1
|
Tian W, Ju J, Guan B, Wang T, Zhang J, Song L, Xu H. Role of hyperhomocysteinemia in atherosclerosis: from bench to bedside. Ann Med 2025; 57:2457527. [PMID: 39898976 PMCID: PMC11792134 DOI: 10.1080/07853890.2025.2457527] [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: 07/20/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Atherosclerosis is a leading cause of global mortality, driven by complex interactions between genetic, metabolic, and environmental factors. Among these, hyperhomocysteinemia (HHcy) has emerged as a significant and modifiable risk factor, contributing to endothelial dysfunction, oxidative stress, and vascular inflammation. Despite increasing recognition of its role in atherogenesis, the precise mechanisms and clinical implications of HHcy remain incompletely understood, necessitating a comprehensive review to connect recent mechanistic insights with practical applications. METHODS We analyzed the various mechanisms whereby HHcy accelerates the progression of atherosclerosis, and conducted a comprehensive review of publications in the fields of HHcy and atherosclerosis. RESULTS HHcy promotes atherosclerosis through several mechanisms, including inflammation, oxidative stress, epigenetic modification, and lipoprotein metabolism alteration. Moreover, this discussion extends to current strategies for the prevention and clinical management of HHcy-induced atherosclerosis. CONCLUSION This review consolidates and elucidates the latest advancements and insights into the role of HHcy in atherosclerosis. The comprehensive narrative connects fundamental research with clinical applications. Contemporary studies highlight the complex interplay between HHcy and atherosclerosis, establishing HHcy as not only a contributing risk factor but also an accelerator of various atherogenic processes.
Collapse
Affiliation(s)
- Wende Tian
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing China
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tongxin Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing China
| | - Jiqian Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Luxia Song
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing China
| |
Collapse
|
2
|
Huang J, Chen L, Li W, Chang CJ. Anti-inflammatory and antioxidative effects of Perilla frutescens-derived extracellular vesicles: Insights from Zebrafish models. Mol Immunol 2025; 182:126-138. [PMID: 40267772 DOI: 10.1016/j.molimm.2025.04.008] [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: 02/05/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Plant-derived extracellular vesicles have recently been extracted and recognized as promising bioactive molecules, owing to their distinctive biological properties and inherent therapeutic activities. In this study, we investigated the physicochemical characteristics, bioactive properties, and therapeutic potential of Perilla frutescens-derived exosome-like nanoparticles (PELNs). Transmission electron microscopy (TEM) revealed that PELNs exhibited a cup-shaped morphology, with a lipid bilayer and a size distribution ranging from 40 to 200 nm (mean: 68.4 ± 13.0 nm). The cargoes in PELNs were analyzed through multi-omics and small RNA sequencing. In vivo studies on zebrafish demonstrated that PELNs are non-toxic at experimental concentrations. A reduction in neutrophil migration to injured fins evidenced the anti-inflammatory properties of PELNs. Furthermore, a meta-analysis of transcriptomic data identified hundreds of differentially expressed genes (DEGs) across 12 samples of three experimental groups. These DEGs were annotated into three categories following gene ontology (GO) enrichment analysis. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that these DEGs were involved in immune-related pathways, including complement and coagulation cascades, systemic lupus erythematosus, PPAR signaling pathways, and antigen processing and presentation. Twelve selected DEGs were validated by quantitative real-time PCR (qRT-PCR), with particular confirmation of the mpx and lcp1 genes via in situ hybridization. Furthermore, PELNs demonstrated antioxidative effects by mitigating reactive oxygen species (ROS) levels, as evidenced by measurements of four oxidative stress (OS) indicators (i.e., SOD, CAT, GSH, and MDA) in zebrafish larvae subjected to H2O2-induced OS. In summary, PELNs exhibit substantial anti-inflammatory and antioxidant properties, underscoring their potential as therapeutic agents for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Jinghong Huang
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen, Fujian 362021, China; School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Linxin Chen
- Department of Traditional Chinese Medicine, Xiamen Chang Gung Hospital, Xiamen, Fujian 301028, China
| | - Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, Xiamen, Fujian 362021, China; School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China.
| | - Chih-Jung Chang
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China; Medical Research Center, Xiamen Chang Gung Hospital, Xiamen, Fujian 301028, China; Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, Fujian 301028, China.
| |
Collapse
|
3
|
Goldschmidt-Clermont PJ, Sevilla BA. Redox and actin, a fascinating story. Redox Biol 2025; 83:103630. [PMID: 40328105 DOI: 10.1016/j.redox.2025.103630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Actin is an extraordinarily complex protein whose functions are essential to cell motility, division, contraction, signaling, transport, tissular structures, DNA repair, and many more cellular activities critical to life for both animals and plants. It is one of the most abundant and conserved proteins and it exists in either a soluble, globular (monomeric, G-actin) or an insoluble, self-assembled (polymerized or filamentous actin, F-actin) conformation as a key component of the cytoskeleton. In the early 1990's little, if anything, was known about the impact of reactive oxygen species (ROS) on the biology of actin except that ROS could disrupt the actin cytoskeleton. Instructively, G-actin is susceptible to alteration by ROS, and thus, purification of G-actin is typically performed in the presence of strong antioxidants (like dithiothreitol) to limit its oxidative degradation. In contrast, F-actin is a more stable conformation and thus actin can be kept relatively intact in purified preparations as filaments at low temperature for extended periods of time. Both G- and F-actin interact with a myriad of intracellular proteins and at least with a couple of extracellular proteins, and these interactions are essential to the many actin functions. This review will show how, over the past 30 years, our understanding of the role of ROS for actin biology has evolved from noxious denaturizing agents to remarkable regulators of the actin cytoskeleton in cells and consequent cellular functions.
Collapse
|
4
|
Huerta MÁ, Molina-Álvarez M, García MM, Tejada MA, Goicoechea C, Ghasemlou N, Ruiz-Cantero MC, Cobos EJ. The role of neutrophils in pain: systematic review and meta-analysis of animal studies. Pain 2025; 166:1230-1249. [PMID: 39450928 DOI: 10.1097/j.pain.0000000000003450] [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: 03/26/2024] [Accepted: 09/10/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The peripheral inflammatory response is an attractive therapeutic target for pain treatment. Neutrophils are the first circulating inflammatory cells recruited to sites of injury, but their contribution to pain outcomes is unclear. We performed a systematic review and meta-analysis of original preclinical studies, which evaluated the effect of preemptive neutrophil depletion on pain outcomes (PROSPERO registration number: CRD42022364004). Literature search (PubMed, January 19, 2023) identified 49 articles, which were meta-analyzed using a random-effects model. The risk of bias was evaluated using SYRCLE's tool. The pooled effect considering all studies showed that neutrophil depletion induced a consistent pain reduction. Inflammatory, joint, neuropathic, and visceral pain showed significant pain alleviation by neutrophil depletion with medium-large effect sizes. However, muscle and postoperative pain were not significantly alleviated by neutrophil depletion. Further analysis showed a differential contribution of neutrophils to pain outcomes. Neutrophils had a higher impact on mechanical hyperalgesia, followed by nociceptive behaviors and mechanical allodynia, with a smaller contribution to thermal hyperalgesia. Interspecies (mice or rats) differences were not appreciated. Analyses regarding intervention unveiled a lower pain reduction for some commonly used methods for neutrophil depletion, such as injection of antineutrophil serum or an anti-Gr-1 antibody, than for other agents such as administration of an anti-Ly6G antibody, fucoidan, vinblastine, CXCR1/2 inhibitors, and etanercept. In conclusion, the contribution of neutrophils to pain depends on pain etiology (experimental model), pain outcome, and the neutrophil depletion strategy. Further research is needed to improve our understanding on the mechanisms of these differences.
Collapse
Affiliation(s)
- Miguel Á Huerta
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Miguel M García
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Miguel A Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Carlos Goicoechea
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Nader Ghasemlou
- Pain Chronobiology & Neuroimmunology Laboratory, Departments of Anesthesiology and Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - M Carmen Ruiz-Cantero
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
- Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| |
Collapse
|
5
|
Shu F, Wang Y, Li L, Shi L, Zhang F, Ma Z, Mao D. Multi-omics integration and machine learning identify and validate neutrophil extracellular trap-associated gene signatures in chronic rhinosinusitis with nasal polyps. Clin Immunol 2025; 275:110473. [PMID: 40089249 DOI: 10.1016/j.clim.2025.110473] [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: 01/21/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
This study aimed to explore the molecular characteristics of neutrophil extracellular traps (NETs) in chronic rhinosinusitis with nasal polyps (CRSwNP). Differentially expressed gene analysis, weighted gene co-expression network analysis, and machine learning algorithms identified three core NETs-associated genes: CXCR4, CYBB, and PTAFR, which were significantly upregulated in CRSwNP patients. The diagnostic performance of these genes was evaluated using receiver operating characteristic (ROC) curves, and their clinical relevance was validated using multicenter data. Immune infiltration analysis showed strong correlations between these genes and neutrophil and immune cell infiltration. Single-cell RNA sequencing demonstrated that these genes were predominantly expressed in myeloid and immune cells and exhibited dynamic changes during disease progression. These genes may contribute to CRSwNP pathogenesis through IL-17 signaling and metabolism-related pathways. This study identifies novel biomarkers and therapeutic targets for precise diagnosis and personalized treatment of CRSwNP.
Collapse
Affiliation(s)
- Fu Shu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Yaping Wang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Linglong Li
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Lei Shi
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Feng Zhang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Zhixuan Ma
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Dehong Mao
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China.
| |
Collapse
|
6
|
Chen Q, Yu Y, Tong L, Weiss RM, Wei SG. Targeted delivery of TAPI-1 via biomimetic nanoparticles ameliorates post-infarct left ventricle function and remodelling. Cardiovasc Res 2025; 121:760-774. [PMID: 40038918 DOI: 10.1093/cvr/cvaf039] [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: 07/11/2024] [Revised: 10/21/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
AIMS The potential of nanoparticles as effective drug delivery tools for treating failing hearts in heart failure remains a challenge. Leveraging the rapid infiltration of neutrophils into infarcted hearts after myocardial infarction (MI), we developed a nanoparticle platform engineered with neutrophil membrane proteins for the targeted delivery of TAPI-1, a TACE/ADAM17 inhibitor, to the inflamed myocardium, aiming to treat cardiac dysfunction and remodelling in rats with MI. METHODS AND RESULTS Neutrophil-mimic liposomal nanoparticles (Neu-LNPs) were constructed by integrating synthesized liposomal nanoparticles with LPS-stimulated neutrophil membrane fragments and then loaded with TAPI-1. MI rats were treated with TAPI-1 delivered via Neu-LNPs for 4 weeks. Left ventricular function was assessed by echocardiography and cardiac fibrosis was evaluated post-treatment. The novel Neu-LNPs maintained typical nanoparticle features, but with increased biocompatibility. Neu-LNPs demonstrated improved targeting ability and cellular internalization, facilitated by LFA1/Mac1/ICAM-1 interaction. Neu-LNPs displayed higher accumulation and cellular uptake by macrophages and cardiomyocytes in infarcted hearts post-MI, with a sustained duration. Treatments with TAPI-1-Neu-LNPs demonstrated greater protection against myocardial injury and cardiac dysfunction in MI rats compared to untargeted TAPI-1, along with reduced cardiac collagen deposition and expression of fibrosis biomarkers as well as altered immune cell compositions within the hearts. CONCLUSIONS Targeted treatment with TACE/ADAM17 inhibitor delivered via biomimetic nanoparticles exhibited pronounced advantages in improving left ventricle function, mitigating cardiac remodelling, and reducing inflammatory responses within the infarcted hearts. This study underscores the effectiveness of Neu-LNPs as a drug delivery strategy to enhance therapeutic efficacy in clinical settings.
Collapse
Affiliation(s)
- Qing Chen
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Yang Yu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Lei Tong
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
| | - Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
- Iowa Neuroscience Institute, University of Iowa, 169 Newton Road, Iowa City, IA 52242, USA
| |
Collapse
|
7
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025:1-17. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [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: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Chen DL, Lin YK, Wang GJ, Chang KC. Neutrophil to high-density lipoprotein cholesterol ratio as a potential inflammatory marker for predicting all-cause mortality in out-of-hospital cardiac arrest survivors. Sci Rep 2025; 15:17181. [PMID: 40382458 PMCID: PMC12085707 DOI: 10.1038/s41598-025-01951-x] [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: 12/26/2024] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
Out-of-hospital cardiac arrest (OHCA) survivors have more than one-third mortality rate. Numerous inflammatory indicators are available, and it should be feasible to identify a fast and accurate way to aid medical decisions. This retrospective cohort study included 247 patients with OHCA, hospitalized between January 2015 and August 2024. The study was conducted in the intensive care unit of China Medical University Hospital, Taichung, Taiwan. A variety of inflammatory markers, including interleukin-6, neutrophil to high-density lipoprotein cholesterol ratio (NHR), and C-reactive protein, were screened at 24 h after OHCA. The primary endpoint was the 90-day all-cause mortality. Receiver operating characteristic (ROC) curves and Kaplan-Meier survival curves of NHR were analyzed. Possible risk factors for all-cause mortality were estimated by Cox regression modeling. NHR and interleukin-6 were similarly predictive of all-cause mortality in inflammatory response, and both were superior to C-reactive protein at 24 h after OHCA (p < 0.001). The area under the ROC curve of NHR was 0.74 (95% confidence interval [CI]: 0.66-0.81, p < 0.001), sensitivity: 0.68, and specificity: 0.68, and NHR = 16.1. The 90-day all-cause mortality rate for NHR > 16.1 compared to those with NHR ≤ 16.1 was 0.51 and 0.21, respectively, according to Kaplan-Meier curves analysis. The hazard ratio for NHR > 16.1 was 2.54 (95% CI: 1.68-3.82, p < 0.001). An NHR > 16.1 at 24 h after OHCA is a potential inflammatory marker for predicting all-cause mortality.
Collapse
Affiliation(s)
- Da-Long Chen
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, No. 2, Yuh-Der Road, North District, Taichung, 40447, Taiwan.
| | - Yu-Kai Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, No. 2, Yuh-Der Road, North District, Taichung, 40447, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Guei-Jane Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, No. 2, Yuh-Der Road, North District, Taichung, 40447, Taiwan.
- Pharmacy Department, Wizcare Medical Corporation Aggregate, Taichung, Taiwan.
- School of Medicine, Weifang University of Science and Technology, Weifang, Shandong, China.
| | - Kuan-Cheng Chang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, No. 2, Yuh-Der Road, North District, Taichung, 40447, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
9
|
Ren K, Peng X, Duan X, Feng R, Cook C, Lu M, Li M, Gu H, Wang X, Deng G, Ma H, Liu Y, Xia Y. Synergistic effects of LCN2 and TWEAK on the progression of psoriasis. Cell Mol Immunol 2025:10.1038/s41423-025-01292-9. [PMID: 40369189 DOI: 10.1038/s41423-025-01292-9] [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: 06/22/2024] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
Lipocalin 2 (LCN2) and the TWEAK/Fn14 signaling pathways are pivotal in psoriasis, influencing epidermal development, inflammatory cell chemotaxis, and inflammatory factor release. Despite their significant roles, the intricate relationship between LCN2 and TWEAK/Fn14 pathways remains unclear. Our study revealed the correlation between the expression of TWEAK, LCN2, and Fn14 in psoriatic lesions. We found that TWEAK is expressed by keratinocytes and macrophages, while LCN2 is expressed by keratinocytes and neutrophils. Surface plasmon resonance experiments demonstrated binding between LCN2 and Fn14, which was further validated by co-immunoprecipitation and cellular co-localization via immunofluorescence. In vitro, LCN2 promoted macrophage differentiation and TWEAK secretion, enhanced TWEAK and Fn14 expression in keratinocytes, and activated the MAPK signaling pathway. TWEAK upregulated LCN2 expression in neutrophils but not in keratinocytes. Bulk RNA-seq analysis revealed a synergistic effect of LCN2 and TWEAK in promoting inflammatory cytokine expression in keratinocytes, with enhanced MAPK pathway activation in the presence of M5 cytokines. Lcn2 knockout reduced Fn14 expression in skin lesions and serum TWEAK levels of imiquimod-induced murine psoriasis model, while Fn14 knockout attenuated the epidermal hyperplasia-promoting effects of TWEAK and LCN2. Overexpression of Fn14 in keratinocytes led to higher TWEAK expression upon LCN2 stimulation, suggesting a self-reinforcing loop among TWEAK, LCN2, and Fn14. We propose that LCN2 synergizes with TWEAK through Fn14 to drive psoriasis pathogenesis.
Collapse
Affiliation(s)
- Kaixuan Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xueting Peng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xudong Duan
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongfang Feng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Christopher Cook
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Min Li
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyu Wang
- Department of Dermatology, Jinling Hospital, Nanjing, China
| | - Guorong Deng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huiqun Ma
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yale Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
10
|
Kwak JW, Houghton AM. Targeting neutrophils for cancer therapy. Nat Rev Drug Discov 2025:10.1038/s41573-025-01210-8. [PMID: 40374764 DOI: 10.1038/s41573-025-01210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/18/2025]
Abstract
Neutrophils are among the most abundant immune cell types in the tumour microenvironment and have been associated with poor outcomes across multiple cancer types. Yet despite mounting evidence of their role in tumour progression, therapeutic strategies targeting neutrophils have only recently gained attention and remain limited in scope. This is probably due to the increasing number of distinct neutrophil subtypes identified in cancer and the limited understanding of the mechanisms by which these subsets influence tumour progression and immune evasion. In this Review, we discuss the spectrum of neutrophil subtypes - including those with antitumour activity - and their potential to polarize towards tumour-suppressive phenotypes. We explore the molecular pathways and effector functions by which neutrophils modulate cancer progression, with an emphasis on identifying tractable therapeutic targets. Finally, we examine emerging clinical trials aimed at modulating neutrophil lineages and consider their implications for patient outcomes.
Collapse
Affiliation(s)
- Jeff W Kwak
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
11
|
Su Z, Cao L, Chen H, Zhang P, Wu C, Lu J, He Z. Obesity indicators mediate the association between the aggregate index of systemic inflammation (AISI) and type 2 diabetes mellitus (T2DM). Lipids Health Dis 2025; 24:176. [PMID: 40369536 PMCID: PMC12080010 DOI: 10.1186/s12944-025-02589-4] [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: 02/17/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE This study analyzes data from the 2009-2018 National Health and Nutrition Examination Survey (NHANES) to explore the relationship between the Aggregate Index of Systemic Inflammation (AISI), also referred to as the pan-immune-inflammation value (PIV), and Type 2 Diabetes Mellitus (T2DM) among adults in the United States. Furthermore, it evaluates the mediating effect of obesity indicators on this association. METHODS This study included 9,947 individuals from NHANES and applied appropriate weighting techniques. To examine the relationship between AISI and T2DM, we used various statistical models, including weighted multivariable logistic regression, smooth curve fitting, threshold effect analysis, subgroup analysis, trend tests, mediation analysis, and Shapley additive explanations (SHAP) models. RESULTS This research included a total of 9,947 participants, with 3,220 diagnosed with T2DM, while 6,727 remained undiagnosed. Weighted multiple logistic regression with all covariates adjusted indicated that with every one-unit increment in AISI/1000, there was an 88.3% likelihood of T2DM occurrence (OR: 1.883, 95% CI: 1.378-2.571). The stratified analysis identified significant differences in this association based on age, biological sex, level of education, poverty-income ratio (PIR), tobacco consumption status, and body mass index (BMI). Interaction tests revealed a positive association between AISI and T2DM, apart from PIR, BMI, age, education attainment, race, gender, tobacco use status, Estimated Glomerular Filtration Rate(eGFR), platelet count, and high blood pressure, with none of the interaction p-values falling below 0.05. Nevertheless, the occurrence of cardiovascular disease (CVD) among participants may affect the strength of this relationship, where an interaction p-value was less than 0.05. Additionally, smoothing curve fitting revealed a nonlinear relationship between AISI and T2DM, marking a significant change at AISI/1000 of 0.21. Mediation analysis indicated that five obesity-related indicators-LAP, VAI, WHtR, WWI and ABSI - partly mediated the association between AISI/1000 and T2DM. CONCLUSION An increase in AISI is associated with an elevated probability of T2DM, with obesity indicators potentially mediating this relationship. Reducing AISI and managing obesity may help prevent T2DM. However, with the cross-sectional design of this study, causal relationships cannot be established. Future research should utilize longitudinal studies to confirm these findings.
Collapse
Affiliation(s)
- Ziying Su
- Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Lei Cao
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Hailong Chen
- Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Peng Zhang
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Chunwei Wu
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jing Lu
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ze He
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China.
| |
Collapse
|
12
|
Yan W, Zhao Y, Zhang J, Jiang P, Ma H, Fang M, Xi X. Causal relationship between molecular markers of biological aging and orthopedic diseases: A two-sample bidirectional Mendelian randomization study. Exp Gerontol 2025; 206:112785. [PMID: 40373834 DOI: 10.1016/j.exger.2025.112785] [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: 02/17/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Studies indicate an association between biological aging and orthopedic diseases, but the causality remains unclear. AIMS This study aims to investigate the bidirectional causal relationship between molecular markers of biological aging age and orthopedic conditions. METHODS A two-sample Mendelian randomization (MR) analysis based on a genome-wide association study (GWAS) was conducted to explore these causal relationships. Analysis methods included inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode. Sensitivity analyses involved Cochran's Q, MR-Egger, leave-one-out, and MR pleiotropy residual sum and outlier (MR-PRESSO) tests. RESULTS The forward MR analysis identified several causal relationships: granulocyte proportions influenced intervertebral disc degeneration (IVDD) (OR 0.2316, P = 0.0101) and low back pain (LBP) (OR 0.2624, P = 0.007); telomere length (TL) affected cervical spondylosis (C/S) (OR 0.8759, P = 0.0167) and IVDD (OR 0.9184, P = 0.023); fibroblast growth factor-23 (FGF-23) impacted frozen shoulder (FS) (OR 1.2424, P = 0.0316); and HannumAge influenced C/S (OR 0.9518, P = 0.0233). The reverse MR analysis found that FS influenced TL (OR 0.9582, P = 0.0002) and α-Klotho (OR 0.7592, P = 0.0256), while sciatica affected TL (OR 0.9344, P = 0.0055) and C/S impacted PhenoAge (OR 1.6583, P = 0.0131) after outlier exclusion. Cochran's Q indicated heterogeneity in certain analyses, and MR-Egger showed no horizontal pleiotropy in significant causal associations. CONCLUSIONS This study suggests a potential causal associations between molecular markers of biological aging and orthopedic diseases, suggesting avenues for future research into the underlying mechanisms.
Collapse
Affiliation(s)
- Wei Yan
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichen Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahui Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Clinical Laboratory, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, JiangSu, China
| | - Ping Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honghong Ma
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Fang
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaobing Xi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
von Ehr A, Steenbuck ID, Häfele C, Remmersmann F, Vico TA, Ehlert C, Lindner D, Wolf D, Tholen S, Schilling O, Czerny M, Westermann D, Hilgendorf I. Experimental evidence on colchicine's mode of action in human carotid artery plaques. Atherosclerosis 2025; 406:119239. [PMID: 40381496 DOI: 10.1016/j.atherosclerosis.2025.119239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND AIMS Atherosclerosis, driven by inflammation, is a leading cause of cardiovascular events. Recent clinical trials have highlighted the therapeutic potential of anti-inflammatory treatments. Consequently, colchicine is being recommended for secondary prevention in current guidelines, although the drug's mechanistic actions are not fully understood. METHODS To this end, we conducted a multiomic investigation of colchicine's effect on human carotid plaques. Sections from endarterectomy specimens were exposed to colchicine at concentrations of 2 ng/ml and 10 ng/ml ex vivo for 24 h and compared to untreated segments of the same plaque. Gene expression changes were analyzed by bulk RNA sequencing, and plaque secretomes underwent mass spectrometry for proteomic analysis. In situ cell proliferation was assessed by histology. RESULTS Our data indicate, that colchicine suppresses neutrophil and platelet degranulation and activation, collagen degradation and atheromatous plaque macrophage proliferation in a dose-dependent manner in human plaques, while stimulating myofibroblast activation. Unexpectedly, interleukine (IL)-1beta release from colchicine treated plaques was not reduced. These results indicate that the inflammasome may not be the predominant target of low-dose colchicine in human carotid artery plaques. CONCLUSION Our study identifies multifactorial pathways through which colchicine, the first cardiovascular guideline-recommended anti-inflammatory drug, predominantly acts on human atherosclerotic lesions beyond the inflammasome. Targeting neutrophil and platelet degranulation, collagen degradation and macrophage proliferation, selectively, may provide substantial therapeutic benefit in atherosclerotic cardiovascular disease without colchicine's undesired side effects.
Collapse
Affiliation(s)
- Alexander von Ehr
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Ines Derya Steenbuck
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Häfele
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Remmersmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamara A Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolin Ehlert
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Yang X, Guo C, Yang Y, Huang L, Luo L, Zhou Y, Xiao Y, Deng L, Li S. Targeting neutrophil extracellular traps: SERPINE1 and THBS1 as non-invasive biomarkers for early detection of liver fibrosis in metabolic dysfunction-associated Steatotic liver disease. Int Immunopharmacol 2025; 158:114828. [PMID: 40349409 DOI: 10.1016/j.intimp.2025.114828] [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: 02/17/2025] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Metabolic dysfunction - Associated Steatotic Liver Disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), poses a significant clinical burden due to its high prevalence and potential progression to fibrosis. While neutrophil extracellular traps (NETs) have been implicated in MASLD progression, their specific role in fibrosis remains unclear. This study integrates transcriptomic and single-cell data using weighted gene co-expression network analysis (WGCNA), causal WGCNA (CWGCNA), single-sample gene set enrichment analysis (ssGSEA), gene set variation analysis (GSVA), and linear models for microarray data (Limma) to identify key genes driving steatosis-to-fibrosis transition. Validation in human serum, mouse liver tissue, and mouse serum confirmed that SERPINE1 and THBS1 as robust non-invasive biomarkers with strong diagnostic performance. When combined with clinical features, these markers improved fibrosis prediction accuracy in MASLD patients. Additionally, SERPINE1 appears to mediate interactions between hepatic stellate cells and neutrophils, highlighting a novel therapeutic target. Overall, our findings reveal that NETs-related genes, particularly SERPINE1 and THBS1, hold strong diagnostic value for early-stage fibrosis in MASLD. Targeting SERPINE1 in hepatic stellate cells offers a promising strategy for therapeutic intervention. This study provides a novel framework for non-invasive MASLD fibrosis prediction and lays the foundation for targeted interventions to mitigate disease progression.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunhong Guo
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yichun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Huang
- Department of Pediatric Research Institute, Chongqing, China; National Clinical Research Center for Child Health and Disorders, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Luo
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, China
| | - Youping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuliang Xiao
- Department of Gastroenterology, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Liang Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Shan Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
15
|
Xiang R, Ben-Eghan C, Liu Y, Roberts D, Ritchie S, Lambert SA, Xu Y, Takeuchi F, Inouye M. Genome-wide analyses of variance in blood cell phenotypes provide new insights into complex trait biology and prediction. Nat Commun 2025; 16:4260. [PMID: 40335489 PMCID: PMC12059119 DOI: 10.1038/s41467-025-59525-4] [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: 05/13/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Blood cell phenotypes are routinely tested in healthcare to inform clinical decisions. Genetic variants influencing mean blood cell phenotypes have been used to understand disease aetiology and improve prediction; however, additional information may be captured by genetic effects on observed variance. Here, we mapped variance quantitative trait loci (vQTL), i.e. genetic loci associated with trait variance, for 29 blood cell phenotypes from the UK Biobank (N ~ 408,111). We discovered 176 independent blood cell vQTLs, of which 147 were not found by additive QTL mapping. vQTLs displayed on average 1.8-fold stronger negative selection than additive QTL, highlighting that selection acts to reduce extreme blood cell phenotypes. Variance polygenic scores (vPGSs) were constructed to stratify individuals in the INTERVAL cohort (N ~ 40,466), where the genetically most variable individuals had increased conventional PGS accuracy (by ~19%) relative to the genetically least variable individuals. Genetic prediction of blood cell traits improved by ~10% on average combining PGS with vPGS. Using Mendelian randomisation and vPGS association analyses, we found that alcohol consumption significantly increased blood cell trait variances highlighting the utility of blood cell vQTLs and vPGSs to provide novel insight into phenotype aetiology as well as improve prediction.
Collapse
Affiliation(s)
- Ruidong Xiang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia.
- The School of Applied Systems Biology, La Trobe University, Melbourne, VIC, 3086, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Chief Ben-Eghan
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Yang Liu
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - David Roberts
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant-Oxford Centre, John Radcliffe Hospital and Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Scott Ritchie
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Yu Xu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Fumihiko Takeuchi
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Bioinformatics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
| |
Collapse
|
16
|
Chen L, Kadoya K, Endo T, Iwasaki N, Terkawi MA. Efferocytosis at the frontline of homeostasis: Shaping the bone microenvironment and therapeutic implications in related diseases. Cytokine Growth Factor Rev 2025:S1359-6101(25)00048-6. [PMID: 40368727 DOI: 10.1016/j.cytogfr.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Bone is a dynamic tissue that constantly undergoes remodeling processes throughout life to maintain its structure and integrity. During this process, physiological bone turnover, which is shaped by apoptosis, occurs in cells in the bone microenvironment. The clearance of these apoptotic cells (ACs) is executed by phagocytes through a process called efferocytosis, which simply means taking to the grave "burial." Efferocytosis is a multistage process involving the recognition, binding, internalization, and digestion of ACs, culminating in the resolution of inflammation. Critically, aberrations in efferocytosis lead to the accumulation of apoptotic corpses, impairing tissue homeostasis and contributing to various pathologies as well as bone-related diseases. Emerging evidence suggests that modulating/activating efferocytosis at any stage represents a promising therapeutic strategy for managing bone-related diseases, especially those associated with aging and inflammation. This review discusses the current understanding of the cellular and molecular mechanisms of efferocytosis, its roles within the bone microenvironment, and potential therapeutic interventions targeting efferocytosis in age-related bone diseases.
Collapse
Affiliation(s)
- Liyile Chen
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Ken Kadoya
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - M Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan.
| |
Collapse
|
17
|
Kerner G, Kamitaki N, Strober B, Price AL. Mapping disease loci to biological processes via joint pleiotropic and epigenomic partitioning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.05.25327017. [PMID: 40385425 PMCID: PMC12083580 DOI: 10.1101/2025.05.05.25327017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Genome-wide association studies (GWAS) have identified thousands of disease-associated loci, yet their interpretation remains limited by the heterogeneity of underlying biological processes. We propose Joint Pleiotropic and Epigenomic Partitioning (J-PEP), a clustering framework that integrates pleiotropic SNP effects on auxiliary traits and tissue-specific epigenomic data to partition disease-associated loci into biologically distinct clusters. To benchmark J-PEP against existing methods, we introduce a metric-Pleiotropic and Epigenomic Prediction Accuracy (PEPA)-that evaluates how well the clusters predict SNP-to-trait and SNP-to-tissue associations using off-chromosome data, avoiding overfitting. Applying J-PEP to GWAS summary statistics for 165 diseases/traits (average N = 290 K ), we attained 16-30% higher PEPA than pleiotropic or epigenomic partitioning approaches with larger improvements for well-powered traits, consistent with simulations; these gains arise from J-PEP's tendency to upweight correlated structure-signals present in both auxiliary trait and tissue data-thereby emphasizing shared components. For type 2 diabetes (T2D), J-PEP identified clusters refining canonical pathological processes while revealing underexplored immune and developmental signals. For hypertension (HTN), J-PEP identified stromal and adrenal-endocrine processes that were not identified in prior analyses. For neutrophil count, J-PEP identified hematopoietic, hepatic-inflammatory, and neuroimmune processes, expanding biological interpretation beyond classical immune regulation. Notably, integrating single-cell chromatin accessibility data refined bulk-based clusters, enhancing cell-type resolution and specificity. For T2D, single-cell data refined a bulk endocrine cluster to pancreatic islet β -cells, consistent with established β -cell dysfunction in insulin deficiency; for HTN, single-cell data refined a bulk endocrine cluster to adrenal cortex cells, consistent with a GO enrichment for neutrophil-mediated inflammation that implicates feedback between aldosterone production in the adrenal gland and local immune signaling. In conclusion, J-PEP provides a principled framework for partitioning GWAS loci into interpretable, tissue-informed clusters that provide biological insights on complex disease.
Collapse
Affiliation(s)
- Gaspard Kerner
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Nolan Kamitaki
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Benjamin Strober
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Broad Institute of MIT and Harvard, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
18
|
Li X, Zhou W, Zhou L, Li Y, Wu X, Chen J. Neutrophil-derived exosomal S100A8 aggravates lung injury in sepsis by inducing pyroptosis. Mol Immunol 2025; 181:29-39. [PMID: 40056630 DOI: 10.1016/j.molimm.2025.03.003] [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: 11/26/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Acute lung injury (ALI) is a common and life-threatening complication in patients with sepsis, with pro-inflammatory cell pyroptosis playing a crucial role in the associated organ damage. In this study, we aimed to identify potential therapeutic targets. Utilizing the GEO database (GSE232753), we analyzed the differentially expressed genes in the peripheral blood of healthy individuals and sepsis patients, identifying the significantly upregulated gene S100A8. Subsequently, we constructed a septic ALI model using lipopolysaccharide (LPS). Notably, S100A8 was highly expressed not only in serum and bronchoalveolar lavage fluid (BALF) but also in neutrophil exosomes. We then co-incubated BEAS-2B cells with neutrophil exosomes that were either treated or untreated with LPS. Cell proliferation activity was assessed using the CCK-8 assay, cell death was evaluated through propidium iodide (PI) staining, and the changes in pyroptosis indicators were detected via Western blot and ELISA. To further validate that LPS-induced neutrophil exosomes promote BEAS-2B cell pyroptosis through the delivery of S100A8, we conducted additional experiments involving the addition of S100A8 protein alone or S100A8 antibody in conjunction with neutrophil exosome treatment, followed by relevant assessments. Moreover, in vivo validation was also performed. Mechanistically, we revealed that S100A8 induces pyroptosis in BEAS-2B cells through the TLR4 signaling pathway. In conclusion, our findings provide new promising targets for the treatment of septic ALI.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Wei Zhou
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Liangliang Zhou
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Yingbin Li
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Xufeng Wu
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Jianjun Chen
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China.
| |
Collapse
|
19
|
Zhang Y, Zheng Z, Zhu S, Xu L, Zhang Q, Gao J, Ye M, Shen S, Xing J, Wu M, Xu RX. Electroactive Electrospun Nanofibrous Scaffolds: Innovative Approaches for Improved Skin Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416267. [PMID: 40190057 PMCID: PMC12079356 DOI: 10.1002/advs.202416267] [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] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/05/2025] [Indexed: 05/16/2025]
Abstract
The incidence and burden of skin wounds, especially chronic and complex wounds, have a profound impact on healthcare. Effective wound healing strategies require a multidisciplinary approach, and advances in materials science and bioengineering have paved the way for the development of novel wound healing dressing. In this context, electrospun nanofibers can mimic the architecture of the natural extracellular matrix and provide new opportunities for wound healing. Inspired by the bioelectric phenomena in the human body, electrospun nanofibrous scaffolds with electroactive characteristics are gaining widespread attention and gradually emerging. To this end, this review first summarizes the basic process of wound healing, the causes of chronic wounds, and the current status of clinical treatment, highlighting the urgency and importance of wound dressings. Then, the biological effects of electric fields, the preparation materials, and manufacturing techniques of electroactive electrospun nanofibrous (EEN) scaffolds are discussed. The latest progress of EEN scaffolds in enhancing skin wound healing is systematically reviewed, mainly including treatment and monitoring. Finally, the importance of EEN scaffold strategies to enhance wound healing is emphasized, and the challenges and prospects of EEN scaffolds are summarized.
Collapse
Affiliation(s)
- Yang Zhang
- Department of RehabilitationThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Zhiyuan Zheng
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Shilu Zhu
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Liang Xu
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Qingdong Zhang
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Jie Gao
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Min Ye
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Shuwei Shen
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Jinyu Xing
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Ming Wu
- Department of RehabilitationThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Ronald X. Xu
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| |
Collapse
|
20
|
Yang L, Shi F, Cao F, Wang L, She J, He B, Xu X, Kong L, Cai B. Neutrophils in Tissue Injury and Repair: Molecular Mechanisms and Therapeutic Targets. MedComm (Beijing) 2025; 6:e70184. [PMID: 40260014 PMCID: PMC12010766 DOI: 10.1002/mco2.70184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Tissue repair represents a highly intricate and ordered dynamic process, critically reliant on the orchestration of immune cells. Among these, neutrophils, the most abundant leukocytes in the body, emerge as the initial immune responders at injury sites. Traditionally recognized for their antimicrobial functions in innate immunity, neutrophils now garner attention for their indispensable roles in tissue repair. This review delves into their novel functions during the early stages of tissue injury. We elucidate the mechanisms underlying neutrophil recruitment and activation following tissue damage and explore their contributions to vascular network formation. Furthermore, we investigate the pivotal role of neutrophils during the initial phase of repair across different tissue types. Of particular interest is the investigation into how the fate of neutrophils influences overall tissue healing outcomes. By shedding light on these emerging aspects of neutrophil function in tissue repair, this review aims to pave the way for novel strategies and approaches in future organ defect repair, regeneration studies, and advancements in tissue engineering. The insights provided here have the potential to significantly impact the field of tissue repair and regeneration.
Collapse
Affiliation(s)
- Luying Yang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fan Shi
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Feng Cao
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Le Wang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Jianzhen She
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Boling He
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiaoying Xu
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Liang Kong
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bolei Cai
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
21
|
Wei S, Dai Z, Wu L, Xiang Z, Yang X, Jiang L, Du Z. Lactate-induced macrophage HMGB1 lactylation promotes neutrophil extracellular trap formation in sepsis-associated acute kidney injury. Cell Biol Toxicol 2025; 41:78. [PMID: 40304798 PMCID: PMC12043764 DOI: 10.1007/s10565-025-10026-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Neutrophils play a key role in sepsis-associated acute kidney injury (SAKI), a common and life-threatening complication of organ failure. High mobility group box 1 (HMGB1) modulates inflammatory responses and the formation of neutrophil extracellular traps (NETs). The present work aimed to explore whether HMGB1 lactylation promotes NET formation and exacerbates SAKI. METHODS Venous blood samples were collected from healthy volunteers and SAKI patients. A SAKI mouse model was established using the cecal ligation and puncture method. A coculture system of macrophage-derived exosomes and neutrophils was established. Macrophage-derived exosomes were isolated and identified. ELISAs, immunofluorescence staining, coimmunoprecipitation, and Western blotting were utilized to determine protein levels. RESULTS Elevated blood lactate levels were associated with increased HMGB1 levels in patients with SAKI. In mouse models, lactate increased HMGB1 expression, promoted NET formation, and exacerbated SAKI. Lactate stimulated M1 macrophages to secrete exosomes, leading to the accumulation and release of HMGB1 in the cytoplasm. Additionally, lactate promoted HMGB1 lactylation in macrophages, triggering the release of mitochondrial DNA from neutrophils and activating the cyclic GMP‒AMP synthase/stimulator of interferon genes pathway. CONCLUSION This study revealed that lactate-induced HMGB1 lactylation in macrophages plays a role in promoting NET formation in SAKI through the cGAS/STING pathway. These findings suggest that HMGB1 could be a potential target for therapeutic intervention in SAKI.
Collapse
Affiliation(s)
- Siwei Wei
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zijuan Dai
- Department of Anesthesiology, The Fourth Hospital of Changsha (Affiliated Changsha Hospital of Hunan Normal University), Changsha City, Hunan Province, China
| | - Lei Wu
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zhen Xiang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Xiaoxiao Yang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Liubing Jiang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zhen Du
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China.
| |
Collapse
|
22
|
Tu S, Jing X, Bu X, Zhang Q, Liao S, Zhu X, Guo Y, Sha W. Identification of pyroptosis-associated gene to predict fibrosis and reveal immune characterization in non-alcoholic fatty liver disease. Sci Rep 2025; 15:14944. [PMID: 40301412 PMCID: PMC12041580 DOI: 10.1038/s41598-025-96158-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/26/2025] [Indexed: 05/01/2025] Open
Abstract
Despite advances in research, studies on predictive models for Non-Alcoholic Fatty Liver Disease (NAFLD)-related fibrosis remain limited. Identifying new biomarkers to distinguish Non-Alcoholic Steatohepatitis (NASH) from NAFLD would aid in the treatment of NASH. Gene expression and clinical profiles of NAFL and NASH patients were collected from databases. Differentially expressed genes with prognostic value were used to construct predictive model. Validation of fibrosis stage-related pyroptosis-related genes (PRGs) was performed using Sprague-Dawley rats liver fibrosis models induced by CCl4 or PS. Immune cell infiltration assessment demonstrated that stromal score, immune score, and ESTIMATE score were higher in patients with NASH compared to those with NAFL. BAX, BAK1, PYCARD, and NLRP3 were identified as hub genes that exhibit a strong correlation with fibrosis stage. Additionally, the expression of these genes was increased in fibrotic liver tissues induced by CCl4 and PS. The pyroptosis-associated gene signature effectively predicts the degree of liver fibrosis in NASH patients. Our study indicates that BAX, BAK1, PYCARD, and NLRP3 might serve as biomarkers for NASH-associated fibrosis.
Collapse
Affiliation(s)
- Sha Tu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Xi Jing
- School of Nursing, Jinan University, Guangzhou, 510632, China
| | - Xiaoling Bu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Qingfang Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Shanying Liao
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Xiaobo Zhu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Ying Guo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
23
|
Zhang X, Huang X, Zhang X, Lai L, Zhu B, Lin P, Kang Z, Yin D, Tian D, Chen Z, Gao J. The miR-941/FOXN4/TGF-β feedback loop induces N2 polarization of neutrophils and enhances tumor progression of lung adenocarcinoma. Front Immunol 2025; 16:1561081. [PMID: 40352924 PMCID: PMC12061992 DOI: 10.3389/fimmu.2025.1561081] [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: 01/23/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a major subtype of lung cancer and one of the deadliest cancers in humans. Dysregulation of miRNA activity in tumor-associated neutrophils (TANs) in the tumor microenvironment plays an important role in the occurrence and development of LUAD. Method In this study, the miReact algorithm was used to analyze the single-cell RNA sequencing data of LUAD samples to reveal the miRNA profile characteristics of TANs in LUAD patients. The function of miR-941 was investigated in vivo and in vitro. The target gene and underlying signaling pathway of miR-941 were predicted and validated with qPCR, luciferase assay, WB and ELISA assay. Results The results indicated the crucial role of TANs, especially N2-TANs in LUAD and miR-941 activity was significantly upregulated in TANs of LUAD patients. MiR-941 overexpression promoted the proliferation, invasion, migration and anti-apoptosis of A549 and H1299. In vivo xenograft mouse model confirmed that miR-941 overexpression enhanced the growth of tumors formed by H1299 cells. Bioinformatics analysis showed that miR-941 targeted the tumor suppressor gene FOXN4, and we confirmed that FOXN4 overexpression could counteract the malignant effects of miR-941. In addition, miR-941 may drive LUAD progression through the FOXN4/TGF-β feedback signaling loop and participate in the N2-TAN polarization. Conclusion In summary, these findings reveal the key role of N2-TANs and the miR-941/FOXN4/TGF-β signaling loop in LUAD progression and provide potential therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Xitong Huang
- Department of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, China
| | - Xianying Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lichang Lai
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Baoyi Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Peibin Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Zhanfang Kang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Dazhong Yin
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Dongbo Tian
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Zisheng Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Jun Gao
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
24
|
Smith AK, Katrinli S, Maihofer AX, Aiello AE, Baker DG, Boks MP, Brick LA, Chen CY, Dalvie S, Fani N, Fortier CB, Gelernter J, Geuze E, Gillespie CF, Hayes JP, Hong S, Kessler RC, King AP, Koen N, Koenen KC, Liberzon I, Linnstaedt SD, McLean SA, Michopoulos V, Milberg WP, Miller MW, Mufford MS, Nugent NR, Orcutt HK, Powers A, Rauch SAM, Ressler KJ, Risbrough VB, Rutten BPF, Smoller JW, Stein DJ, Stein MB, Ursano RJ, Verfaellie MH, Vermetten E, Vinkers CH, Wani AH, WareVinkers EB, Wildman DE, Wolf EJ, Zhao Y, Logue MW, Nievergelt CM, Uddin M, Zannas AS. Cell-type-specific and inflammatory DNA methylation patterns associated with PTSD. Brain Behav Immun 2025; 128:540-548. [PMID: 40286993 DOI: 10.1016/j.bbi.2025.04.031] [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: 12/06/2024] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Epigenetic modifications, including DNA methylation (DNAm), can change in response to traumatic stress exposure, and may help to distinguish between individuals with and without PTSD. Here, we examine the DNAm patterns specific to immune cell types and inflammation in those with PTSD. METHODS This study includes 3,277 participants from 11 cohorts participating in the Psychiatric Genomics Consortium (PGC) PTSD Epigenetics Workgroup. DNAm was assayed from blood with the MethylationEPIC BeadChip. A standardized QC pipeline was applied and used to impute cell composition. Within each cohort, we identified cell-type-specific DNAm patterns associated with PTSD, controlling for sex (if applicable), age, and ancestry. Meta-analyses were performed from summary statistics. RESULTS PTSD cases had lower proportions of B cells and NK cells as well as higher proportions of neutrophils when compared to trauma-exposed controls. Overall, we identified 96 PTSD-associated CpGs across six types of immune cells. Most of these differences were identified in B cells, with 95 % exhibiting lower methylation levels in those with PTSD. Interestingly, the PTSD-associated CpGs annotated to a gene in B cells were enriched in a recent GWAS of PTSD (p < 0.0001). CONCLUSIONS This study identifies novel PTSD-associated CpGs in individual immune cell types and supports the role of immune dysregulation and inflammation in PTSD.
Collapse
Affiliation(s)
- Alicia K Smith
- Emory University, Department of Gynecology and Obstetrics, Atlanta, GA, USA; Emory University, Department of Human Genetics, Atlanta, GA, USA; Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA.
| | - Seyma Katrinli
- Emory University, Department of Gynecology and Obstetrics, Atlanta, GA, USA
| | - Adam X Maihofer
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA; Veterans Affairs San Diego Healthcare System, Research Service, San Diego, CA, USA
| | - Allison E Aiello
- Columbia University, Robert N Butler Columbia Aging Center, Department of Epidemiology, New York, NY, USA
| | - Dewleen G Baker
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA
| | - Marco P Boks
- Amsterdam UMC, Department of Psychiatry, Amsterdam, NH, Netherland; Dimence Specialised Mental Health, Mood Disorders, Deventer, OV, Netherland; University Medical Center Utrecht, Department Psychiatry, Utrecht, UT, Netherland
| | - Leslie A Brick
- Alpert Medical School of Brown University, Department of Psychiatry and Human Behavior, Providence, RI, USA
| | - Chia-Yen Chen
- Biogen Inc., Translational Medicine, Cambridge, MA, USA
| | - Shareefa Dalvie
- University of Cape Town, Division of Human Genetics, Cape Town, Western Province, South Africa
| | - Negar Fani
- Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Catherine B Fortier
- Harvard Medical School, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Boston, MA, USA; VA Boston Healthcare System, Translational Research Center for Traumatic Brain Injury and Stress Disorders (TRACTS), Boston, MA, USA
| | - Joel Gelernter
- VA Connecticut Healthcare Center, Psychiatry Service, West Haven, CT, USA; Yale University School of Medicine, Departments of Psychiatry, Genetics and Neuroscience, New Haven, CT, USA
| | - Elbert Geuze
- Netherlands Ministry of Defence, Brain Research and Innovation Centre, Utrecht, UT, Netherland; UMC Utrecht Brain Center Rudolf Magnus, Department of Psychiatry, Utrecht, UT, Netherland
| | - Charles F Gillespie
- Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Jasmeet P Hayes
- The Ohio State University, Department of Psychology, Columbus, OH, USA
| | - Suzi Hong
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA; University of California San Diego, Herbert Wertheim School of Public Health and Human Longevity Science, La Jolla, CA, USA
| | - Ronald C Kessler
- Harvard Medical School, Department of Health Care Policy, Boston, MA, USA
| | - Anthony P King
- The Ohio State University, College of Medicine, Institute for Behavioral Medicine Research, Columbus, OH, USA; The Ohio State University, College of Medicine, Psychiatry & Behavioral Health, Columbus, OH, USA
| | - Nastassja Koen
- University of Cape Town, Department of Psychiatry & Mental Health, Cape Town, Western Province, South Africa; University of Cape Town, Neuroscience Institute, Cape Town, Western Province, South Africa; University of Cape Town, SA MRC Unit on Risk & Resilience in Mental Disorders, Cape Town, Western Province, South Africa
| | - Karestan C Koenen
- Broad Institute of MIT and Harvard, Stanley Center for Psychiatric Research, Cambridge, MA, USA; Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA; Massachusetts General Hospital, Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Boston, MA, USA
| | - Israel Liberzon
- Texas A&M University College of Medicine, Department of Psychiatry and Behavioral Sciences, Bryan, TX, USA
| | - Sarah D Linnstaedt
- University of North Carolina at Chapel Hill, Department of Anesthesiology, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Institute for Trauma Recovery, Chapel Hill, NC, USA
| | - Samuel A McLean
- University of North Carolina at Chapel Hill, Institute for Trauma Recovery, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Department of Psychiatry, Chapel Hill, NC, USA
| | - Vasiliki Michopoulos
- Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - William P Milberg
- Harvard Medical School, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Boston, MA, USA; VA Boston Healthcare System, Translational Research Center for Traumatic Brain Injury and Stress Disorders (TRACTS), Boston, MA, USA
| | - Mark W Miller
- Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, National Center for PTSD, Boston, MA, USA
| | - Mary S Mufford
- University of Cape Town, Department of Psychiatry and Mental Health, Cape Town, Western Province, South Africa
| | - Nicole R Nugent
- Alpert Brown Medical School, Department of Emergency Medicine, Providence, RI, USA; Alpert Brown Medical School, Department of Pediatrics, Providence, RI, USA; Alpert Brown Medical School, Department of Psychiatry and Human Behavior, Providence, RI, USA
| | - Holly K Orcutt
- Northern Illinois University, Department of Psychology, DeKalb, IL, USA
| | - Abigail Powers
- Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Sheila A M Rauch
- Emory University School of Medicine, Department of Psychiatry & Behavioral Sciences, Atlanta, GA, USA; Joseph Maxwell Cleland Atlanta Veterans Affairs Healthcare System, Mental Health Service Line, Atlanta, GA, USA
| | - Kerry J Ressler
- Emory University, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA; Harvard Medical School, Department of Psychiatry, Boston, MA, USA; McLean Hospital, Division of Depression and Anxiety, Belmont, MA, USA
| | - Victoria B Risbrough
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA; Veterans Affairs San Diego Healthcare System, Research Service, San Diego, CA, USA
| | - Bart P F Rutten
- Broad Institute of MIT and Harvard, Stanley Center for Psychiatric Research, Cambridge, MA, USA; Maastricht University, Department of Psychiatry and Neuropsychology, Maastricht, Limburg, Netherland
| | - Jordan W Smoller
- Broad Institute of MIT and Harvard, Stanley Center for Psychiatric Research, Cambridge, MA, USA; Massachusetts General Hospital, Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Boston, MA, USA; Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA
| | - Dan J Stein
- University of Cape Town, Department of Psychiatry & Mental Health, Cape Town, Western Province, South Africa; University of Cape Town, Neuroscience Institute, Cape Town, Western Province, South Africa; University of Cape Town, SA MRC Unit on Risk & Resilience in Mental Disorders, Cape Town, Western Province, South Africa
| | - Murray B Stein
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA; University of California San Diego, School of Public Health, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, Psychiatry Service, San Diego, CA, USA
| | - Robert J Ursano
- Uniformed Services University of Health Sciences, Center for the Study of Traumatic Stress, Department of Psychiatry, Bethesda, MD, USA
| | - Mieke H Verfaellie
- Boston University School of Medicine, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, Memory Disorders Research Center, Boston, MA, USA
| | - Eric Vermetten
- Leiden University Medical Center, Department of Psychiatry, Leiden, ZH, Netherland; New York University School of Medicine, Department of Psychiatry, New York, NY, USA
| | - Christiaan H Vinkers
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, NH, Netherland; Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam, Holland, Netherland; Amsterdam University Medical Center, Amsterdam Neuroscience Research Institute, Mood, Anxiety, Psychosis, Stress & Sleep Program, Amsterdam, NH, Netherland
| | - Agaz H Wani
- University of South Florida College of Public Health, Genomics Program, Tampa, FL, USA
| | | | - Derek E Wildman
- Boston University Chobanian & Avedisian School of Medicine, Department of Biomedical Genetics, Boston, MA, USA
| | - Erika J Wolf
- Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, National Center for PTSD, Boston, MA, USA
| | - Ying Zhao
- University of North Carolina at Chapel Hill, Department of Anesthesiology, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Institute for Trauma Recovery, Chapel Hill, NC, USA
| | - Mark W Logue
- Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA; VA Boston Healthcare System, National Center for PTSD, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA; University of North Carolina at Chapel Hill, Carolina Stress Initiative, Chapel Hill, NC, USA
| | - Caroline M Nievergelt
- University of California San Diego, Department of Psychiatry, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA; Veterans Affairs San Diego Healthcare System, Research Service, San Diego, CA, USA
| | - Monica Uddin
- University of South Florida College of Public Health, Genomics Program, Tampa, FL, USA
| | - Anthony S Zannas
- University of North Carolina at Chapel Hill, Institute for Trauma Recovery, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Department of Psychiatry, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Carolina Stress Initiative, Chapel Hill, NC, USA; University of North Carolina at Chapel Hill, Department of Genetics, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Ledderose C, Valsami EA, Elevado M, Stevenson A, Abutabikh R, Curatolo J, Junger WG. Adenosine accumulation in the blood of newborn mice weakens antimicrobial host defenses. J Leukoc Biol 2025; 117:qiaf003. [PMID: 39824218 PMCID: PMC12022637 DOI: 10.1093/jleuko/qiaf003] [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: 05/23/2024] [Revised: 10/30/2024] [Accepted: 01/16/2025] [Indexed: 01/20/2025] Open
Abstract
Pediatric intensive care patients are particularly susceptible to severe bacterial infections because of ineffective neutrophil responses. The reasons why neutrophils of newborns are less responsive than those of adults are not clear. Because adenosine triphosphate and adenosine tightly regulate neutrophils, we studied whether the adenosine triphosphate and adenosine levels in the blood of newborn mice could impair the function of their neutrophils. We observed significant changes in plasma adenosine triphosphate and adenosine levels throughout the lifespan of mice. Adenosine levels in newborns were significantly higher than in older mice, while adenosine triphosphate levels were significantly lower. These changes were particularly striking in newborn and juvenile mice with adenosine triphosphate and adenosine levels of about 80 and 600 nM in newborns vs 130 and 190 nM in juveniles, respectively. The ratios of the adenosine triphosphate vs adenosine levels of newborns were (with 0.2) significantly lower than those of juveniles (1.4) and adults (0.5). These low adenosine triphosphate/adenosine ratios correlated with significantly weakened neutrophil activation responses following in vitro stimulation with a formyl peptide receptor agonist and a markedly higher morbidity and mortality rate of newborns following bacterial infection. We found that enhanced adenosine monophosphate hydrolysis via CD73, a lack of adenosine breakdown by adenosine deaminase, and reduced adenosine uptake by nucleoside transporters are responsible for the low adenosine triphosphate/adenosine ratios in blood of newborn mice. We conclude that the extracellular adenosine accumulation in newborn mice impairs inflammatory responses and reduces the ability of neutrophils to mount effective antimicrobial defenses against bacterial infections.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Eleftheria-Angeliki Valsami
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Mark Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Ava Stevenson
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Reem Abutabikh
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Julian Curatolo
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Wolfgang G Junger
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| |
Collapse
|
26
|
Son S, Xu C, Jang J, Dinh M, Skorobogatko Y, Fu H, Valentine JM, An G, Ying W, Yu RT, Downes M, Evans RM, Saltiel AR. Sympathetic activation of white adipose tissue recruits neutrophils to limit energy expenditure. RESEARCH SQUARE 2025:rs.3.rs-6414640. [PMID: 40321773 PMCID: PMC12047989 DOI: 10.21203/rs.3.rs-6414640/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Adipose tissue maintains energy homeostasis by storing lipids during nutrient surplus and releasing them through lipolysis in times of energy demand. While lipolysis is essential for short term metabolic adaptation, prolonged metabolic stress requires adaptive changes that preserve energy reserves. Here, we report that β-adrenergic activation of adipocytes induces a transient and depot-specific infiltration of neutrophils into white adipose tissue (WAT), particularly in lipid-rich visceral WAT. Neutrophil recruitment requires the stimulation of both lipolysis and p38 MAPK activation in adipocytes. Recruited neutrophils locally secrete IL-1β, which suppresses lipolysis and limits excessive energy expenditure. Neutrophil depletion or blockade of IL-1β production increased lipolysis, leading to reduced WAT mass upon repeated β3-adrenergic stimulation. Together, these findings reveal an unexpected role of neutrophil-derived IL-1β in preserving lipid stores during metabolic stress, highlighting a physiological function of innate immune cells in maintaining energy homeostasis.
Collapse
Affiliation(s)
- Seunghwan Son
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Cindy Xu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Janice Jang
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Maddox Dinh
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Yuliya Skorobogatko
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Haipeng Fu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Joseph M. Valentine
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Garam An
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Alan R. Saltiel
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
27
|
Gao C, Liu M, Sun Y, Zhao Z, Wei F, Xu X. Association between advanced lung cancer inflammation index and gallstone prevalence among U.S. adults: A population-based study. PLoS One 2025; 20:e0321733. [PMID: 40233085 PMCID: PMC11999145 DOI: 10.1371/journal.pone.0321733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
INTRODUCTION Gallstones are a common digestive disorder, with a global prevalence of 10%-15%, posing a significant economic burden on public health. The formation of gallstones is closely associated with inflammation and nutritional status. The Advanced Lung Cancer Inflammation Index (ALI) is a composite measure for assessing inflammation and nutritional status; however, its relationship with gallstone risk remains unclear. This study aims to investigate the association between ALI and gallstone prevalence among U.S. adults. METHODS This study is based on data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES) and includes 5,826 adults aged 20 years and older. The Advanced Lung Cancer Inflammation Index (ALI) was calculated using body mass index (BMI), serum albumin levels, and the neutrophil-to-lymphocyte ratio (NLR). The prevalence of gallstones was determined through questionnaire surveys. Multivariable logistic regression models were employed to analyze the relationship between ALI and the risk of gallstones. Additionally, trend analysis, smooth curve fitting, and subgroup analyses were conducted. RESULTS The study results showed a significant positive correlation between ALI levels and the risk of gallstone disease. After fully adjusting for covariates, each unit increase in lnALI was associated with a 42% increase in the risk of gallstone disease (OR = 1.42, 95% CI: 1.12-1.80). Trend analysis indicated a significant dose-response relationship between ALI and gallstone risk (P for trend < 0.01). Subgroup analysis further revealed that the correlation between ALI and gallstone risk was more pronounced in females, non-diabetic patients, individuals with higher education levels, those with insufficient physical activity, and non-drinkers, with gender showing a significant interaction effect (interaction P < 0.05). Smooth curve fitting further validated the linear relationship between ALI and gallstone risk, and this association was particularly prominent in the female population. CONCLUSIONS This study demonstrates that ALI is significantly associated with the risk of gallstones, particularly among women. As a simple and readily accessible indicator, ALI may help identify high-risk populations and provide a new clinical tool for the prevention and management of gallstones. Future longitudinal studies should further validate these findings and evaluate the predictive value of ALI across different populations.
Collapse
Affiliation(s)
- Chaofeng Gao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | | | - Yuan Sun
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Zekun Zhao
- Department of Cardiovascular Surgery, Xi’an Third Hospital, Xi’an, China
| | - Fengxian Wei
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaodong Xu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
28
|
Wang N, Chen SQ, Tang YJ, Chen QZ, Wang JH, Bai YF, Zhou YL, Xu HH, Li WL, Chen J, Cui JH, Wang YC, Zhang YL, Yu Y. Identification of the key genes in Sanhe cattle for health and milk composition traits based on the WGCNA. J Dairy Sci 2025:S0022-0302(25)00214-0. [PMID: 40221034 DOI: 10.3168/jds.2024-25842] [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: 10/17/2024] [Accepted: 03/08/2025] [Indexed: 04/14/2025]
Abstract
Sanhe cattle are domestically bred dual-purpose (i.e., milk and meat) cattle in northeast China that exhibit exceptional adaptability, resilience, and milk composition traits. Nevertheless, few studies that have analyzed the transcriptome of Sanhe cattle and elucidated the key pathways and genes accountable for its immune and production traits. Weighted gene co-expression network analysis is effective for cattle genetic studies. In this study, with Holstein cattle (n = 82) serving as the dairy breed, we aimed to investigate the manifestations and regulatory pathways of the health and milk composition traits for the Sanhe cattle population (n = 61). We employed principal composition analysis and pathway scoring to compare the distinct characteristics between the 2 cattle breeds using transcriptome, routine blood examination, milk composition, and dairy herd improvement data. We then identified 20 hub genes and potentially crucial pathways that were specifically and significantly correlated with the health and milk composition traits of Sanhe cattle by weighted co-expression network analysis and enrichment analysis. We discovered 56 hub genes that might give rise to commonalities and differences in health and milk composition between Sanhe cattle and Holstein cattle. Finally, we examined the differences in hub gene expression in health and milk composition traits between Sanhe cattle and Holstein cattle using correlation analysis. Overall, this study establishes a foundation for further exploration of the molecular markers related to the health and milk composition traits of Sanhe cattle.
Collapse
Affiliation(s)
- N Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - S Q Chen
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Y J Tang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Q Z Chen
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - J H Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Y F Bai
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Y L Zhou
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - H H Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - W L Li
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - J Chen
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Hulunbuir 021012, China
| | - J H Cui
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Hulunbuir 021012, China
| | - Y C Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Y L Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Y Yu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
29
|
Irakli K, Ekaterina K, Mladen K. The immunosuppressive role of neutrophils in infectious and oncological conditions: A study of chemokine receptor CXCR3 and human neutrophil lipocalin levels. BMC Res Notes 2025; 18:148. [PMID: 40200380 PMCID: PMC11980113 DOI: 10.1186/s13104-025-07229-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Neutrophils are key players in the innate immune system, responsible for rapid responses to infections through mechanisms such as phagocytosis and the release of reactive oxygen species (ROS). Beyond their role in host defense, neutrophils also contribute to the pathogenesis of various diseases, including infections, metabolic disorders, autoimmune diseases, and cancer. Understanding the immunosuppressive role of neutrophils, particularly through markers like human neutrophil lipocalin (HNL) and the chemokine receptor CXCR3, is crucial for developing targeted therapeutic strategies. MATERIALS AND METHODS This study involved 200 participants divided into four groups: 50 patients with acute respiratory infection, 50 COVID-19 recovered patients, 50 oncology patients, and 50 healthy donors as controls. Peripheral blood samples were collected and analyzed using enzyme-linked immunoassay (ELISA) to quantify levels of HNL and CXCR3. Data were analyzed using SPSS version 25.0, employing descriptive statistics, the Shapiro-Wilk test for normality, one-way ANOVA for normally distributed variables, and the Kruskal-Wallis test for non-normally distributed variables. Post-hoc comparisons were conducted using Tukey's HSD and Dunn's tests. RESULTS CXCR3 levels were stable across groups, with no significant differences found. Acute respiratory infection patients had an average CXCR3 level of 150 ± 20 pg/ml, while COVID-19 recovered patients had slightly lower levels at 140 ± 18 pg/ml. Oncology patients had elevated CXCR3 levels at 160 ± 22 pg/ml, similar to healthy donors at 150 ± 19 pg/ml. HNL levels varied more, with COVID-19 recovered patients showing notably lower levels (100 ± 12 ng/ml) compared to other groups. Oncology patients exhibited higher HNL levels, especially those with prostate cancer (150 ± 20 ng/ml). CONCLUSION The findings highlight the consistent expression of CXCR3 across various conditions, making it a reliable marker for immune response assessment. The distinct HNL profiles, particularly the lower levels in COVID-19 recovered patients and higher levels in prostate cancer patients, suggest unique neutrophil activities and immune responses. These insights into neutrophil-mediated immunosuppression and inflammation could inform the development of targeted therapies for infections, cancer, and autoimmune diseases. Further research is needed to elucidate the specific mechanisms underlying neutrophil-induced immunosuppression.
Collapse
Affiliation(s)
- Khuntsaria Irakli
- Petre Shotadze Tbilisi Medical Academy, Ketevan Tsamebuli avenue 51/2, Tbilisi, 0144, Georgia
| | - Kldiashvili Ekaterina
- Petre Shotadze Tbilisi Medical Academy, Ketevan Tsamebuli avenue 51/2, Tbilisi, 0144, Georgia.
| | - Krajacic Mladen
- Petre Shotadze Tbilisi Medical Academy, Ketevan Tsamebuli avenue 51/2, Tbilisi, 0144, Georgia
- University of Zagreb, Ul. Radoslava Cimermana 88, Zagreb, 10000, Croatia
| |
Collapse
|
30
|
Canè S, Geiger R, Bronte V. The roles of arginases and arginine in immunity. Nat Rev Immunol 2025; 25:266-284. [PMID: 39420221 DOI: 10.1038/s41577-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Arginase activity and arginine metabolism in immune cells have important consequences for health and disease. Their dysregulation is commonly observed in cancer, autoimmune disorders and infectious diseases. Following the initial description of a role for arginase in the dysfunction of T cells mounting an antitumour response, numerous studies have broadened our understanding of the regulation and expression of arginases and their integration with other metabolic pathways. Here, we highlight the differences in arginase compartmentalization and storage between humans and rodents that should be taken into consideration when assessing the effects of arginase activity. We detail the roles of arginases, arginine and its metabolites in immune cells and their effects in the context of cancer, autoimmunity and infectious disease. Finally, we explore potential therapeutic strategies targeting arginases and arginine.
Collapse
Affiliation(s)
- Stefania Canè
- The Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Università della Svizzera italiana, Bellinzona, Switzerland
| | | |
Collapse
|
31
|
Martínez‐López J, Ortiz‐Fernandez L, Estupiñán‐Moreno E, Kerick M, Andrés‐León E, Terron‐Camero LC, Carnero‐Montoro E, Barturen G, Beretta L, Almeida I, Alarcón‐Riquelme ME, Ballestar E, Acosta‐Herrera M, Martín J. A Strong Dysregulated Myeloid Component in the Epigenetic Landscape of Systemic Sclerosis: An Integrated DNA Methylome and Transcriptome Analysis. Arthritis Rheumatol 2025; 77:439-449. [PMID: 39468422 PMCID: PMC11936501 DOI: 10.1002/art.43044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/10/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE Nongenetic factors influence systemic sclerosis (SSc) pathogenesis, underscoring epigenetics as a relevant contributor to the disease. We aimed to unravel DNA methylation abnormalities associated with SSc through an epigenome-wide association study. METHODS We analyzed DNA methylation data from whole-blood samples in 179 patients with SSc and 241 unaffected individuals to identify differentially methylated positions (DMPs) with a false discovery rate (FDR) <0.05. These results were further integrated with RNA sequencing data from the same patients to assess their functional consequence. Additionally, we examined the impact of DNA methylation changes on transcription factors and analyzed the relationship between alterations of the methylation and gene expression profile and serum proteins levels. RESULTS This analysis yielded 525 DMPs enriched in immune-related pathways, with leukocyte cell-cell adhesion being the most significant (FDR = 4.91 × 10-9), prioritizing integrins as they were exposed by integrating methylome and transcriptome data. Furthermore, through this integrative approach, we observed an enrichment of neutrophil-related pathways, highlighting this myeloid cell type as a relevant contributor in SSc pathogenesis. In addition, we uncovered novel profibrotic and proinflammatory mechanisms involved in the disease. Finally, the altered epigenetic and transcriptomic signature revealed an increased activity of CCAAT/enhancer-binding protein transcription factor family in SSc, which is crucial in the myeloid lineage development. CONCLUSION Our findings uncover the impaired epigenetic regulation of the disease and its impact on gene expression, identifying new molecules for potential clinical applications and improving our understanding of SSc pathogenesis.
Collapse
Affiliation(s)
- Javier Martínez‐López
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones Científicas and Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria de GranadaGranadaSpain
| | - Lourdes Ortiz‐Fernandez
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones CientíficasGranadaSpain
| | | | - Martin Kerick
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones CientíficasGranadaSpain
| | - Eduardo Andrés‐León
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones CientíficasGranadaSpain
| | - Laura C. Terron‐Camero
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones CientíficasGranadaSpain
| | - Elena Carnero‐Montoro
- Centre for Genomics and Oncological Research, Pfizer, University of Granada/Andalusian Regional GovernmentGranadaSpain
| | - Guillermo Barturen
- Centre for Genomics and Oncological Research, Pfizer, University of Granada/Andalusian Regional GovernmentGranadaSpain
| | - Lorenzo Beretta
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di MilanoMilanItaly
| | - Isabel Almeida
- Centro Hospitalar Universitário do Porto and Instituto de Ciências Biomédicas Abel Salazar, Universidade do PortoPortoPortugal
| | - Marta E. Alarcón‐Riquelme
- Centre for Genomics and Oncological Research, Pfizer, University of Granada/Andalusian Regional GovernmentGranadaSpain
| | - Esteban Ballestar
- Josep Carreras Research Institute, Barcelona, Spain, and Health Science Center, East China Normal UniversityShanghaiChina
| | - Marialbert Acosta‐Herrera
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones Científicas and Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria de GranadaGranadaSpain
| | - Javier Martín
- Institute of Parasitology and Biomedicine López‐Neyra, Consejo Superior de Investigaciones CientíficasGranadaSpain
| |
Collapse
|
32
|
Chalmers JD, Metersky M, Aliberti S, Morgan L, Fucile S, Lauterio M, McDonald PP. Neutrophilic inflammation in bronchiectasis. Eur Respir Rev 2025; 34:240179. [PMID: 40174958 PMCID: PMC11962982 DOI: 10.1183/16000617.0179-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/11/2025] [Indexed: 04/04/2025] Open
Abstract
Noncystic fibrosis bronchiectasis, hereafter referred to as bronchiectasis, is a chronic, progressive lung disease that can affect people of all ages. Patients with clinically significant bronchiectasis have chronic cough and sputum production, as well as recurrent respiratory infections, fatigue and impaired health-related quality of life. The pathophysiology of bronchiectasis has been described as a vicious vortex of chronic inflammation, recurring airway infection, impaired mucociliary clearance and progressive lung damage that promotes the development and progression of the disease. This review describes the pivotal role of neutrophil-driven inflammation in the pathogenesis and progression of bronchiectasis. Delayed neutrophil apoptosis and increased necrosis enhance dysregulated inflammation in bronchiectasis and failure to resolve this contributes to chronic, sustained inflammation. The excessive release of neutrophil serine proteases, such as neutrophil elastase, cathepsin G and proteinase 3, promotes a protease-antiprotease imbalance that correlates with increased inflammation in bronchiectasis and contributes to disease progression. While there are currently no licensed therapies to treat bronchiectasis, this review will explore the evolving evidence for neutrophilic inflammation as a novel treatment target with meaningful clinical benefits.
Collapse
Affiliation(s)
- James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Mark Metersky
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - Lucy Morgan
- Department of Respiratory Medicine, Concord Clinical School, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
33
|
Mandal M, Mamun MAA, Rakib A, Kumar S, Park F, Hwang DJ, Li W, Miller DD, Singh UP. Modulation of occludin, NF-κB, p-STAT3, and Th17 response by DJ-X-025 decreases inflammation and ameliorates experimental colitis. Biomed Pharmacother 2025; 185:117939. [PMID: 40036995 DOI: 10.1016/j.biopha.2025.117939] [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: 11/20/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
SCOPE Inflammatory bowel disease (IBD) involves a range of immune-mediated disorders marked by systemic and local intestinal inflammation. We synthesized a novel compound DJ-X-025 and uncovered its anti-inflammatory properties using lipopolysaccharide (LPS)-induced RAW 264.7 macrophages in vitro and a dextran sodium sulfate (DSS)-induced model of colitis. METHODS AND RESULTS We evaluated the alteration in cell morphology, cytoskeletal proteins, and inflammatory markers of DJ-X-025 treated LPS-stimulated RAW 264.7 macrophages. We administered DJ-X-025 by oral gavage in DSS-induced colitis, examined colon histology, and alterations of immune cells by flow cytometry, and performed molecular studies using RT-qPCR and western blot analysis. DJ-X-025 treatment markedly altered the morphology of LPS-treated RAW 264.7 macrophages from elongated to round shapes, modulated actin and tubulin, and reduced the level of inflammatory markers like TNF-α, IL-1β, IL-6, and iNOS. Further, we observed that DJ-X-025 steered to improve colon length, muscularis mucosa thickness, and colon inflammatory score compared to the DSS group alone. DJ-X-025 effectively inverted the increased population of activated T cells, Th17, and macrophages in lamina propria by DSS treatment, leading to a substantial reduction in the inflammatory response in the colon. Strikingly, DJ-X-025 treatment enhanced the expression of occludin and diminished the expression of NF-κB and phosphorylation of STAT3 in the colon of DSS-treated mice compared to DSS-alone. Additionally, DJ-X-025 induced the expression of Foxp3 in the colon and, reduced systemic inflammatory cytokine/chemokine levels further supporting its immunomodulatory effects. These results suggest that DJ-X-025 is linked to the induction of occludin expression and decreased expression of p-STAT3/NF-κB and Th17 response in the colon, which together suppresses systemic and colon inflammatory cytokines for effective amelioration of experimental colitis. CONCLUSION These findings suggest that DJ-X-025 might be a promising therapeutic agent for the treatment of IBD.
Collapse
Affiliation(s)
- Mousumi Mandal
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Md Abdullah Al Mamun
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Ahmed Rakib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
34
|
Liu Z, Xu J, Wang X. Bioactive hemostatic materials: a new strategy for promoting wound healing and tissue regeneration. MedComm (Beijing) 2025; 6:e70113. [PMID: 40123833 PMCID: PMC11928890 DOI: 10.1002/mco2.70113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/25/2025] Open
Abstract
Wound healing remains a critical global healthcare challenge, with an annual treatment cost exceeding $50 billion worldwide. Over the past decade, significant advances in wound care have focused on developing sophisticated biomaterials that promote tissue regeneration and prevent complications. Despite these developments, there remains a crucial need for multifunctional wound healing materials that can effectively address the complex, multiphase nature of wound repair while being cost effective and easily applicable in various clinical settings. This review systematically analyzes the latest developments in wound healing materials, examining their chemical composition, structural design, and therapeutic mechanisms. We comprehensively evaluate various bioactive components, including natural polymers, synthetic matrices, and hybrid composites, along with their different forms, such as hydrogels, powders, and smart dressings. Special attention is given to emerging strategies in material design that integrate multiple therapeutic functions, including sustained drug delivery, infection prevention, and tissue regeneration promotion. The insights provided in this review illuminate the path toward next-generation wound healing materials, highlighting opportunities for developing more effective therapeutic solutions that can significantly improve patient outcomes and reduce healthcare burden.
Collapse
Affiliation(s)
- Zhengyuan Liu
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijingChina
- Sino‐Danish CollegeUniversity of Chinese Academy of Sciences (UCAS)BeijingChina
- Nano‐Science CenterUniversity of CopenhagenCopenhagenDenmark
| | - Junnan Xu
- Department of Urologythe Third Medical Center of PLA General HospitalBeijingChina
| | - Xing Wang
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijingChina
| |
Collapse
|
35
|
Brodjonegoro SR, Rizal DM, Arfian N, Luzman RA, Pikatan NW, Robert R, Febriyanto T, Liliana B, Yogahutama N, Dwiaji IW. Association of MPV, NLR, PLR and CRP on testicular salvage in testicular torsion: A systematic review and meta-analysis. NARRA J 2025; 5:e1785. [PMID: 40352219 PMCID: PMC12059872 DOI: 10.52225/narra.v5i1.1785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/08/2025] [Indexed: 05/14/2025]
Abstract
Testicular torsion, a critical urological emergency caused by twisting of the spermatic cord, poses a risk of ischemia, particularly in children who often struggle to pinpoint symptoms onset. Delay in managing testicular torsion can lead to the need for orchiectomy. The aim of this study was to assess the association between hematologic parameters-mean platelet volume (MPV), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and C-reactive protein (CRP)-and testicular salvage in cases of testicular torsion. Four databases (PubMed, Embase (Ovid), Science Direct, and Scopus) were systematically searched for eligible studies published up to November 4, 2024. The primary outcome was testicular salvage. Sensitivity analysis was performed using leave- one-out plot. Subgroup analysis was performed based on age, country, region, duration to orchiopexy and duration to orchiectomy. Heterogeneity was examined using I 2 statistics, and a random-effect model was applied. Out of 363 studies identified, nine observational studies involving 796 patients were included, with 338 (42.3%) in orchiopexy group. The meta-analysis indicated that MPV value was significantly elevated in orchiectomy group (mean difference (MD): -0.4; 95% confidence interval (95%CI): -0.62-(-0.18); p < 0.01), with higher MPV levels associated with an increased likelihood of orchiectomy (odds ratio (OR): 2.12; 95%CI: 1.35-3.33; p < 0.01). NLR, PLR, and CRP showed no significant association with testicular salvage, as demonstrated by pooled MD and OR analyses (p > 0.05). No significant differences were observed after sensitivity and subgroup analysis (p > 0.05). These findings suggest that elevated MPV levels are associated with non- salvageable testis, requiring orchiectomy highlighting its potential utility in clinical evaluation for testicular torsion.
Collapse
Affiliation(s)
- Sakti R. Brodjonegoro
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Dicky M. Rizal
- Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nur Arfian
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Raedi A. Luzman
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Narpati W. Pikatan
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Robert Robert
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Toni Febriyanto
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Belinda Liliana
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Noka Yogahutama
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Iqbal W. Dwiaji
- Division of Urology, Department of Surgery, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Division of Urology, Department of Surgery, Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| |
Collapse
|
36
|
Liu Q, Zhao Y, Dong S, Bai X, Chen B, Liu X, Shen J, Zhu D. Characteristics of Neutrophil Migration and Function in Acute Inflammation Induced by Zymosan and Carrageenan in the Mice Air Pouch Model. Inflammation 2025; 48:607-620. [PMID: 38902540 DOI: 10.1007/s10753-024-02064-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
Deciphering the complex and redundant process of acute inflammation remains challenging. The failure of numerous clinical trials assessing anti-inflammation agents which had promising preclinical effects inevitably questions the validity of current animal models of inflammation. This study aimed to better understand the process of immune inflammatory response and to select more suitable models to evaluate the effect of potential anti-inflammatory drugs. Zymosan and λ-carrageenan are the most used representatives of particulate and soluble irritants that trigger acute inflammation in the air pouch inflammation model. When zymosan was used, the number of exudate cells first increased at 4 h-8 h, followed by a drop at 12 h-24 h. While, the changes in number of leukocytes in peripheral blood and proportion of neutrophils in bone marrow have the opposite trend. Meanwhile, neutrophils released neutrophil extracellular traps (NETs) to clean zymosan particles. In contrast, the cell migration response to carrageenan increased during 4 h to 24 h, no obvious NETs were observed, and the number of leukocytes in peripheral blood increased and the proportion of neutrophils in bone marrow decreased slightly. This study indicated that although both zymosan and carrageenan are sterile irritants, the characteristics of the inflammatory response induced by each other were different. In the acute phase of inflammation, zymosan-stimulated neutrophils were mobilized, recruited, and engulfed, and then died by NETs. Carrageenan stimulated the production of cytokines/chemokines by neutrophils or macrophages, but did not lead to an obvious death by releasing NETs.
Collapse
Affiliation(s)
- Qi Liu
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yubo Zhao
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Shuai Dong
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Xingyuan Bai
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Bin Chen
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Xijuan Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing), Core Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jing Shen
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing), Core Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Dan Zhu
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China.
| |
Collapse
|
37
|
Nigi L, Pedace E, Dotta F, Sebastiani G. Neutrophils in Type 1 Diabetes: Untangling the Intricate Web of Pathways and Hypothesis. Biomolecules 2025; 15:505. [PMID: 40305198 PMCID: PMC12025241 DOI: 10.3390/biom15040505] [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: 03/03/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Neutrophils are increasingly recognized as key contributors to the pathogenesis of Type 1 Diabetes (T1D), yet their precise mechanistic role in disease onset and progression remains incompletely understood. While these innate immune cells reside in pancreatic tissue and support tissue homeostasis under physiological conditions, they can also drive tissue damage by triggering innate immune responses and modulating inflammation. Within the inflammatory milieu, neutrophils establish complex, bidirectional interactions with various immune cells, including macrophages, dendritic cells, natural killer cells, and lymphocytes. Once activated, they may enhance the innate immune response through direct or indirect crosstalk with immune cells, antigen presentation, and β-cell destruction or dysfunction. These mechanisms underscore the multifaceted and dynamic role of neutrophils in T1D, shaped by their intricate immunological interactions. Further research into the diverse functional capabilities of neutrophils is crucial for uncovering novel aspects of their involvement in T1D, potentially revealing new therapeutic targets to modulate disease progression.
Collapse
Affiliation(s)
- Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Erika Pedace
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
- Tuscany Centre for Precision Medicine, 53100 Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| |
Collapse
|
38
|
Walker SL, Leete P, Boldison J. Tissue Resident and Infiltrating Immune Cells: Their Influence on the Demise of Beta Cells in Type 1 Diabetes. Biomolecules 2025; 15:441. [PMID: 40149976 PMCID: PMC11939886 DOI: 10.3390/biom15030441] [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: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease that results in the selective loss of pancreatic beta cells and an eventual deficit in insulin production to maintain glucose homeostasis. It is now increasingly accepted that this dynamic disease process is multifactorial; involves a variety of immune cells which contribute to an inflamed pancreatic microenvironment; and that the condition is heterogenous, resulting in variable rates of subsequent beta cell damage. In this review, we will explore the current understanding of the cellular interactions between both resident and infiltrating immune cells within the pancreatic environment, highlighting key mechanisms which may promote the beta cell destruction and islet damage associated with T1D.
Collapse
Affiliation(s)
| | | | - Joanne Boldison
- Department of Clinical and Biomedical Sciences, University of Exeter, RILD Building (Level 4), Barrack Road, Exeter EX2 5DW, UK; (S.L.W.); (P.L.)
| |
Collapse
|
39
|
Liu J, Li X, Wang Y, Xu Z, Lv Y, He Y, Chen L, Feng Y, Liu G, Bai Y, Xie W, Wu Q. Predicting postoperative pulmonary infection in elderly patients undergoing major surgery: a study based on logistic regression and machine learning models. BMC Pulm Med 2025; 25:128. [PMID: 40108569 PMCID: PMC11921591 DOI: 10.1186/s12890-025-03582-4] [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: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Postoperative pulmonary infection (POI) is strongly associated with a poor prognosis and has a high incidence in elderly patients undergoing major surgery. Machine learning (ML) algorithms are increasingly being used in medicine, but the predictive role of logistic regression (LR) and ML algorithms for POI in high-risk populations remains unclear. METHODS We conducted a retrospective cohort study of older adults undergoing major surgery over a period of six years. The included patients were randomly divided into training and validation sets at a ratio of 7:3. The features selected by the least absolute shrinkage and selection operator regression algorithm were used as the input variables of the ML and LR models. The random forest of multiple interpretable methods was used to interpret the ML models. RESULTS Of the 9481 older adults in our study, 951 developed POI. Among the different algorithms, LR performed the best with an AUC of 0.80, whereas the decision tree performed the worst with an AUC of 0.75. Furthermore, the LR model outperformed the other ML models in terms of accuracy (88.22%), specificity (90.29%), precision (44.42%), and F1 score (54.25%). Despite employing four interpretable methods for RF analysis, there existed a certain degree of inconsistency in the results. Finally, to facilitate clinical application, we established a web-friendly version of the nomogram based on the LR algorithm; In addition, patients were divided into three significantly distinct risk intervals in predicting POI. CONCLUSIONS Compared with popular ML algorithms, LR was more effective at predicting POI in older patients undergoing major surgery. The constructed nomogram could identify high-risk elderly patients and facilitate perioperative management planning. TRIAL REGISTRATION The study was retrospectively registered (NCT06491459).
Collapse
Affiliation(s)
- Jie Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yanting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhenzhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Lv
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yuyao He
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yiqi Feng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guoyang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yunxiao Bai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wanli Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
40
|
Stephan JK, Knerr T, Gu Z, Li H, Brittian KR, Dassanayaka S, Singhal R, Nong Y, Jones SP, Wysoczynski M. Neutrophil-secreted CHI3L1 exacerbates cardiac dysfunction and inflammation after myocardial infarction. FASEB J 2025; 39:e70422. [PMID: 40013912 PMCID: PMC11963974 DOI: 10.1096/fj.202401654r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Myocardial infarction (MI) triggers acute inflammation, marked by neutrophil infiltration. Although neutrophils are central to this response, the exact role of various neutrophil-derived factors is not fully understood. Clinical studies have linked one such enigmatic factor, chitinase-3 like-1, to MI outcomes. Hence, we investigated its role in post-MI remodeling. We found that chitinase-3 like-1 (CHI3L1) is upregulated after MI and secreted by activated neutrophils but does not directly affect neutrophil activity. To assess whether increased CHI3L1 influences ventricular remodeling, we subjected mice to non-reperfused MI and administered recombinant CHI3L1. Increased CHI3L1 levels worsened ventricular remodeling. In contrast, CHI3L1-deficient mice showed reduced ventricular remodeling after MI. To explore the underlying mechanisms, we assessed interactions with other cells known to be important in ventricular remodeling. Immunoprofiling of infarcted CHI3L1-deficient mouse hearts revealed a faster decline in neutrophil and monocyte numbers, indicating quicker resolution of inflammation. These findings provide direct evidence that CHI3L1 exacerbates ventricular inflammation and remodeling following MI through gain- and loss-of-function approaches.
Collapse
Affiliation(s)
- Jonah K. Stephan
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Taylor Knerr
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Zhen Gu
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Hong Li
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Kenneth R. Brittian
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Sujith Dassanayaka
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Richa Singhal
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Yibing Nong
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Steven P. Jones
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| | - Marcin Wysoczynski
- Center for Cardiometabolic Science, Christina Lee Brown Envirome InstituteUniversity of Louisville School of MedicineLouisvilleKentuckyUSA
| |
Collapse
|
41
|
Ye T, Wu Z, Liu X, Wu J, Fu Q, Cao J, Zhang D, Shi P. Engineered mesenchymal stromal cells with bispecific polyvalent peptides suppress excessive neutrophil infiltration and boost therapy. SCIENCE ADVANCES 2025; 11:eadt7387. [PMID: 40053594 PMCID: PMC11887798 DOI: 10.1126/sciadv.adt7387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Excessive neutrophil infiltration can exacerbate inflammation and tissue damage, contributing to conditions like autoimmune disorders and liver diseases. Mesenchymal stromal cells (MSCs) share homing mechanisms with neutrophils, showing promise for treating such diseases. However, ex vivo expanded MSCs often suffer from reduced homing efficiency due to the loss of essential ligands. Here, we engineer MSCs with P-selectin and E-selectin targeting peptides, assembling them into bispecific polyvalent structures using DNA self-assembly technology. This modification allows engineered MSCs to compete with chemotactic neutrophils for selectin binding sites on endothelial cells. In a mouse model of acute liver failure, engineered MSCs effectively home to the damaged liver and substantially inhibit excessive neutrophil infiltration. The combination of inhibiting neutrophil infiltration and the MSCs' inherent therapeutic properties lead to superior therapeutic outcomes. Single-cell RNA sequencing reveals that engineered MSCs elevate the levels of Marco_macrophage, which have neutrophil-inhibitory effects. Our study offers a perspective for advancing MSC-based therapies in tissue repair.
Collapse
Affiliation(s)
- Tenghui Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Zixin Wu
- Department of General Surgery, Guangzhou Digestive Disease Center, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Xi Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Jiamin Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Qin Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Di Zhang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, PR China
| |
Collapse
|
42
|
Jia R, Yin Y, Shan H. Systemic inflammatory response index as a novel biomarker for age-related macular degeneration: a cross-sectional study from NHANES (2005-2008). Front Nutr 2025; 12:1540933. [PMID: 40115389 PMCID: PMC11922706 DOI: 10.3389/fnut.2025.1540933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Background Chronic low-grade systemic inflammation plays a significant role in age-related macular degeneration (AMD) pathogenesis. The systemic inflammatory response index (SIRI), a novel inflammatory marker, may predict various diseases. However, data on the relationship between SIRI and AMD are limited. This study examines the relationship between SIRI and AMD and assesses its potential as a predictive biomarker. Methods A cross-sectional analysis of the National Health and Nutrition Examination Survey (NHANES) data from 2005 to 2008 was conducted on participants aged ≥40 years with SIRI and AMD status data. Multivariable logistic regression models adjusted for confounders were used to assess the association. Sensitivity and subgroup analyses, along with restricted cubic spline (RCS) curve analysis, were performed. Results Among 5,365 participants, 425 (7.9%) had AMD. The median SIRI was higher in AMD patients (1.23 vs. 1.04, p < 0.001). Higher SIRI was independently associated with increased odds (adjusted OR: 1.18, 95% CI:1.07-1.29, p = 0.001). RCS analyses revealed a dose-response relationship (p = 0.002). Subgroup analyses showed a positive association in male participants, individuals with hypertension, individuals with obesity, and non-smokers. Higher SIRI levels were independently associated with increased AMD risk (adjusted OR: 1.27, 95% CI: 1.03-1.56, p = 0.023). Conclusion Elevated SIRI is independently associated with increased AMD risk in the U.S. population. SIRI may serve as a biomarker for identifying high-risk individuals, enabling early intervention. The cross-sectional design limits causal inference, and unmeasured confounders may affect the results. SIRI could potentially serve as a non-invasive biomarker for AMD risk, pending further validation through longitudinal studies.
Collapse
Affiliation(s)
- Ruoshuang Jia
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huimin Shan
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Newman PP, Schmitt BL, Maurmann RM, Pence BD. Polysaccharides with Arabinose: Key Players in Reducing Chronic Inflammation and Enhancing Immune Health in Aging. Molecules 2025; 30:1178. [PMID: 40076400 PMCID: PMC11901799 DOI: 10.3390/molecules30051178] [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: 12/12/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Aging is associated with a decline in physiological performance leading to increased inflammation and impaired immune function. Polysaccharides (PLs) found in plants, fruits, and fungi are emerging as potential targets for therapeutic intervention, but little is known about their effects on chronic inflammation and aging. This review aims to highlight the current advances related to the use of PLs, with the presence of arabinose, to attenuate oxidative stress and chronic and acute inflammation, and their immunomodulatory effects associated with antioxidant status in monocytes, macrophages, and neutrophil infiltration, and leukocyte rolling adhesion in neutrophils. In addition, recent studies have shown the importance of investigating the 'major' monosaccharide, such as arabinose, present in several of these polysaccharides, and with described effects on gut microbiome, glucose, inflammation, allergy, cancer cell proliferation, neuromodulation, and metabolic stress. Perspectives and opportunities for further investigation are provided. By promoting a balanced immune response and reducing inflammation, PLs with arabinose or even arabinose per se may alleviate the immune dysregulation and inflammation seen in the elderly, therefore providing a promising strategy to mitigate a variety of diseases.
Collapse
Affiliation(s)
- Patricia Pantoja Newman
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (B.L.S.); (R.M.M.)
| | | | | | - Brandt D. Pence
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (B.L.S.); (R.M.M.)
| |
Collapse
|
44
|
Bani Hani DA, Alshraideh JA, Saleh A, Alduraidi H, Alwahadneh AA, Al-Zaiti SS. Lymphocyte-based inflammatory markers: Novel predictors of significant coronary artery disease ✰,✰✰. Heart Lung 2025; 70:23-29. [PMID: 39549307 DOI: 10.1016/j.hrtlng.2024.11.006] [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: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Lymphocyte-based inflammatory indices such as monocyte-to-lymphocyte ratio (MLR) have long been recognized as reliable coronary artery disease (CAD) predictors. More recently, novel indices like the Systemic Inflammatory Index (SII), Systemic Inflammatory Response Index (SIRI), and Systemic Immune-Inflammation Index (SIIRI) have emerged. These newer markers offer a more comprehensive assessment of inflammation by integrating multiple immune cell types, potentially enhancing the prediction of cardiovascular outcomes. OBJECTIVES We evaluated the predictive value of novel inflammatory markers in estimating the pretest probability of severe CAD in high-risk patients. METHODS We enrolled consecutive patients undergoing diagnostic coronary angiography in a single tertiary care hospital. Inflammatory markers were calculated based on pre-procedural complete blood count laboratory measurements. Severe CAD was defined as critical (>70 %) and actionable narrowing of a primary coronary artery. Classification performance was assessed using multivariate logistic regression. RESULTS The study sample included 363 patients (age 58.9± 11 years, 44.9 % females, 30 % severe CAD). In univariate analysis, MLR, SIRI, and SIIRI were significant predictors of severe CAD, with age- and sex-adjusted OR of 1.98 [1.25-3.14], 1.79 [1.24-2.59], and 1.63 [1.11-2.38], respectively. In multivariate analysis, SIRI remained an independent predictor of severe CAD (OR = 1.98, 95 % CI 1.13-3.46, p = 0.02). CONCLUSION Our results suggest that novel inflammatory markers derived from routine blood tests are predictive of severe CAD in high-risk patients. Such simple, practical, and cost-effective inflammatory markers may enhance cardiac risk stratification and prediction of severe CAD.
Collapse
Affiliation(s)
| | | | - Akram Saleh
- The University of Jordan, Jordan University Hospital, Amman, Jordan.
| | - Hamza Alduraidi
- Community Health Nursing Department, School of Nursing, The University of Jordan, Amman, Jordan.
| | | | - Salah S Al-Zaiti
- School of Nursing, the University of Jordan, Amman, Jordan; University of Rochester, Rochester, NY, USA.
| |
Collapse
|
45
|
Zahoor I, Bala R, Wani SN, Chauhan S, Madaan R, Kumar R, Hakeem KR, Malik IA. Potential role of NSAIDs loaded nano-formulations to treat inflammatory diseases. Inflammopharmacology 2025; 33:1189-1207. [PMID: 39953360 DOI: 10.1007/s10787-025-01644-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/25/2024] [Indexed: 02/17/2025]
Abstract
Inflammation is a necessary immunological response that promotes survival and preserves tissue homeostasis, a common characteristic linked to various diseases. However, in some circumstances, the inflammatory response is deleterious and contributes to disease pathogenesis. Anti-inflammatory substances have poor affinity for inflamed tissues, resulting in low concentrations in the target tissue and a higher incidence of severe adverse effects. To address this issue, several potential approaches have been proposed, such as chemical modification of drug molecules and the development of nanocarriers for drug delivery. Since the development of nanotechnology at the beginning of the twenty-first century, researchers have been using the pathophysiological characteristics of inflammation, primarily leaky vasculature, and biomarker overexpression to develop nanomedicines that can deliver therapeutics via passive and active targeting mechanisms to sites of inflammation and produce therapeutic effects. Drug carriers based on nanoparticles can enhance the safety and efficacy of drugs by increasing their capacity, enhancing their solubility, combining several drugs, protecting them from metabolism, and regulating their release. An approach that shows promise in the treatment of various inflammatory diseases is the application of nanomedicines. Nanomedicine involves nanoparticles that have been loaded with a therapeutically active component. Nanomedicines can target inflammation by recognizing molecules highly expressed on endothelial cells or activated macrophage surfaces, enhancing the permeability of vessels, or even by biomimicry. A review of the research findings shows significant potential for the use of nanotechnology to enhance the quality of life for people using NSAIDs for chronic disorders by minimizing drug side effects or the duration of administration. After a brief introduction to inflammation, its various forms- acute and chronic inflammation, and the pathophysiology of inflammation, this review highlights the main innovative nanocarriers utilized for carrying various nonsteroidal anti-inflammatory drugs that have been utilized in treating various inflammatory disorders.
Collapse
Affiliation(s)
- Ishrat Zahoor
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India.
| | - Rajni Bala
- University School of Pharmaceutical Sciences, Rayat-Bhara University, Kharar, Punjab, India
| | - Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Aman Pharmacy College, Dholakhera Udaipurwati, Jhunjhunu, Rajasthan, India
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Reecha Madaan
- Adesh College of Pharmacy, NH1 Shahabad Kurukshetra, Haryana, India
| | - Rajesh Kumar
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Khalid Rehman Hakeem
- Department of Biological Sciences, Faculty of Science, King Adualaziz University, 21589, Jeddah, Saudi Arabia
- Department of Public Health, Daffodil International University, Dhaka, 1341, Bangladesh
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Irfan Ahmad Malik
- Department of Pharmacology, Sanjivani College of Pharmaceutical Education and Research, Kopargaon, 423603, Maharashtra, India
| |
Collapse
|
46
|
Zoroddu S, Di Lorenzo B, Paliogiannis P, Mangoni AA, Carru C, Zinellu A. Vascular endothelial growth factor in inflammatory bowel disease: A systematic review and meta-analysis. Eur J Clin Invest 2025; 55:e14361. [PMID: 39545600 PMCID: PMC11810564 DOI: 10.1111/eci.14361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
AIM Vascular endothelial growth factor (VEGF) is linked to inflammation and angiogenesis, indicating a possible role in inflammatory bowel disease (IBD) and its main clinical manifestations, Crohn's disease (CD) and ulcerative colitis (UC). This systematic review and meta-analysis investigated studies assessing circulating VEGF concentrations in IBD patients and healthy controls, considering the effect of IBD type, sample type and geographical location. METHODS A systematic search identified 18 studies (28 group comparators) investigating 1741 IBD patients and 1291 controls. Data were extracted and analysed using standardized mean differences (SMD) with 95% confidence intervals (CI). RESULTS VEGF concentrations were significantly higher in IBD patients (SMD = .71, 95% CI .38 to 1.04; p < .001). UC patients showed higher VEGF concentrations than CD patients. Serum samples indicated significant VEGF elevations, unlike plasma samples. Significant VEGF increases were observed in studies conducted in Western Europe and Asia, but not in Eastern Europe. No significant differences were found between active and inactive disease. CONCLUSIONS VEGF concentrations are elevated in IBD patients, with variations by disease type, sample type and geography. However, VEGF is not a reliable marker of disease activity. Future research should standardize methods and explore regional influences to enhance VEGF's clinical utility as a biomarker of IBD.
Collapse
Affiliation(s)
- Stefano Zoroddu
- Department of Biomedical SciencesUniversity of SassariSassariItaly
| | - Biagio Di Lorenzo
- Department of Biomedical SciencesUniversity of SassariSassariItaly
- Department of Medicine and SurgeryLUM UniversityCasamassimaItaly
| | - Panagiotis Paliogiannis
- Department of Medicine, Surgery and PharmacyUniversity of SassariSassariItaly
- Anatomic Pathology and Histology UnitUniversity Hospital (AOU) of SassariSassariItaly
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- Department of Clinical Pharmacology, Flinders Medical CentreSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Ciriaco Carru
- Department of Biomedical SciencesUniversity of SassariSassariItaly
- Medical Oncology UnitUniversity Hospital (AOU) of SassariSassariItaly
| | - Angelo Zinellu
- Department of Biomedical SciencesUniversity of SassariSassariItaly
| |
Collapse
|
47
|
Zheng Y, Zhong D, Li J, Zhang Y, Li H, Liu L, Ren C, Zhong S, Liu X, He X, Jin S, Luo L. Systemic immune-inflammation index and long-term mortality in patients with hypertension: a cohort study. J Hypertens 2025; 43:464-473. [PMID: 39670474 PMCID: PMC11789614 DOI: 10.1097/hjh.0000000000003927] [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: 06/24/2024] [Revised: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVE The objective of this study was to examine the relationship between systemic inflammation and long-term mortality in patients with hypertension. METHODS The study employed a retrospective cohort design. The study population was derived from the National Health and Nutrition Examination Survey (NHANES), and the mortality data for this population was acquired from the National Death Index (NDI) database. Systemic inflammation was quantified by the Systemic Immune Inflammation Index (SII) and the Systemic Inflammatory Response Index (SIRI), which were then categorized into four groups (Q1-Q4, with Q4 representing the highest level of SII or SIRI). Weighted Cox regression models were constructed to investigate the association between mortality and SII and SIRI, with hazard ratios (HRs) subsequently calculated. RESULTS A total of 7431 participants were included in the analysis. The highest quantile (Q4) of SII was associated with a higher risk of all-cause mortality (hazard ratio 1.36, 95% CI 1.1-1.69, P < 0.001). After adjustment for important covariates, the association remained significant (hazard ratio 1.70, 95% CI 1.27-2.30, P < 0.001). The highest quantile (Q4) of SIRI was also associated with the highest risk of mortality (hazard ratio 2.11, 95% CI 1.64-2.70, P < 0.001), and this association remained significant after adjustment for important covariates (hazard ratio 1.64, 95% CI 0.61-1.22, P = 0.001). CONCLUSION Both SII and SIRI scores were found to be associated with mortality rates in patients with hypertension. The findings suggest that these scores may serve as complementary biomarkers to the neutrophil-to-lymphocyte ratio (NLR) for assessing mortality risk in patients with hypertension. Further investigation is warranted to elucidate the underlying mechanisms that underpin this association.
Collapse
Affiliation(s)
- Yaling Zheng
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Dongling Zhong
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine
| | - Juan Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine
- Affiliated Sichuan Provincial Rehabilitation Hospital of Chengdu University of TCM
| | - Yue Zhang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine
| | - Huijing Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luoji Liu
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Chi Ren
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Shan Zhong
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Xicen Liu
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Xia He
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Shiqi Jin
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| | - Lun Luo
- Department of Rehabilitative Medicine, Chengdu Second People's Hospital
| |
Collapse
|
48
|
Liu Y, Liu W, Yang Y, Liu H, Liu J, Liu Y. The association between dietary dark green vegetable intake and cognitive function in US older adults. NUTR BULL 2025; 50:69-81. [PMID: 39572249 DOI: 10.1111/nbu.12720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 09/26/2024] [Accepted: 10/30/2024] [Indexed: 02/13/2025]
Abstract
Dark green vegetables include dark green leafy vegetables and broccoli. They are sources of many essential nutrients, including vitamins A, B and C, folate, fibre, carotenoids and flavonoids. This study aimed to explore the association between dietary dark green vegetable intake and cognitive function in US older adults. We included 2344 older adults (≥60 years old) from the National Health and Nutrition Examination Survey (NHANES) 2011-2014 cycles. Dark green vegetable consumption was assessed using a continuous variable (cups/day) and two categorical variables. The first categorical variable classified participants into non-consumers and consumers based on whether they consumed dark green vegetables. The second categorical variable grouped participants into four levels of dark green vegetable consumption (non-consumers, consumers with low intakes, consumers with moderate intakes and consumers with high intakes). We used five continuous variables with non-normal distribution to assess cognitive function, including a composite z-score and the standardised scores of four individual cognitive tests. The four cognitive tests included the Immediate Recall Test (IRT), the Delayed Recall Test (DRT), the Animal Mobility Test (AFT) and the Digit Symbol Substitution Test (DSST). The standardised scores of the four cognitive tests were calculated using the mean and standard deviation of each cognitive test score. The composite z-score was calculated by averaging the standardised scores of four cognitive tests to evaluate global cognition. We used multiple linear regression models to examine the association between dietary dark green vegetable intake and cognitive function. Our findings indicated that dark green vegetable intake was positively associated with global cognition (β [95% CI]: 0.17 [0.04, 0.30]; p = 0.016) and IRT (β [95% CI]: 0.26 [0.08, 0.43]; p = 0.009) and DRT (β [95% CI]: 0.21 [0.05, 0.36]; p = 0.012) standardised scores. Individuals with high intake of dark green vegetables showed notably better global cognition (β [95% CI]: 0.16 [0.05, 0.28]; p = 0.010) and showed higher IRT (β [95% CI]: 0.22 [0.07, 0.38]; p = 0.010) and DRT standardised scores (β [95% CI]: 0.21 [0.07, 0.36]; p = 0.007) compared with the non-consumers. Blood neutrophil counts mediated the cognitive benefits of dark green vegetables (Proportion: 9.5%, p = 0.006). In conclusion, our findings suggest that dark green vegetable consumption may have favourable effects on cognitive function in US older adults, especially on immediate and delayed learning abilities. The underlying mechanisms include the ability of dark green vegetables to reduce blood neutrophil levels, an indicator of decreased systemic inflammation. Increasing dietary intake of dark green vegetables may be a beneficial intervention to improve cognitive health in the older US population.
Collapse
Affiliation(s)
- Yuqian Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Wen Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Yang Yang
- Qilu Hospital of Shandong University, Jinan, China
| | - Heyin Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Jinde Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Yiming Liu
- Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
49
|
You YL, Byun HJ, Chang YB, Kim H, Lee H, Suh HJ, Jeon JY, Kim BR, Hwang JE, Lee JH, Choi HS. Euglena gracilis-derived β-glucan ameliorates particulate matter (PM 2.5)-induced airway inflammation by modulating nuclear factor kappa B, mitogen-activated protein kinase, and nuclear factor erythroid 2-related factor 2 signaling pathways in A549 cells and BALB/c mice. Int J Biol Macromol 2025; 296:139671. [PMID: 39798741 DOI: 10.1016/j.ijbiomac.2025.139671] [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: 09/29/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
This study aimed to investigate the effects of β-glucan derived from Euglena gracilis (EGB), an edible microalga, on particulate matter (PM2.5)-induced airway inflammation in A549 cells and BALB/c mice. EGB effectively suppressed the mRNA and protein levels of inflammatory cytokines (IL-6, IL-1β, TNF-α, IL-8) and mediators (iNOS, COX-2), while inhibiting the NF-κB and MAPK signaling pathways triggered by PM2.5 exposure and reducing nuclear NF-κB levels. Additionally, EGB decreased PM2.5-induced ROS production and increased the protein levels of NRF2 and HO-1, along with genes encoding antioxidant enzymes (catalase, GPx, SOD1), associated with elevated nuclear NRF2 levels. EGB reduced immune cell infiltration and inflammatory cytokine levels in BALF and serum, both of which increased by PM2.5 exposure. EGB also significantly increased alveolar numbers while decreasing the gene expression of MMP1/9/13. Furthermore, EGB suppressed PM2.5-induced bronchial thickening and collagen-1 deposition by downregulating TGF-β1 expression, and alleviated goblet cell hyperplasia and mucin production in lung tissues. These results suggest that EGB effectively reduces PM2.5-induced airway inflammation by suppressing NF-κB and MAPK signaling pathways, lowering pro-inflammatory cytokines, and activating the NRF2-HO-1 signaling pathway to enhance antioxidant enzyme expression. This study highlights the potential of EGB as an edible functional agent for controlling PM-related airway inflammation.
Collapse
Affiliation(s)
- Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea
| | - Ha-Jun Byun
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea
| | - Yeok Boo Chang
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyeongyeong Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyowon Lee
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jin-Young Jeon
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Bo-Ra Kim
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Ji Eun Hwang
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Jun Hee Lee
- Health R&D Institute, Daesang Corp., Seoul 07789, Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea.
| |
Collapse
|
50
|
Wang X, He S, Gong X, Lei S, Zhang Q, Xiong J, Liu Y. Neutrophils in colorectal cancer: mechanisms, prognostic value, and therapeutic implications. Front Immunol 2025; 16:1538635. [PMID: 40092983 PMCID: PMC11906667 DOI: 10.3389/fimmu.2025.1538635] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Neutrophils, the most abundant myeloid cells in human peripheral blood, serve as the first defense line against infection and are also significantly involved in the initiation and progression of cancer. In colorectal cancer (CRC), neutrophils exhibit a dual function by promoting tumor events and exerting antitumor activity, which is related to the heterogeneity of neutrophils. The neutrophil extracellular traps (NETs), gut microbiota, and various cells within the tumor microenvironment (TME) are involved in shaping the heterogeneous function of neutrophils. This article provides an updated overview of the complex functions and underlying mechanisms of neutrophils in CRC and their pivotal role in guiding prognosis assessment and therapeutic strategies, aiming to offer novel insights into neutrophil-associated treatment approaches for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yang Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|