination used to prevent this in the clinic places a significant burden on patients. To address this issue, this work develops a nanotherapeutic platform that incorporates bimetallic gold-silver hollow nanoshells (AuAg HNSs) with exceptional near-infrared (NIR) absorption capability, the small-molecule tyrosine kinase inhibitor pyrotinib (PYR), and Herceptin (HCT). This platform realizes targeted delivery of multiple therapeutic effects, including chemo-and photothermal activities, oxidative stress, and immune response. In vitro assays reveal that the HCT-modified nanoparticles exhibit specific recognition ability and effective internalization by cells. The released PYR inhibit cell proliferation by downregulating HER2 and its associated pathways. NIR laser application induces a photothermal effect and tumor cell apoptosis, whereas an intracellular reactive oxygen species burst amplifies oxidative stress and triggers cancer cell ferroptosis. Importantly, this multimodal therapy also promotes the upregulation of genes related to TNF and NF-κB signaling pathways, enhancing immune activation and immunogenic cell death. In vivo studies confirm a significant reduction in tumor volume after treatment, substantiating the potential effectiveness of these nanocarriers.
Collapse
Affiliation(s)
- Li Zhao
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061China
| | - Fei Chang
- The Second Hospital of Shandong UniversityJinanShandong250033China
| | - Yao Tong
- The Second Hospital of Shandong UniversityJinanShandong250033China
| | - Jiawei Yin
- The Second Hospital of Shandong UniversityJinanShandong250033China
| | - Jiawen Xu
- Department of PathologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanShandong250021China
| | - Hui Li
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061China
| | - Lutao Du
- Department of Clinical LaboratoryQilu Hospital of Shandong UniversityJinanShandong250012China
- Shandong Provincial Key Laboratory of Innovation Technology in Laboratory MedicineJinanShandong250033China
- Shandong Provincial Clinical Medicine Research Center for Clinical LaboratoryJinanShandong250033China
| | - Yanyan Jiang
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061China
| |
Collapse
|
23
|
Cezar R, Kundura L, André S, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. T4 apoptosis in the acute phase of SARS-CoV-2 infection predicts long COVID. Front Immunol 2024; 14:1335352. [PMID: 38235145 PMCID: PMC10791767 DOI: 10.3389/fimmu.2023.1335352] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Background As about 10% of patients with COVID-19 present sequelae, it is important to better understand the physiopathology of so-called long COVID. Method To this aim, we recruited 29 patients hospitalized for SARS-CoV-2 infection and, by Luminex®, quantified 19 soluble factors in their plasma and in the supernatant of their peripheral blood mononuclear cells, including inflammatory and anti-inflammatory cytokines and chemokines, Th1/Th2/Th17 cytokines, and endothelium activation markers. We also measured their T4, T8 and NK differentiation, activation, exhaustion and senescence, T cell apoptosis, and monocyte subpopulations by flow cytometry. We compared these markers between participants who developed long COVID or not one year later. Results None of these markers was predictive for sequelae, except programmed T4 cell death. T4 lymphocytes from participants who later presented long COVID were more apoptotic in culture than those of sequelae-free participants at Month 12 (36.9 ± 14.7 vs. 24.2 ± 9.0%, p = 0.016). Conclusions Our observation raises the hypothesis that T4 cell death during the acute phase of SARS-CoV-2 infection might pave the way for long COVID. Mechanistically, T4 lymphopenia might favor phenomena that could cause sequelae, including SARS-CoV-2 persistence, reactivation of other viruses, autoimmunity and immune dysregulation. In this scenario, inhibiting T cell apoptosis, for instance, by caspase inhibitors, could prevent long COVID.
Collapse
Affiliation(s)
- Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| | - Sonia André
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Ahn Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
- Laval University Research Center, Quebec City, QC, Canada
| | - Pierre Corbeau
- Immunology Department, Nîmes University Hospital, Nîmes, France
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| |
Collapse
|
24
|
Liu Q, Bode AM, Chen X, Luo X. Metabolic reprogramming in nasopharyngeal carcinoma: Mechanisms and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:189023. [PMID: 37979733 DOI: 10.1016/j.bbcan.2023.189023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
The high prevalence of metabolic reprogramming in nasopharyngeal carcinoma (NPC) offers an abundance of potential therapeutic targets. This review delves into the distinct mechanisms underlying metabolic reprogramming in NPC, including enhanced glycolysis, nucleotide synthesis, and lipid metabolism. All of these changes are modulated by Epstein-Barr virus (EBV) infection, hypoxia, and tumor microenvironment. We highlight the role of metabolic reprogramming in the development of NPC resistance to standard therapies, which represents a challenging barrier in treating this malignancy. Furthermore, we dissect the state of the art in therapeutic strategies that target these metabolic changes, evaluating the successes and failures of clinical trials and the strategies to tackle resistance mechanisms. By providing a comprehensive overview of the current knowledge and future directions in this field, this review sets the stage for new therapeutic avenues in NPC.
Collapse
Affiliation(s)
- Qian Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China.
| | - Xiangjian Luo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|