1
|
Feng Y, Wu Z, Hu K, Yuan S, Li J, Wang Y, Wang Z, Yang H, Luo ZH, Zhou J. Inflammatory metabolite 7α,25-OHC promotes TIMP1 expression in COVID-19 monocytes through synergy effect of SMARCC1/JUND/H3K27ac. Cell Mol Life Sci 2025; 82:208. [PMID: 40399563 PMCID: PMC12095718 DOI: 10.1007/s00018-025-05721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/24/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025]
Abstract
Chromatin remodeling factors are involved in the inflammatory responses, contributing to tissue damage and multi-organ dysfunction in COVID-19 patients. However, the underlying mechanisms remain unclear. In this study, high-dimensional analyses of single-cell RNA sequencing and single-cell ATAC sequencing data revealed increased chromatin accessibility at the promoters or enhancers of the pro-inflammatory cytokine tissue inhibitor of metalloproteinase-1 (TIMP1), as well as altered gene transcription profiles in monocytes from COVID-19 patients. Motif enrichment and positive regulators analyses identified SMARCC1, the core subunit of the chromatin remodeling complex, and the transcription factor JUND as positive regulators to co-modulate TIMP1 expression. In-vitro experiments, co-immunoprecipitation and chromatin immunoprecipitation (ChIP)-qPCR, and others, demonstrated the collaboration of SMARCC1 and JUND. Increased 7α,25-dihydroxycholesterol (7α,25-OHC) enhanced SMARCC1-JUND interactions to co-regulate TIMP1 expression. Further investigation indicated that 7α,25-OHC promoted the expression of SMARCC1 and its co-localization with H3K27ac, which involved in the expression of TIMP1 and inflammatory responses. Our study highlights the critical roles of SMARCC1 and JUND in COVID-19 inflammation, and offers the potential targets for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Ying Feng
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zheng Wu
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Kefan Hu
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Shenzhen Yuan
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jun Li
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhongyi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Han Yang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Zhi-Hui Luo
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Jingjiao Zhou
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Zhao L, Du GL, Ruze A, Qi HZ, Zhang CS, Li QL, Deng AX, Zhao BH, Hu S, Gai MT, Gao XM. Novel function of macrophage migration inhibitory factor in regulating post-infarct inflammation and the therapeutic significance. J Adv Res 2025:S2090-1232(25)00348-0. [PMID: 40383291 DOI: 10.1016/j.jare.2025.05.030] [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: 02/04/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
INTRODUCTION Recent studies indicate that macrophage migration inhibitory factor (MIF) has a dual role in myocardial infarction (MI), with different cellular sources of MIF influencing inflammation and healing differentially. OBJECTIVES To investigate the role and underlying mechanism of MIF in MI and interventional efficacy targeting MIF. METHODS Wild-type (WT), global MIF gene knockout (KO) and chimeric mice were subjected to coronary artery occlusion. The inflammatory responses and healing processes following MI were studied in both in vivo and in vitro settings. Furthermore, the therapeutic potential of pharmacological MIF inhibition to improve the prognosis of MI was explored. RESULTS Globally, MIF enhanced systemic and local inflammatory responses, as well as splenic monocyte mobilization, in mice with MI. MIF promoted monocyte migration through CCR2 and CXCR4 in peripheral blood mononuclear cells (PBMCs) and the infarcted myocardium. Additionally, MIF augmented angiotensin Ⅱ type 1 receptor (AT-1R) expression and interacted with AT-1R to promote the splenic monocyte mobilization following acute MI. MIF derived from bone marrow cells (KOWT mice) had stronger systemic and local inflammatory responses and augmented mobilization of splenic monocytes. In contrast, deficiency of MIF in leukocytes (WTKO mice) increased Ly-6Clow monocyte accumulation, M2 macrophage infiltration, and degree of myocardial fibrosis in infarcted myocardium. In vitro, MIF derived from ischemic heart enhanced M2 but impaired M1 macrophage marker expression in PBMCs. Anti-MIF treatment effectively attenuated splenic monocyte mobilization and both systemic and regional inflammatory responses post-MI without affecting the healing process, thereby improving the long-term prognosis. CONCLUSION Deletion of global and inflammatory-cell-derived MIF diminished inflammation following MI by inhibiting monocyte mobilization and downregulating pro-inflammatory mediators, while cardiac-derived MIF exerted anti-inflammatory influence and facilitated healing. Furthermore, MIF antibody therapy protected the heart from severe ischemic injury and improved long-term prognosis.
Collapse
Affiliation(s)
- Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China; Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Hong-Zhi Qi
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chuan-Shan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Su Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Min-Tao Gai
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China.
| |
Collapse
|
3
|
Wang S, Xiao R, Chen Y, Ye Y, He T, Yang Y, Chen X, Chou CK. Anti-tumor necrosis factor therapy in the treatment of systemic autoinflammatory diseases: the responses of innate immune cells. J Leukoc Biol 2025; 117:qiaf026. [PMID: 40084825 DOI: 10.1093/jleuko/qiaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/09/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025] Open
Abstract
Systemic autoinflammatory diseases are rare conditions resulting from dysregulation of the innate immune system, culminating in repetitive bouts of systemic inflammation without the presence of external or self-antigens. Most systemic autoinflammatory diseases are associated with mutations in genes affecting the innate immune response. Tumor necrosis factor is a central player in the pathogenesis of numerous chronic inflammatory disorders, and anti-tumor necrosis factor therapy is widely used in the clinical management of systemic autoinflammatory diseases. Tumor necrosis factor inhibitors block the interaction of tumor necrosis factor with its 2 receptors, tumor necrosis factor receptor 1 and tumor necrosis factor receptor 2. These inhibitors primarily target soluble tumor necrosis factor, which mainly binds to tumor necrosis factor receptor 1, exerting anti-inflammatory effects. Interestingly, tumor necrosis factor inhibitors also affect transmembrane tumor necrosis factor, which engages tumor necrosis factor receptor 2 to initiate reverse signaling. This reverse signaling can activate innate immune cells, prevent apoptosis, or paradoxically inhibit the production of pro-inflammatory cytokines. Tumor necrosis factor inhibitors also promote the release of soluble tumor necrosis factor receptor 2, which neutralizes circulating tumor necrosis factor. Some agents targeting tumor necrosis factor receptor 2 can even act as agonists, triggering reverse signaling by binding to transmembrane tumor necrosis factor. While effective, prolonged use of tumor necrosis factor inhibitors may cause significant side effects due to the widespread expression and pleiotropic functions of tumor necrosis factor receptors. A more thorough understanding of the mechanisms underlying the action of tumor necrosis factor inhibitors is required to develop a more effective and safer treatment for systemic autoinflammatory diseases. This article reviews current studies on the role of the innate immune system in systemic autoinflammatory disease pathogenesis, the impact of anti-tumor necrosis factor therapy on innate immune cells, and perspectives on developing improved agents targeting tumor necrosis factor or its receptors.
Collapse
Affiliation(s)
- Shuyi Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
| | - Rufei Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
| | - Yibo Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
| | - Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, P. R. China
| | - Tianzhen He
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, P. R. China
| | - Yang Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
| | - Xin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
- Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau 999078, P. R. China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, P. R. China
| | - Chon-Kit Chou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau 999078, P. R. China
| |
Collapse
|
4
|
Wang Q, Wang S, Cui L, Zhang Y, Waterhouse GIN, Sun-Waterhouse D, Ma C, Kang W. Flammulina velutipes polysaccharide exerts immunomodulatory function involving RSAD2 to regulate the NF-κB/MAPK signaling pathway in RAW264.7 macrophage cells and in mouse spleen cells. Int J Biol Macromol 2025; 309:142985. [PMID: 40210026 DOI: 10.1016/j.ijbiomac.2025.142985] [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: 01/14/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
There are ongoing efforts to explore the potential of natural bioactive substances including polysaccharides in immunological regulation and understand the mechanisms under their immune-regulating function. In this study, a polysaccharide from Flammulina velutipes (FVP-1) exhibited immunomodulatory in RAW264.7 macrophage cells and mouse spleen cells. FVP-1 increased the secretion of cytokines (like TNF-α, IL-6 and IL-1β) and their mRNA expression, upregulated the transcription and translation expression of COX-2 and iNOS, and enhanced the release of reactive oxygen species the phagocytic activity in macrophages, thereby promoting the maturation and transformation of certain lymphocytes. All these functions of FVP-1 depended to some extent on its concentration. The RSAD2 effector was involved in the immunomodulatory function of FVP-1 towards macrophages and mouse splenocytes, through mediating FVP-1's activation and regulation of the NF-κB/MAPK signaling pathway. These findings indicate the potential of FVP-1 as a natural immunomodulator and approach for improving immune function.
Collapse
Affiliation(s)
- Qiuyi Wang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Senye Wang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Lili Cui
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China
| | - Yu Zhang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Geoffrey I N Waterhouse
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; School of Chemical Sciences, the University of Auckland, Auckland 1142, New Zealand
| | - Dongxiao Sun-Waterhouse
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; School of Chemical Sciences, the University of Auckland, Auckland 1142, New Zealand.
| | - Changyang Ma
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; Functional Food Engineering Technology Research Center, Kaifeng 475004, China; College of Agriculture, Henan University, Kaifeng 475004, China.
| | - Wenyi Kang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; Functional Food Engineering Technology Research Center, Kaifeng 475004, China; College of Agriculture, Henan University, Kaifeng 475004, China.
| |
Collapse
|
5
|
Mabatha KC, Letuka P, Aremu O, Zulu MZ. Macrophages of the Heart: Homeostasis and Disease. Biomed J 2025:100867. [PMID: 40300670 DOI: 10.1016/j.bj.2025.100867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025] Open
Abstract
Cardiac macrophages (CMs) are the most abundant immune cell type in the heart. They are critical for maintaining cardiac homeostasis and in the orchestration of immune responses to ischemic and non-ischemic cardiomyopathies. Their functions are highly heterogeneous and regulated by their tissue microenvironment. CMs have high plasticity, which allows them to perform various functions in the myocardium to bring about homeostasis within the cardiovascular system (CVS). CMs also play critical roles in coronary development and angiogenesis, tissue repair and remodeling, cardiac conduction and in the clearance of necrotic and apoptotic cells. However, there is a paucity of studies on the biology of cardiac macrophages in both steady state and disease, especially, in humans. In this review, we discuss the multifaceted roles of CMs in the heart, focusing on their ontogeny, homeostatic functions and immunological responses during inflammation and reparative processes post-injury. We highlight the heterogeneity of CMs in their ontogeny, phenotypes and functions as well as their roles in the pathogenesis of pathological conditions such as myocarditis, myocardial fibrosis and heart failure. Understanding the unique characteristics of cardiac macrophages in the cardiac milieu is critical for the development of macrophage-specific therapeutic interventions to alleviate the global burden of cardiovascular disease (CVD). Therefore, future studies should focus on further improving the understanding of the biology of cardiac macrophages to harness their potential as therapeutic targets for cardiovascular disorders.
Collapse
Affiliation(s)
- Koketso C Mabatha
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; SAMRC Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Pheletso Letuka
- SAMRC Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa; Division of Cardiology, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Olukayode Aremu
- SAMRC Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa; Division of Cardiology, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Michael Z Zulu
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; SAMRC Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
6
|
Liang L, Dang B, Ouyang X, Zhao X, Huang Y, Lin Y, Cheng X, Xie G, Lin J, Mi P, Ye Z, Guleng B, Cheng SC. Dietary succinate supplementation alleviates DSS-induced colitis via the IL-4Rα/Hif-1α Axis. Int Immunopharmacol 2025; 152:114408. [PMID: 40086056 DOI: 10.1016/j.intimp.2025.114408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/22/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
Inflammatory bowel disease (IBD) remains a pressing global health challenge, necessitating novel therapeutic strategies. Succinate, a metabolite known for its role in type 2 immunity and tuft cell activation in the small intestine, presents its potential in IBD management. However, its impact on colonic inflammation has not been explored. Here, we demonstrate that succinate administration induces a type 2 immune response, significantly alleviating dextran sulfate sodium (DSS)-induced colonic inflammation. Succinate enhances antibacterial capacity, reduces intestinal permeability, and reshapes the colonic cytokine milieu. Mechanistically, succinate promotes myeloid cell expansion in peripheral blood, mesenteric lymph nodes, and the colonic lamina propria. The protective effects of succinate were abolished in Ccr2-/- mice, confirming the role of monocyte recruitment, but persisted in Rag1-/- mice, indicating independence from adaptive immunity. Adoptive transfer of monocytes from succinate-treated donors mitigated intestinal inflammation in recipient mice. Transcriptomic analysis revealed heightened expression of Il1b and Il6, and higher lactate production in monocytes upon lipopolysaccharide (LPS) stimulation, highlighting a reprogrammed pro-inflammatory trained immunity phenotype. Finally, we identify the IL-4Rα/Hif-1α axis is critical for succinate-mediated protection. These findings reveal the ability of succinate to reprogram monocytes into protective intestinal macrophages via induction of type 2 response, restoring homeostasis through enhanced barrier function and immune modulation. Our study positions thus uncover succinate as a promising therapeutic candidate for IBD.
Collapse
Affiliation(s)
- Laiying Liang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; Department of Laboratory Medicine, West China Xiamen Hospital of Sichuan University, Xiamen 361000, China
| | - Buyun Dang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaomei Ouyang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xianling Zhao
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Yongdong Huang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xiaoshen Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Guijing Xie
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Junhui Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Peng Mi
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Zhenyu Ye
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China.
| | - Shih-Chin Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
7
|
Scafidi A, Lind-Holm Mogensen F, Michelucci A. Protocol for the generation and assessment of functional macrophages from mouse bone marrow cells. STAR Protoc 2025; 6:103706. [PMID: 40120113 PMCID: PMC11976235 DOI: 10.1016/j.xpro.2025.103706] [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: 10/30/2024] [Revised: 12/10/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Macrophages play essential roles in tissues, wherein they exert beneficial or detrimental functions depending on the signals they encounter during their differentiation. Here, we present a protocol to differentiate mouse bone marrow cells into macrophages under specific cues to evaluate their impact on macrophage phenotypic acquisition. We provide detailed instructions for optimal cell culture conditions, quality controls, and examples of concluding immunological functional assays. This protocol is applicable in short- and long-term drug-based modulation of macrophage functions. For complete details on the use and execution of this protocol, please refer to Scafidi et al.1.
Collapse
Affiliation(s)
- Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg.
| | - Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 1210 Luxembourg, Luxembourg.
| |
Collapse
|
8
|
Mir MM, Jeelani M, Alharthi MH, Rizvi SF, Sohail SK, Wani JI, Sabah ZU, BinAfif WF, Nandi P, Alshahrani AM, Alfaifi J, Jehangir A, Mir R. Unraveling the Mystery of Insulin Resistance: From Principle Mechanistic Insights and Consequences to Therapeutic Interventions. Int J Mol Sci 2025; 26:2770. [PMID: 40141412 PMCID: PMC11942988 DOI: 10.3390/ijms26062770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Insulin resistance (IR) is a significant factor in the development and progression of metabolic-related diseases like dyslipidemia, T2DM, hypertension, nonalcoholic fatty liver disease, cardiovascular and cerebrovascular disorders, and cancer. The pathogenesis of IR depends on multiple factors, including age, genetic predisposition, obesity, oxidative stress, among others. Abnormalities in the insulin-signaling cascade lead to IR in the host, including insulin receptor abnormalities, internal environment disturbances, and metabolic alterations in the muscle, liver, and cellular organelles. The complex and multifaceted characteristics of insulin signaling and insulin resistance envisage their thorough and comprehensive understanding at the cellular and molecular level. Therapeutic strategies for IR include exercise, dietary interventions, and pharmacotherapy. However, there are still gaps to be addressed, and more precise biomarkers for associated chronic diseases and lifestyle interventions are needed. Understanding these pathways is essential for developing effective treatments for IR, reducing healthcare costs, and improving quality of patient life.
Collapse
Affiliation(s)
- Mohammad Muzaffar Mir
- Department of Clinical Biochemistry, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohammed Jeelani
- Department of Physiology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Muffarah Hamid Alharthi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Syeda Fatima Rizvi
- Department of Pathology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (S.F.R.); (S.K.S.)
| | - Shahzada Khalid Sohail
- Department of Pathology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (S.F.R.); (S.K.S.)
| | - Javed Iqbal Wani
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia; (J.I.W.); (Z.U.S.)
| | - Zia Ul Sabah
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia; (J.I.W.); (Z.U.S.)
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Partha Nandi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Abdullah M. Alshahrani
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Adnan Jehangir
- Biomedical Sciences Department, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | - Rashid Mir
- Prince Fahd Bin Sultan Research Chair, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia;
| |
Collapse
|
9
|
Fu C, Mao Y. Association between pan-immune-inflammation value and heart failure: Evidence from the NHANES 2011-2020. J Int Med Res 2025; 53:3000605251325176. [PMID: 40079437 PMCID: PMC11907545 DOI: 10.1177/03000605251325176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/15/2025] [Indexed: 03/15/2025] Open
Abstract
ObjectiveThis study aimed to explore the association between pan-immune-inflammation value and heart failure outcomes using data from the nationally representative National Health and Nutrition Examination Survey database.MethodsWe conduct a cross-sectional cohort analysis using National Health and Nutrition Examination Survey data, including participants aged ≥20 years with available pan-immune-inflammation value data. We performed smooth curve fitting and threshold analysis, using both linear and non-linear regression models, to assess dose-response relationship and explore the continuous effect of pan-immune-inflammation value on heart failure outcomes.ResultsOur analysis revealed a significant and independent non-linear association between elevated pan-immune-inflammation value levels and an increased risk of heart failure occurrence. After adjustment for multiple covariates, these findings remained consistent and each increment unit in logarithmic pan-immune-inflammation value is associated with 34% increase in the risk of heart failure occurrence. Furthermore, we identified an inflection point of logarithmic pan-immune-inflammation value = 5.98 as a critical threshold. Stratified analyses revealed that the association between pan-immune-inflammation value and heart failure occurrence remains consistent across different subgroups.ConclusionsThis study confirmed the clinical value of pan-immune-inflammation value as a novel inflammatory biomarker in the assessment and monitoring of heart failure.
Collapse
Affiliation(s)
- Chen Fu
- Department of Cardiology, the First people’s Hospital of Xiaoshan District, Hangzhou, China
| | - Yijie Mao
- Department of Training, Hangzhou Xiaoshan No. 4 Secondary Vocational School, Hangzhou, China
| |
Collapse
|
10
|
Blokhina T, Kirichenko T, Markina Y, Khovantseva U, Melnikov I, Guseva O, Bazanovich S, Kozlov S, Orekhov A. Features of the monocyte inflammatory response in patients with premature coronary artery disease. BIOPHYSICS REPORTS 2025; 11:12-17. [PMID: 40070664 PMCID: PMC11891073 DOI: 10.52601/bpr.2024.240030] [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: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 03/14/2025] Open
Abstract
The purpose of this study was to examine the secretion of inflammatory cytokines by cultured monocytes/macrophages in patients with premature coronary artery disease (CAD). The study included 38 patients with premature CAD and 35 patients without CAD. A primary culture of CD14+ monocytes was obtained by immunomagnetic separation. The inflammatory response was induced by incubation of a cell culture with lipopolysaccharide (LPS) for 24 hours on Days 1 and 6. Basal and LPS-stimulated secretion of the cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8) and monocyte chemotactic protein-1 (MCP-1) was assessed by enzyme immunoassay on Days 2 and 7 of cultivation. The level of basal secretion of TNF-α, IL-1β, IL-6, MCP-1 was higher in patients with CAD compared to patients in the control group. The levels of re-stimulated TNF-α secretion and the levels of LPS-stimulated and re-stimulated IL-1β secretion on the second and sixth days were also higher in patients with CAD. LPS-stimulated MCP-1 secretion on the second day did not differ in patients of both groups, but re-stimulated MCP-1 secretion was higher in patients with CAD. The results of logistic regression analysis showed that the basal secretion levels of IL-1β and IL-6 were independently associated with premature CAD, along with smoking, body mass index and serum HDL-cholesterol levels.
Collapse
Affiliation(s)
- Tatiana Blokhina
- Department of problems of atherosclerosis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Tatiana Kirichenko
- Laboratory of medical genetics, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Yuliya Markina
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Ulyana Khovantseva
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Ivan Melnikov
- Laboratory of cell hemostasis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
- Laboratory of Gas Exchange, Biomechanics and Barophysiology, State Scientific Center of the Russian Federation, The Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow 123007, Russia
| | - Olga Guseva
- Laboratory of cell hemostasis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey Bazanovich
- Laboratory of stem cells, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey Kozlov
- Department of problems of atherosclerosis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alexander Orekhov
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| |
Collapse
|
11
|
Eskin FP, Patir P, Yilmaz U, Duman F, Buber H. The Relationship Between the Monocyte-to-High-Density Lipoprotein Cholesterol Ratio and Platelet Volume Indices With Diabetic Retinopathy. Cureus 2025; 17:e79822. [PMID: 40161128 PMCID: PMC11955199 DOI: 10.7759/cureus.79822] [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] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Aim This study aims to evaluate the roles of monocyte count-to-high-density lipoprotein (HDL) cholesterol ratio (MHR) and platelet volume indices (PVI) in predicting diabetic retinopathy (DR) in patients diagnosed with type 2 diabetes mellitus (DM) and to explore new methods for early prediction of retinopathy. Methods This prospective study included 120 patients aged over 18 years with type 2 DM diagnosed according to the American Diabetes Association criteria, along with a control group of 40 healthy individuals. Patients with type 2 DM were divided into three groups: 40 without retinopathy, 40 with non-proliferative diabetic retinopathy (NPDR), and 40 with proliferative diabetic retinopathy (PDR). Parameters such as complete blood count, PVI (mean platelet volume, platelet distribution width, plateletcrit, platelet large cell ratio), lipid profile, inflammatory markers (sedimentation rate, C-reactive protein), MHR, renal function tests, and glucose metabolism markers were analyzed alongside demographic, clinical, and laboratory data to assess their associations with DR in both patient and control groups. Results This study investigated the relationship between DR and hematological parameters (PVI, MHR). Hypertension, hyperlipidemia, and chronic kidney disease were common comorbidities. NPDR patients more frequently used metformin and dipeptidyl peptidase (DPP)-4 inhibitors, while PDR patients more often used insulin (p<0.05). Diabetic neuropathy and nephropathy were more prevalent in the PDR group. PDR patients had a significantly longer DM duration. Mean platelet volume (MPV) and platelet large cell ratio (PLCR) were higher in the PDR group. ROC analysis identified MHR (>11.9, 65% sensitivity, 35% specificity) and plateletcrit (PCT<0.29%, 61% sensitivity, 80% specificity) cut-offs for DR prediction. MHR's area under the curve (AUC) was greater than PCT's (p<0.05). MPV differed between PDR and NPDR, but MHR did not differ among DR groups or between DR/non-DR individuals. Glycosylated hemoglobin (HbA1c) and DM duration correlated positively with MHR, which also correlated with leukocyte count. HbA1c and glucose correlated with platelet distribution width (PDW). Each unit increase in DM duration increased MHR 1.13-fold, and each unit increase in HbA1c increased MHR 1.9-fold. Conclusions The duration of DM was significantly longer in patients with PDR compared to those with NPDR and those without retinopathy. MPV and PLCR levels were significantly higher in PDR patients than in NPDR patients. In evaluating the relationship between PVI and MHR for predicting diabetic retinopathy, significant cut-off values of MHR and PCT were found to be predictive indicators of diabetic retinopathy.
Collapse
Affiliation(s)
- Firdevs Pelin Eskin
- Department of Hematology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, TUR
| | - Pusem Patir
- Department of Hematology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, TUR
| | - Ustun Yilmaz
- Department of Nephrology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, TUR
| | - Fulya Duman
- Department of Ophthalmology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, TUR
| | - Hakan Buber
- Department of Ophthalmology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, TUR
| |
Collapse
|
12
|
Wang H, Li J, Wang Y, Chen Y, Zhang W, Pan X, Su C, Li Z, Wang L, Gu J. IgG4-mediated M2 macrophage polarization in tertiary lymphoid structures of esophageal cancer: implications for immunosuppression. Front Immunol 2025; 15:1497783. [PMID: 39896813 PMCID: PMC11782137 DOI: 10.3389/fimmu.2024.1497783] [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: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Background Our previous research highlighted the potential role of immunoglobulin G4 (IgG4) in mediating immunosuppression within the tumor microenvironment (TME). Tertiary lymphoid structures (TLS) in the TME have important immune-related functions. This study aims to analyze the distribution characteristics of IgG4-expressing cells, regulatory T cells (Tregs), and M2-type macrophages as well as to elucidate the relationship between IgG4 and the polarization of M2 macrophages within TLS in esophageal cancer. Object To elucidate the distribution of IgG4, Treg cells, and M2 macrophages in TLS and to assess the impact of IgG4 on macrophage polarization. Methods Esophageal cancer tissue were analyzed with multiplex immunofluorescence to determine the spatial distribution and density of B cells, T cells, and their subtypes. The relationship between IgG4 and CD8+ T cells in TLS, along with interleukin-10 (IL-10) expression and Treg presence, was studied. Serum IgG4 and IL-10 levels were compared between patients and healthy controls. In vitro, the impact of IgG4 on monocyte differentiation into M2 macrophages was observed. Results IgG4 density was inversely related with CD8+ T cells in mature TLS indicating a potential immunosuppressive role (P<0.05,*). Serum analysis revealed that both IgG4 (P<0.01, **) and IL-10 (P<0.0001, ****) were significantly elevated and positively correlated in tumor patients compared to controls (P<0.01, **). In vitro experiments confirmed that IgG4 monocyte differentiation into M2 macrophages, potentially enhancing the immunosuppressive phenotype in TLS. Conclusion IgG4 and IL-10 may contribute to immunosuppression in esophageal cancer by promoting the polarization of M2 macrophages within TLS, which could be a therapeutic target.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jirui Li
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yinghai Wang
- Department of Gynecology, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yang Chen
- Department of Pathology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Weifeng Zhang
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xinyan Pan
- Department of Pathology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Chanjuan Su
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, China
| | - Ziteng Li
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, China
| | - Li Wang
- Department of Pathology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiang Gu
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| |
Collapse
|
13
|
Echevarria-Lima J, Moles R. Monocyte and Macrophage Functions in Oncogenic Viral Infections. Viruses 2024; 16:1612. [PMID: 39459945 PMCID: PMC11512331 DOI: 10.3390/v16101612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Monocytes and macrophages are part of innate immunity and constitute the first line of defense against pathogens. Bone marrow-derived monocytes circulate in the bloodstream for one to three days and then typically migrate into tissues, where they differentiate into macrophages. Circulatory monocytes represent 5% of the nucleated cells in normal adult blood. Following differentiation, macrophages are distributed into various tissues and organs to take residence and maintain body homeostasis. Emerging evidence has highlighted the critical role of monocytes/macrophages in oncogenic viral infections, mainly their crucial functions in viral persistence and disease progression. These findings open opportunities to target innate immunity in the context of oncogenic viruses and to explore their potential as immunotherapies.
Collapse
Affiliation(s)
- Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| | - Ramona Moles
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
14
|
Alamoudi JA, El-Masry TA, El-Nagar MMF, El Zahaby EI, Elmorshedy KE, Gaballa MMS, Alshawwa SZ, Alsunbul M, Alharthi S, Ibrahim HA. Chitosan/hesperidin nanoparticles formulation: a promising approach against ethanol-induced gastric ulcers via Sirt1/FOXO1/PGC-1α/HO-1 pathway. Front Pharmacol 2024; 15:1433793. [PMID: 39314751 PMCID: PMC11417028 DOI: 10.3389/fphar.2024.1433793] [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: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
Hesperidin (Hes) protects different organs from damage by acting as a potent antioxidant and anti-inflammatory. This study aims to evaluate the gastroprotective effects of free hesperidin and its chitosan nanoparticles (HNPs) against ethanol-induced gastric ulcers in rats, hypothesizing that HNPs will enhance bioavailability and therapeutic efficacy due to improved solubility and targeted delivery. HNPs were synthesized via ion gelation and characterized using TEM, SEM, and zeta potential analyses. Key assessments included gastric acidity, histological analysis, and markers of inflammation, oxidative stress, and apoptosis. HNPs significantly decreased gastric acidity, reduced inflammatory and apoptotic markers, and enhanced antioxidant enzyme activities compared to free hesperidin and esomeprazole. Furthermore, Sirt-1, PGC-1α, HO-1, and FOXO1 gene expression were also evaluated. HNPs raised Sirt-1, PGC-1α, HO-1, and downregulated FOXO1, and they suppressed the activities of NF-κB p65, COX-2, IL-1β, CD86, FOXO1 P53, and caspase-3 and increased Sirt-1 activity. HNPs treatment notably restored antioxidant enzyme activity, reduced oxidative stress and inflammatory markers, and improved histological outcomes more effectively than free hesperidin and esomeprazole. These results indicate that chitosan nanoparticles significantly enhance the gastroprotective effects of hesperidin against ethanol-induced gastric ulcers, potentially offering a more effective therapeutic strategy. Further research should explore the clinical applications of HNPs in human subjects.
Collapse
Affiliation(s)
- Jawaher Abdullah Alamoudi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Maysa M. F. El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Enas I. El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Kadreya E. Elmorshedy
- Department of Anatomy, Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Anatomy, King Khaled College of Medicine, Riyadh, Saudi Arabia
| | - Mohamed M. S. Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Samar Zuhair Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Maha Alsunbul
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sitah Alharthi
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| | - Hanaa A. Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
15
|
Lupica-Tondo GL, Arner EN, Mogilenko DA, Voss K. Immunometabolism of ferroptosis in the tumor microenvironment. Front Oncol 2024; 14:1441338. [PMID: 39188677 PMCID: PMC11345167 DOI: 10.3389/fonc.2024.1441338] [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: 05/30/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Ferroptosis is an iron-dependent form of cell death that results from excess lipid peroxidation in cellular membranes. Within the last decade, physiological and pathological roles for ferroptosis have been uncovered in autoimmune diseases, inflammatory conditions, infection, and cancer biology. Excitingly, cancer cell metabolism may be targeted to induce death by ferroptosis in cancers that are resistant to other forms of cell death. Ferroptosis sensitivity is regulated by oxidative stress, lipid metabolism, and iron metabolism, which are all influenced by the tumor microenvironment (TME). Whereas some cancer cell types have been shown to adapt to these stressors, it is not clear how immune cells regulate their sensitivities to ferroptosis. In this review, we discuss the mechanisms of ferroptosis sensitivity in different immune cell subsets, how ferroptosis influences which immune cells infiltrate the TME, and how these interactions can determine epithelial-to-mesenchymal transition (EMT) and metastasis. While much focus has been placed on inducing ferroptosis in cancer cells, these are important considerations for how ferroptosis-modulating strategies impact anti-tumor immunity. From this perspective, we also discuss some promising immunotherapies in the field of ferroptosis and the challenges associated with targeting ferroptosis in specific immune cell populations.
Collapse
Affiliation(s)
- Gian Luca Lupica-Tondo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Emily N. Arner
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Denis A. Mogilenko
- Department of Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kelsey Voss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
16
|
Manole CG, Voiculescu VM, Soare C, Ceafalan LC, Gherghiceanu M, Hinescu ME. Skin Telocytes Could Fundament the Cellular Mechanisms of Wound Healing in Platelet-Rich Plasma Administration. Cells 2024; 13:1321. [PMID: 39195210 PMCID: PMC11353115 DOI: 10.3390/cells13161321] [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] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
For more than 40 years, autologous platelet concentrates have been used in clinical medicine. Since the first formula used, namely platelet-rich plasma (PRP), other platelet concentrates have been experimented with, including platelet-rich fibrin and concentrated growth factor. Platelet concentrates have three standard characteristics: they act as scaffolds, they serve as a source of growth factors and cytokines, and they contain live cells. PRP has become extensively used in regenerative medicine for the successful treatment of a variety of clinical (non-)dermatological conditions like alopecies, acne scars, skin burns, skin ulcers, muscle, cartilage, and bone repair, and as an adjuvant in post-surgery wound healing, with obvious benefits in terms of functionality and aesthetic recovery of affected tissues/organs. These indications were well documented, and a large amount of evidence has already been published supporting the efficacy of this method. The primordial principle behind minimally invasive PRP treatments is the usage of the patient's own platelets. The benefits of the autologous transplantation of thrombocytes are significant, representing a fast and economic method that requires only basic equipment and training, and it is biocompatible, thus being a low risk for the patient (infection and immunological reactions can be virtually disregarded). Usually, the structural benefits of applying PRP are attributed to fibroblasts only, as they are considered the most numerous cell population within the interstitium. However, this apparent simplistic explanation is still eluding those different types of interstitial cells (distinct from fibroblasts) that are residing within stromal tissue, e.g., telocytes (TCs). Moreover, dermal TCs have an already documented potential in angiogenesis (extra-cutaneous, but also within skin), and their implication in skin recovery in a few dermatological conditions was attested and described ultrastructurally and immunophenotypically. Interestingly, PRP biochemically consists of a series of growth factors, cytokines, and other molecules, to which TCs have also proven to have a positive expression. Thus, it is attractive to hypothesize and to document any tissular collaboration between cutaneous administered PRP and local dermal TCs in skin recovery/repair/regeneration. Therefore, TCs could be perceived as the missing link necessary to provide a solid explanation of the good results achieved by administering PRP in skin-repairing processes.
Collapse
Affiliation(s)
- Catalin G. Manole
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M. Voiculescu
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Gherghiceanu
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihail E. Hinescu
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|