1
|
Li C, Yu J, Issa R, Wang L, Ning M, Yin S, Li J, Wu C, Chen Y. CoronaVac-induced antibodies that facilitate Fc-mediated neutrophil phagocytosis track with COVID-19 disease resolution. Emerg Microbes Infect 2025; 14:2434567. [PMID: 39584817 PMCID: PMC11731273 DOI: 10.1080/22221751.2024.2434567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants raise concerns about decreased vaccine efficacy, vaccines continue to confer robust protection in humans, implying that immunity beyond neutralization contributes to vaccine efficacy. In addition to neutralization, antibodies can mediate various Fc-dependent effector functions, including antibody-dependent cellular phagocytosis (ADCP), antibody-dependent neutrophil phagocytosis (ADNP) and antibody-dependent cellular cytotoxicity (ADCC). However, the specific role of each Fc-mediated effector function in contributing to COVID-19 disease attenuation in human remains unclear. To fully define the potential immune correlates of Fc-mediated effector functions, we comprehensively analysed the above Fc-mediated effector functions in two study cohorts. In the CoronaVac vaccinee cohort, individuals without breakthrough infection exhibited higher levels of ADCP and ADNP activities with a greater degree of cross-reactivity compared to those who had breakthrough infection. A predictive model was established incorporating ADNP activity and IgG titre, achieving an area under the curve (AUC) of 0.837. In the COVID-19 patient cohort, BA.5-specific ADCP and ADNP responses were significantly reduced in COVID-19 patients with fatal outcomes compared to milder outcomes. The prognostic model incorporating WT, BA.5, and XBB.1.5 spike-specific ADNP demonstrated effective predictive ability, achieving an AUC of 0.890. Meanwhile, transcriptomic analysis of peripheral blood mononuclear cells (PBMCs) from COVID-19 patients in the acute phases of infection highlighted remarkably upregulation of neutrophil activity and phagocytic function, further reinforcing the essential role of ADNP. Collectively, our findings underscored Fc-mediated effector activities, especially neutrophil phagocytosis, as significant antibody biomarkers for the risk of SARS-CoV-2 breakthrough infection and COVID-19 prognosis.
Collapse
Affiliation(s)
- Chuang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, People’s Republic of China
| | - Jie Yu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Rahma Issa
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Department of Pharmacy, Ismailia Teaching Oncology Hospital (GOTHI), Ismailia, Egypt
| | - Lili Wang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, People’s Republic of China
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine. Zhong Fu Road, Nanjing, People’s Republic of China
| | - Mingzhe Ning
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Shengxia Yin
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, People’s Republic of China
| | - Chao Wu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, People’s Republic of China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, People’s Republic of China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Wang L, Yang L, Tian L, Guo B, Dai T, Lv Q, Xie J, Liu F, Bao H, Cao F, Liu Y, Gao Y, Hou Y, Ye Z, Wang S, Zhang Q, Kong L, Cai B. Exosome-capturing scaffold promotes endogenous bone regeneration through neutrophil-derived exosomes by enhancing fast vascularization. Biomaterials 2025; 319:123215. [PMID: 40023128 DOI: 10.1016/j.biomaterials.2025.123215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 01/19/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Exosomes (Exos), extracellular vesicles of endosomal origin, are a promising therapeutic platform for tissue regeneration. In the current study, an exosome-capturing scaffold (ECS) was designed to attract and anchor exosomes via electrostatic adherence followed by lipophilic interactions. Our findings demonstrate that local enrichment of exosomes in the ECS implanted into critical mandibular defects could significantly accelerate endogenous bone regeneration by enhancing vascularization at the defect site. Notably, neutrophil (PMN)-derived exosomes (PMN-Exos) were identified as the predominant exosome subtype among all captured exosomes. During endogenous bone regeneration, PMN-Exos promoted endogenous vascularization primarily by stimulating the proliferation of endothelial progenitor cells (EPCs), which play a pivotal role in the vasculogenesis of new blood vessels. Mechanistically, vascularization involved PMN-Exo-derived miR455-3p, which promotes EPC proliferation by targeting the Smad4 pathway. In conclusion, this study offers an ECS with broad application prospects for enhancing tissue regeneration by accelerating vascularization. The elucidation of underlying mechanisms paves the way for developing novel strategies to regenerate various tissues and organs.
Collapse
Affiliation(s)
- Le Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Luying Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Lei Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Baolin Guo
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Taiqiang Dai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Qianxin Lv
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Jirong Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Fuwei Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Han Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Feng Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Ya Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China; College of Life Sciences, Northwest University, Xi'an, 710069, China.
| | - Ye Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yan Hou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R, China.
| | - Shenqiang Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Qiuyu Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of the Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, China.
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Bolei Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Wang Y, Yin J, Yu C, Wu D, Chen Y, Han Q, Li S, Zhang R, Wang W, Xu J. Neutrophil extracellular traps activate STING signaling to promote dendritic cell-driven rejection after liver transplantation. Int Immunopharmacol 2025; 160:114763. [PMID: 40449271 DOI: 10.1016/j.intimp.2025.114763] [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: 02/08/2025] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 06/03/2025]
Abstract
PURPOSE Post-transplant immune rejection affects graft function. Interaction between neutrophil extracellular traps (NETs) with specific immune cells and the specific mechanism in liver transplantation were still unclear. METHOD Clinical patients RNA-Seq results were used for GSEA and KEGG analysis. C57BL/6 and C3H mouse models and clinical samples were use to describe the disease phenotype characteristics through multiple immunofluorescence, flow cytometry and etc. Cell co-culture experiments were performed to clarify the mechanism pathway process. RESULTS RNA-Seq results analysis indicated that the NETs formation pathway was upregulated. Animal models confirmed that in liver transplant immune rejection status the formation of NETs in situ and peripheral cells increased and the level of cell-free DNA (cf-DNA) in peripheral cells increased. Reactive oxygen species (ROS) as a predisposing factor for NETs accumulated more in immune rejection status and NETs are rich in mitochondrial DNA (mtDNA). NETs promote dendritic cell maturation through STING-related pathways. NETs formation increases in patients with liver transplant immune rejection and is positively correlated with disease severity. CONCLUSION We found that NETs can regulate dendritic cell maturation through STING-related pathways after liver transplantation, which may ultimately promote the occurrence of liver transplant rejection, providing a new perspective for clinical diagnosis, treatment and prevention of liver transplant rejection.
Collapse
Affiliation(s)
- Yan Wang
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jie Yin
- Basic Medicine School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chenjiang Yu
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Dongdong Wu
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yizhang Chen
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qi Han
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Shipeng Li
- Department of Hepatopancreaticobiliary Surgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Rui Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, National Health Commission Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
4
|
Li F, Fan X, Wang B, Tong J, Ling M, Tong H, Huang Y. Phillyrin counters β2 integrin-mediated neutrophil adhesion and chemotaxis to alleviate endotoxin-induced acute lung injury in neonatal rats. Biochem Pharmacol 2025; 237:116934. [PMID: 40210127 DOI: 10.1016/j.bcp.2025.116934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/15/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Acute lung injury (ALI) in neonates presents a grave threat to infant health, characterized by a heightened risk of mortality. Phillyrin, an extraordinary constituent derived from a traditional Chinese medicinal herb Forsythia suspensa, has garnered considerable attention for its pronounced anti-inflammatory properties. However, its therapeutic potential for acute inflammatory diseases in neonates remains unclear. Therefore, our current study endeavors to assess the protective effects of phillyrin against lipopolysaccharide (LPS)-induced ALI in neonates and elucidate the underlying mechanisms. Phillyrin exhibited significant amelioration of lung damage in neonatal rats with LPS-induced ALI, accompanied by reductions in the total cell counts, neutrophil counts, and total protein level in bronchoalveolar lavage fluid (BALF). Notably, phillyrin substantially attenuated proinflammatory cytokine secretion and suppressed NF-κB activation in the lungs of neonatal ALI rats; however, it demonstrated inefficacy in mitigating LPS-induced cytokine secretion and NF-κB activation in vitro. Notably, phillyrin effectively inhibited β2 integrin-mediated neutrophil adhesion, migration, and chemotaxis. Moreover, phillyrin robustly suppressed β2 integrin engagement-induced actin polymerization and the Vav1/Rac1/PAK1/LIMK1/cofilin pathway. From a mechanistic standpoint, phillyrin exhibited direct interaction with β2 integrin, effectively antagonizing its function and significantly disrupting its binding affinity to intercellular adhesion molecule 1 (ICAM-1). This investigation unveils the promising therapeutic prospects of phillyrin as a novel compound against neonatal ALI.
Collapse
Affiliation(s)
- Feng Li
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Xinrong Fan
- Department of Durg Preparation, Lishui Hospital of Traditional Chinese Medicine, Lishui 323000, China
| | - Bohao Wang
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Jingyang Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Menglai Ling
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China.
| | - Yumei Huang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
5
|
Lu L, Lin J, Wei F, Huang W, Sang Y, Zhou Y, Yu C, Yuan W, Feng Y, Kong X. Yin-chen Wu-ling powder inhibits MAPKs/CXCL1/CXCR2-induced neutrophil infiltration to alleviate LPS/D-GalN-induced acute liver failure. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119957. [PMID: 40368257 DOI: 10.1016/j.jep.2025.119957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute liver failure (ALF) is the result of progression from acute liver injury with high mortality, and novel treatments are needed. Yin-chen Wu-ling powder (YWP), a traditional herbal medicine in China, has been used for treating acute liver injury for thousands of years. However, the mechanism of YWP is unknown. AIM OF THE STUDY In vitro and in vivo studies were conducted to clarify YWP's protective effect on ALF and investigate its hepatoprotective mechanism. MATERIALS AND METHODS We established an LPS/D-GalN-induced ALF mouse model and in vitro system to evaluate the effect of YWP. We characterized YWP's chemical composition via UHPLC-Q-Exactive Orbitrap HRMS. Enzyme-linked immunosorbent assay, hematoxylin and eosin staining, immunohistochemistry and immunofluorescence, flow cytometry, qPCR, Western blot were used to discover key mechanisms both in vitro and in vivo. RESULTS YWP alleviated liver dysfunction and liver necrosis. YWP reduced hepatocyte death and inflammatory responses. Importantly, YWP markedly inhibited neutrophil infiltration into the liver. We examined key chemokines that contribute to neutrophil recruitment. The results showed that YWP inhibited CXCL1, which is sourced from inflammation-activated hepatocytes. In addition, YWP inhibited TNF-α-induced CXCL1 transcription via the inhibition of MAPKs signaling in vitro. Furthermore, the anti-ALF effect of YWP was weakened when CXCL1/CXCR2 signaling was suppressed. CONCLUSION YWP alleviates inflammatory liver injury in ALF by suppressing neutrophil infiltration into the liver, potentially through inhibition of the MAPKs/CXCL1/CXCR2 axis. We suggest that YWP is a potential anti-inflammatory treatment for ALF.
Collapse
Affiliation(s)
- Liyue Lu
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Wei
- Department of Emergency Surgeon Medicine, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weifan Huang
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yali Sang
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuge Zhou
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang Yu
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weian Yuan
- Department of Liver Diseases, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yu Feng
- Pudong New Area Hospital of Traditional Chinese Medicine Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
6
|
Xiong J, Liu L, Zhang X. Biological and Molecular Mechanisms of Wound Healing: Emerging Therapeutic Strategies and Future Directions. Am Surg 2025:31348251350983. [PMID: 40492867 DOI: 10.1177/00031348251350983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
Abstract
This review comprehensively examines the biological and molecular mechanisms underlying wound healing, focusing on inflammation, signaling pathways, and microenvironmental imbalances. By comparing traditional and emerging therapies, we highlight innovative approaches such as gene editing, stem cell therapy, and nanotechnology, which offer new perspectives for enhancing wound treatment outcomes. This study underscores the integration of modern medicine with traditional practices, providing a robust theoretical foundation for future clinical applications.
Collapse
Affiliation(s)
- Jun Xiong
- Department of Day Surgery Ward, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, China
| | - Li Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, China
| | - Xuebing Zhang
- Department of Day Surgery Ward, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, China
| |
Collapse
|
7
|
Loggini A, Hornik J, Henson J, Wesler J, Hornik A. Association between neutrophil-to-lymphocyte ratio and hematoma expansion in spontaneous intracerebral hemorrhage: A systematic review and meta-analysis. World J Crit Care Med 2025; 14:99445. [PMID: 40491877 PMCID: PMC11891842 DOI: 10.5492/wjccm.v14.i2.99445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Hematoma expansion (HE) typically portends a poor prognosis in spontaneous intracerebral hemorrhage (ICH). Several radiographic and laboratory values have been proposed as predictive markers of HE. AIM To perform a systematic review and meta-analysis on the association of neutrophil-to-lymphocyte ratio (NLR) and HE in ICH. A secondary outcome examined was the association of NLR and perihematomal (PHE) growth. METHODS Three databases were searched (PubMed, EMBASE, and Cochrane) for studies evaluating the effect of NLR on HE and PHE growth. The inverse variance method was applied to estimate an overall effect for each specific outcome by combining weighted averages of the individual studies' estimates of the logarithm odds ratio (OR). Given heterogeneity of the studies, a random effect was applied. Risk of bias was analyzed using the Newcastle-Ottawa Scale. The study was conducted following the Preferred Reporting Items for Systematic Review and Meta-analysis guidelines. The protocol was registered in PROSPERO (No. CRD42024549924). RESULTS Eleven retrospective cohort studies involving 2953 patients were included in the meta-analysis. Among those, HE was investigated in eight studies, whereas PHE growth was evaluated in three. Blood sample was obtained on admission in ten studies, and at 24 hours in one study. There was no consensus on cut-off value among the studies. NLR was found to be significantly associated with higher odds of HE (OR = 1.09, 95%CI: 1.04-1.15, I 2 = 86%, P < 0.01), and PHE growth (OR = 1.28, 95%CI: 1.19-1.38, I 2 = 0%, P < 0.01). Qualitative analysis of each outcome revealed overall moderate risk of bias mainly due to lack of control for systemic confounders. CONCLUSION The available literature suggests that a possible association may exist between NLR on admission and HE, and PHE growth. Future studies controlled for systemic confounders should be designed to consolidate this finding. If confirmed, NLR could be added as a readily available and inexpensive biomarker to identify a subgroup of patients at higher risk of developing HE.
Collapse
Affiliation(s)
- Andrea Loggini
- Brain and Spine Institute, Southern Illinois Healthcare, Carbondale, IL 62901, United States
- Southern Illinois University School of Medicine, Carbondale, IL 62901, United States
| | - Jonatan Hornik
- Brain and Spine Institute, Southern Illinois Healthcare, Carbondale, IL 62901, United States
- Southern Illinois University School of Medicine, Carbondale, IL 62901, United States
| | - Jessie Henson
- Brain and Spine Institute, Southern Illinois Healthcare, Carbondale, IL 62901, United States
| | - Julie Wesler
- Brain and Spine Institute, Southern Illinois Healthcare, Carbondale, IL 62901, United States
| | - Alejandro Hornik
- Brain and Spine Institute, Southern Illinois Healthcare, Carbondale, IL 62901, United States
- Southern Illinois University School of Medicine, Carbondale, IL 62901, United States
| |
Collapse
|
8
|
Gao Y, Mao M, Li Y, Xuan M, Wu Y, He Q. A self-directed Trojanbot-enzymatic nanobot in neutrobot for active target therapy of glioblastoma. Nat Commun 2025; 16:5263. [PMID: 40480985 PMCID: PMC12144173 DOI: 10.1038/s41467-025-60422-z] [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: 01/13/2025] [Accepted: 05/22/2025] [Indexed: 06/11/2025] Open
Abstract
Chemotherapy is an important treatment for glioblastoma (GBM) and a key component of comprehensive GBM therapy. However, the blood-brain barrier (BBB) and complex tumor microenvironment (TME) restrict the diffusion of drugs, which greatly reduces the chemotherapeutic effect on GBM. Single strategies, such as cell-based nanobots to cross the BBB or enzymatic nanobots propelled by enriched substrates in the TME for deep tumor penetration, remain inadequate to address multiple barriers and achieve precise targeting. Here, we develop a Trojan horse-inspired enzymatic nanobot-in-neutrobot system (Trojanbot) to greatly enhance targeted GBM therapy. Trojanbots traverse the BBB by leveraging positive chemotaxis in response to tumor-derived chemokine gradients, after which the released catalase-driven nanobots (CatNbot) undergo directional movement along the H2O2 gradients in TME, facilitating deep tumor penetration. This multi-stage targeting strategy improves drug delivery efficiency, providing considerable potential as a clinical approach for brain tumor treatment.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Key Lab of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, 150001, Harbin, China
| | - Meng Mao
- Key Lab of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, 150001, Harbin, China.
- Key Lab of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, 450000, Zhengzhou, China.
| | - Yue Li
- Key Lab of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, 150001, Harbin, China
| | - Mingjun Xuan
- Wenzhou Institute, University of Chinese Academy of Sciences, 325000, Wenzhou, China
| | - Yingjie Wu
- Key Lab of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, 150001, Harbin, China.
- Key Lab of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, 450000, Zhengzhou, China.
| | - Qiang He
- Key Lab of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, 150001, Harbin, China.
- Key Lab of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, 450000, Zhengzhou, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, 325000, Wenzhou, China.
| |
Collapse
|
9
|
Aslanian-Kalkhoran L, Nouri N, Soltani-Zangbar MS, Mardi A, Aghebati-Maleki L. Immunoglobulin therapy for infertility and the role of immune cells in pregnancy success: An extensive investigation and update. J Reprod Immunol 2025; 169:104458. [PMID: 40015106 DOI: 10.1016/j.jri.2025.104458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
In the United States, roughly one out of every eight couples, or 7.5 million women, experience challenges related to conceiving or maintaining a pregnancy. The body's immune response is vital during pregnancy. T cells, natural killer (NK) cells, B cells, and macrophages (MQ) are immune cells in the female reproductive tract. They are in charge of maintaining tissue homeostasis and regulating the immune system's response to invasive pathogens. Failure to regulate these immune cells might result in inflammation, which reduces fertility. The immune system modulation of pregnancy loss has been studied with intralipid, intravenous immunoglobulin (IVIG), and paternal leukocyte vaccination. A concentrated antibody called intravenous immunoglobulin (IVIG) is utilized as a biological agent to treat autoimmune, viral, and inflammatory diseases and some immunodeficiencies. The main objective of this treatment is to restore a damaged immune system. IgGs, through binding to specific antigens, promote the innate immunity's cellular and humoral immune response by activating complements and binding to Fc receptors of several immune cells. Contrariwise, IVIG regulates pathogenic autoimmunity in animal models, including skin-blister diseases, nephrotoxic nephritis, and K/BxN arthritis. IVIG has, therefore, been of great interest as an immune modulator in several immune disorders. This review aims to investigate the immunological reasons of reproductive failure, focusing on the immunomodulatory effects of IVIG in its treatment.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narjes Nouri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Chen SH, Chen CH, Lin HC, Yeh SA, Hwang TL, Chen PJ. Drug repurposing of cyclin-dependent kinase inhibitors for neutrophilic acute respiratory distress syndrome and psoriasis. J Adv Res 2025; 72:485-500. [PMID: 39089617 DOI: 10.1016/j.jare.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Neutrophilic inflammation, characterized by dysregulated neutrophil activation, triggers a variety of inflammatory responses such as chemotactic infiltration, oxidative bursts, degranulation, neutrophil extracellular traps (NETs) formation, and delayed turnover. This type of inflammation is pivotal in the pathogenesis of acute respiratory distress syndrome (ARDS) and psoriasis. Despite current treatments, managing neutrophil-associated inflammatory symptoms remains a significant challenge. AIM OF REVIEW This review emphasizes the role of cyclin-dependent kinases (CDKs) in neutrophil activation and inflammation. It aims to highlight the therapeutic potential of repurposing CDK inhibitors to manage neutrophilic inflammation, particularly in ARDS and psoriasis. Additionally, it discusses the necessary precautions for the clinical application of these inhibitors due to potential off-target effects and the need for dose optimization. KEY SCIENTIFIC CONCEPTS OF REVIEW CDKs regulate key neutrophilic functions, including chemotactic responses, degranulation, NET formation, and apoptosis. Repurposing CDK inhibitors, originally developed for cancer treatment, shows promise in controlling neutrophilic inflammation. Clinical anticancer drugs, palbociclib and ribociclib, have demonstrated efficacy in treating neutrophilic ARDS and psoriasis by targeting off-label pathways, phosphoinositide 3-kinase (PI3K) and phosphodiesterase 4 (PDE4), respectively. While CDK inhibitors offer promising therapeutic benefits, their clinical repurposing requires careful consideration of off-target effects and dose optimization. Further exploration and clinical trials are necessary to ensure their safety and efficacy in treating inflammatory conditions.
Collapse
Affiliation(s)
- Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan.
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Hsin-Chieh Lin
- Department of Chinese Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824410, Taiwan; School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Shyh-An Yeh
- Medical Physics and Informatics Laboratory of Electronic Engineering and Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan; Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung 824410, Taiwan; Department of Radiation Oncology, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan.
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan; Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan.
| |
Collapse
|
11
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Maiocchi S, Burnham EE, Cartaya A, Lisi V, Buechler N, Pollard R, Babaki D, Bergmeier W, Pinkerton NM, Bahnson EM. Development of DNase-1 Loaded Polymeric Nanoparticles Synthesized by Inverse Flash Nanoprecipitation for Neutrophil-Mediated Drug Delivery to In Vitro Thrombi. Adv Healthc Mater 2025; 14:e2404584. [PMID: 40341904 DOI: 10.1002/adhm.202404584] [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: 11/17/2024] [Revised: 03/31/2025] [Indexed: 05/11/2025]
Abstract
Activated neutrophils release Neutrophil Extracellular Traps (NETs), comprising decondensed chromatin, peroxidases, and serine proteases, which aid in host defense but are also implicated in thrombosis and resistance to thrombolysis. Recombinant DNase 1, which degrades NETs, may aid in thrombus dissolution synergistically with fibrinolytics. However, its short half-life and susceptibility to plasma proteases limit its therapeutic applicability. To address these limitations, DNase1 is encapsulated into polymeric nanoparticles (DNPs) using inverse Flash Nanoprecipitation (iFNP), a scalable nanoparticle synthesis technique. Previously only used with model proteins, the study demonstrates for the first time the feasibility of extending iFNP to the encapsulation of therapeutic proteins. Conditions that promote DNase1 solubility, preserve activity, and demonstrate release resulting in ex vivo NET degradation are detailed. Furthermore, the use of neutrophils, the source of NETs, as carriers for DNPs to enhance targeted delivery is investigated. These findings confirm that DNP-loaded neutrophils maintain key functionalities, including viability and oxidative burst, and associate with in vitro blood clots to deliver nanoparticles, and DNase1 protein. This study not only extends the feasibility of applying iFNP to encapsulate therapeutic proteins into polymeric nanoparticles, a promising alternative to lipid nanoparticles, but also contributes to the emerging literature on neutrophils as delivery vectors for nanocarriers.
Collapse
Affiliation(s)
- Sophie Maiocchi
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Erica E Burnham
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Ana Cartaya
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Light Microscopy Core Facility, Duke University, Durham, NC, 27710, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Veronica Lisi
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Nancy Buechler
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Rachel Pollard
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Danial Babaki
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nathalie M Pinkerton
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Edward M Bahnson
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
13
|
Sul C, Lewis CV, Posey J, Jordan M, Colon Hidalgo D, Porfilio T, Elajaili H, McCormack G, Burciaga S, Delaney C, Nozik ES. Increased Circulating Extracellular Superoxide Dismutase Attenuates Platelet-Neutrophil Interactions. Am J Respir Cell Mol Biol 2025; 72:653-662. [PMID: 39531632 DOI: 10.1165/rcmb.2024-0292oc] [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: 06/25/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024] Open
Abstract
Acute respiratory distress syndrome is a serious illness accounting for 10% of ICU admissions and has a high mortality of 31-45%, with a paucity of pharmacologic treatment options. Dysregulated inflammation and oxidative stress are hallmark features of acute respiratory distress syndrome. We previously showed that transgenic mice expressing a naturally occurring polymorphism of the antioxidant enzyme EC-SOD (extracellular superoxide dismutase) are protected against Staphylococcus aureus pneumonia, acute lung injury, and pulmonary neutrophilia. In this mouse strain, an R213G amino acid substitution leads to lower tissue-binding affinity and elevated alveolar and plasma EC-SOD amounts, although the redox-regulated mechanisms responsible for protection against S. aureus are not yet elucidated. Neutrophils are recruited to the areas of injury and inflammation, in part by activated platelets, which contain multiple redox-sensitive targets. Thus, we hypothesize that increased circulating EC-SOD due to the EC-SOD R213G variant protects against S. aureus pneumonia by reducing platelet activation and subsequent neutrophil recruitment to the lung. We demonstrate that, compared with wild-type mice with S. aureus pneumonia, platelet activation, formation of platelet-neutrophil aggregates, and influx of neutrophils and platelet-neutrophil aggregates into the lung are decreased in the infected R213G mice. Furthermore, pretreatment with a MnTE-2-PyP SOD mimetic protects against S. aureus-induced platelet activation, pulmonary neutrophilia, and acute lung injury. Our data highlight the redox regulation of platelet activation as a driver of S. aureus-induced acute lung injury.
Collapse
Affiliation(s)
- Christina Sul
- Division of Critical Care
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Caitlin V Lewis
- Division of Critical Care
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | - Daniel Colon Hidalgo
- Division of Pulmonology and Critical Care, Department of Internal Medicine, and
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Hanan Elajaili
- Division of Critical Care
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | - Cassidy Delaney
- Division of Neonatology, Department of Pediatrics
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Eva S Nozik
- Division of Critical Care
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
14
|
Vetsika EK, Katsianou MA, Sarantis P, Palamaris K, Papavassiliou AG, Piperi C. Pediatric gliomas immunity challenges and immunotherapy advances. Cancer Lett 2025; 618:217640. [PMID: 40090572 DOI: 10.1016/j.canlet.2025.217640] [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: 02/11/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Pediatric gliomas, the most frequent brain tumors in children, are characterized by heterogeneity and a unique tumor immune microenvironment. They are categorized into different subtypes, including low-grade gliomas like pilocytic astrocytomas and high-grade gliomas such as diffuse midline gliomas and diffuse intrinsic pontine gliomas, each exhibiting distinct immunological profiles. The tumor immune microenvironment in pediatric gliomas is shaped by cellular and non-cellular components, including immune cells, cytokines, and the extracellular matrix, involved in tumor progression, immune evasion, and response to therapy. While pediatric low-grade gliomas often display an immunosuppressed microenvironment, high-grade gliomas are characterized by complex immune infiltrates and intricate immunosuppressive mechanisms. The blood-brain barrier further obscures immune cell recruitment and therapeutic delivery. Despite advances in understanding adult gliomas, the immunobiology of pediatric tumors is poorly investigated, with limited data on the interactions between glioma cells and immune populations such as T and natural killer cells, as well as tumor-associated macrophages. Herein, we provide an update of the current knowledge on tumor immune microenvironment interactions in pediatric gliomas, highlighting the immunosuppressive mechanisms and emerging immunotherapeutic strategies aiming at overcoming these barriers to improve clinical outcomes for affected children.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria A Katsianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Palamaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
15
|
Ma X, Huo K, Gao H, Ge H. The clinical value of lymphocyte percentages and the monocyte-to-lymphocyte ratio in differentiating immune-mediated necrotizing myopathy from dermatomyositis. Front Neurol 2025; 16:1581206. [PMID: 40492171 PMCID: PMC12146153 DOI: 10.3389/fneur.2025.1581206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Objective Immune-mediated necrotizing myopathy (IMNM) and dermatomyositis (DM) represent distinct subtypes of idiopathic inflammatory myopathies (IIMs). While both conditions share clinical manifestations, including muscle weakness and inflammatory infiltrates on muscle biopsy, their pathophysiological characteristics differ significantly. This study investigated the clinical utility of hematological inflammatory biomarkers in differentiating these two entities. Methods In this retrospective analysis, we compared complete blood count parameters among 27 patients with IMNM, 14 patients with DM, and 85 healthy controls (HC). Demographic characteristics, clinical presentations, and hematological indices including the neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR) were analyzed. Results Myalgia and skin rash were observed more frequently in the DM group compared to the IMNM group. The patients with IMNM exhibited significantly higher serum creatine kinase (CK) and lactate d0.2815ehydrogenase levels. Red blood cell distribution width (RDW), monocyte counts, and MLR were elevated in the patients with IMNM compared to the HC. The patients with DM showed significantly increased neutrophil percentages, monocyte percentages, monocyte counts, NLR, MLR, and PLR, as well as decreased lymphocyte percentages and counts, compared to the HC. When directly comparing DM and IMNM, the patients with DM had lower lymphocyte percentages and counts, along with higher NLR and MLR. Receiver operating characteristic (ROC) curve analysis revealed that lymphocyte percentages and the MLR had moderate predictive value for differentiating IMNM from DM, with area under the curve (AUC) values of 0.709 and 0.7487, respectively. Conclusion RDW and the MLR in IMNM and the NLR, MLR, and PLR in DM represent accessible and cost-effective biomarkers for assessing inflammation. Lymphocyte percentages and the MLR may serve as inexpensive and readily available supplementary markers for distinguishing IMNM from DM.
Collapse
Affiliation(s)
- Xue Ma
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Kaikai Huo
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huajie Gao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huizhen Ge
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Xiang Y, Mao W. Neutrophil-derived ratios as predictors of short-term mortality in HBV-associated decompensated cirrhosis. BMC Gastroenterol 2025; 25:404. [PMID: 40419938 PMCID: PMC12105264 DOI: 10.1186/s12876-025-03991-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Hepatitis B virus-associated decompensated cirrhosis (HBV-DC) is recognized as a critical illness with an increased risk of short-term mortality. Neutrophil-derived ratios, including neutrophil-to-lymphocyte ratio (NLR), neutrophil-to-albumin ratio, neutrophil-to-high-density lipoprotein-cholesterol ratio, neutrophil-to-hemoglobin ratio, and neutrophil-to-platelet ratio, have emerged as potential prognostic markers in various liver diseases. The present study aimed to determine the effectiveness of these neutrophil-derived ratios for prediction of mortality in patients with HBV-DC. METHODS We conducted a retrospective analysis of HBV-DC patients at our hospital between April 2022 and April 2024. The study endpoint was the 30-day mortality rate. These neutrophil-derived ratios were calculated from data obtained during routine laboratory tests on admission. Disease severity was assessed using the Model for End-Stage Liver Disease (MELD) score. Multivariate regression analyses and receiver operating characteristic (ROC) curve analyses were conducted. RESULTS The study investigated 160 HBV-DC patients, of whom 23 (14.4%) experienced mortality within 30 days. Non-survivors exhibited markedly higher values for neutrophil-derived ratios than survivors. All neutrophil-derived ratios were associated with mortality in univariate analyses, but only NLR and MELD score remained as independent predictors of mortality in multivariate analyses. In the ROC analyses, NLR showed a similar prognostic value to MELD score. Moreover, both NLR and MELD score had high specificity for prediction of mortality in HBV-DC patients. CONCLUSIONS Among neutrophil-derived ratios, NLR stands out as a simple and reliable predictor of mortality in HBV-DC patients.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Endocrinology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - WeiLin Mao
- Department of Clinical Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| |
Collapse
|
17
|
Xia C, Liu Y, Qing X. Characteristic genes and immune infiltration analysis of gastric cancer based on bioinformatics analysis and machine learning. Discov Oncol 2025; 16:872. [PMID: 40407862 PMCID: PMC12102041 DOI: 10.1007/s12672-025-02624-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Gastric cancer (GC), a common and deadly malignancy worldwide, is a serious burden on society and individuals. However, available diagnostic biomarkers for GC are very limited. The current study aimed to identify potential diagnostic biomarkers for GC and analyze the activity of infiltrating immune cells in this pathology. METHODS Microarray data for GC were acquired from the Gene Expression Omnibus (GEO) database. The limma package was utilized to normalize these data, thus identifying differentially expressed genes (DEGs). For normalized data of samples, we established a weighted gene co-expression network (WGCNA) to reveal key genes in the significant module. Afterward, we obtained overlapping genes by intersecting the DEGs and the key genes from the WGCNA module. Next, after applying the three algorithms (LASSO, RandomForest, and SVM-RFE) to analyze these overlapping genes and take the intersection, we established a GC diagnosis. The diagnostic significances of these identified genes were evaluated with receiver operating characteristic (ROC) curves and validated in the external dataset. Furthermore, ssGSEA and CIBERSORT were employed for evaluating the infiltrating immune cells and the association of the immune cells and diagnostic biomarkers. RESULTS Herein, we identified 49 overlapping genes, and the results of enrichment analysis demonstrated that these genes may be involved in the signaling transduction-related process. Finally, BANF1, DUSP14, and VMP1 were regarded as key biomarkers in GC patients based on the overlapping genes that we found, and these three biomarkers demonstrated great diagnostic significance. Additionally, the hub biomarkers had different levels of association with macrophages, neutrophils, memory B cells, and plasma cells. CONCLUSIONS BANF1, DUSP14, and VMP1 are promising diagnostic biomarkers for GC, and infiltrating immune cells may dramatically affect gastric carcinogenesis and progression.
Collapse
Affiliation(s)
- Chengwei Xia
- Department of Thyroid and Breast Surgery, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yini Liu
- Department of Anesthesiology, The People's Hospital of Zhongjiang, Deyang, China
| | - Xin Qing
- Department of Hepatobiliary Vascular Surgery, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China.
| |
Collapse
|
18
|
Du LY, Keerthisinghe P, Rolland L, Sung YJ, Darroch H, Linnerz T, Ashimbayeva E, Grant MJ, Kakadia PM, Ramachandran A, Tups A, Spaink HP, Bohlander SK, Cheeseman J, Crosier PS, Astin JW, Warman G, Hall CJ. A light-regulated circadian timer optimizes neutrophil bactericidal activity to boost daytime immunity. Sci Immunol 2025; 10:eadn3080. [PMID: 40408429 DOI: 10.1126/sciimmunol.adn3080] [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: 12/03/2023] [Revised: 01/21/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
The immune response exhibits strong circadian rhythmicity, with enhanced bacterial clearance often synchronized with an organism's active phase. Despite providing the bulk of cellular antibacterial defense, the neutrophil clockwork is poorly understood. Here, we used larval zebrafish to explore the role of clock genes in neutrophils during infection. Per2 was required in neutrophils for reactive oxygen species (ROS) production and bacterial killing by enhancing infection-responsive expression of high-mobility group box 1a (hmgb1a). The Cry binding domain of Per2 was required for regulation of neutrophil bactericidal activity, and neutrophils lacking Cry1a had elevated bactericidal activity and infection-responsive hmgb1a expression. A conserved cis-regulatory element with BMAL1 and nuclear factor κB binding motifs gated infection-responsive hmgb1a expression to the light phase. Mutagenesis of the BMAL1 motif in neutrophils blunted the priming effect of light on bactericidal activity and hmgb1a expression. These findings identify a light-responsive cell-intrinsic timer that controls time-of-day variations in antibacterial activity.
Collapse
Affiliation(s)
- Lucia Yi Du
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Pramuk Keerthisinghe
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Centre For Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Leah Rolland
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yih Jian Sung
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Hannah Darroch
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tanja Linnerz
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Elina Ashimbayeva
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | - Purvi M Kakadia
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Annasuya Ramachandran
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Alexander Tups
- Centre For Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Herman P Spaink
- Institute of Biology, Leiden University, Leiden, Netherlands
| | - Stefan K Bohlander
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - James Cheeseman
- Department of Anaesthesiology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Guy Warman
- Department of Anaesthesiology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
19
|
Zhang Y, Wu Q, Wang Y, Chen Q, Han S, Li B, Zhao Q, Wang Q, Wang Y, Gao Y. Systemic Inflammation and Disruption of the Local Microenvironment Compromise Muscle Regeneration: Critical Pathogenesis of Autoimmune-Associated Sarcopenia. Interact J Med Res 2025; 14:e64456. [PMID: 40407708 PMCID: PMC12124038 DOI: 10.2196/64456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 06/02/2025] Open
Abstract
Unlabelled Sarcopenia is defined by age-related reductions in muscle mass, strength, and physiological function, and it is especially prevalent among individuals with autoimmune diseases. Autoimmune disorders, characterized by immune dysregulation, cause systemic inflammation and damage to multiple tissues through unregulated immune activity. Research indicates that autoimmune diseases negatively impact skeletal muscle functions and may worsen the progression of sarcopenia. This viewpoint comprehensively discusses the pathogenesis and potential mechanism of sarcopenia in 3 autoimmune diseases: inflammatory bowel disease, rheumatoid arthritis, and type 1 diabetes mellitus. Mechanistically, chronic immune microenvironment alterations induce compartment-specific redistribution of leukocyte subsets and cytokine networks. These perturbations disrupt critical signaling pathways governing muscle protein synthesis, satellite cell activation, and mitochondrial bioenergetics, leading to impaired regeneration and accelerated sarcopenia progression. By delineating shared and distinct pathomechanisms across these models, this analysis reframes our understanding of immune-mediated muscle wasting. Beyond mechanistic insights, it establishes a translational framework for targeted therapies and highlights emerging research directions bridging immunology and age-related musculoskeletal decline.
Collapse
Affiliation(s)
- Yingjuan Zhang
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Qingqian Wu
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Yi Wang
- Yixing Center for Disease Control and Prevention, Yixing, Jiangsu Province, China
| | - Qingyan Chen
- Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuang Han
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Bei Li
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Qingwen Zhao
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Qianzhuo Wang
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Yule Wang
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| | - Yue Gao
- Department of Geriatrics, Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China, 86 13706511908
| |
Collapse
|
20
|
Li S, Duan F, Qiu Z, Nan Z, Cao X, Yang C, Li W, Shen B. Polymer-Conjugated SOD-Pt⁰ Micelles Enhance ROS Cascade Scavenging to Alleviate Ischemia-Reperfusion Injury During Kidney Transplantation. Adv Healthc Mater 2025:e2500696. [PMID: 40394947 DOI: 10.1002/adhm.202500696] [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: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Ischemia-reperfusion injury (IRI) during kidney transplantation is linked to oxidative stress induced by excessive reactive oxygen species (ROS), which causes the injury of transplanted kidney, leading to further intensified organ shortages. Protein-based antioxidants have been developed for ROS scavenging via cascade biocatalyst. The in situ growth of metal nanozymes on proteins effectively decreases the steric hindrance between active sites, improving the efficiency of cascade biocatalysts. However, the poor stability of protein during the process of preparation and intracellular delivery leads to low therapeutic effects. In this study, three different functional polymers are conjugated to SOD for the formation of micelles. Surprisingly, it is found that the conjugated ultra-acid sensitive polymer efficiently preserves the enzymatic activity of SOD, due to great endo/lysosomal escape capacity. Subsequently, SOD micelles (SOE) are used as a template to prepare SOE-Pt0 (SOEP) through in situ growth of Pt0 with vicinal enzymatic active sites. The preparation process minimally impacts on the activity of SOD, owing to improved stability. The system exhibits effective cascade ROS scavenging, significantly reducing kidney damage and inflammation caused by IRI. The research offers a novel approach for addressing IRI challenges in organ transplantation and provides a promising strategy to mitigate organ shortages.
Collapse
Affiliation(s)
- Shengzhou Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Fei Duan
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Zhiwen Qiu
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Zhuofan Nan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xiangqian Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Chenkai Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Wei Li
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
- Department of Urology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| |
Collapse
|
21
|
Wei H, Xia D, Li L, Liang L, Ning L, Gan C, Wu Y. Baicalin Modulates Glycolysis via the PKC/Raf/MEK/ERK and PI3K/AKT Signaling Pathways to Attenuate IFN-I-Induced Neutrophil NETosis. Mediators Inflamm 2025; 2025:8822728. [PMID: 40420943 PMCID: PMC12105894 DOI: 10.1155/mi/8822728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/19/2025] [Indexed: 05/28/2025] Open
Abstract
Type I interferon (IFN-I), a pivotal component of the host's innate antiviral immune system, can induce the formation of neutrophil extracellular traps (NETs) and facilitate inflammatory responses. Baicalin exhibits a range of pharmacological activities, including anti-inflammatory and immunomodulatory effects. It has been reported that neutrophil glycolysis plays a pivotal role in the formation of NETs and the regulation of inflammatory response in immune modulation, regulated by IFN-I. However, it remains unclear whether baicalin regulates IFN-I-induced NETs formation through glycolysis. In this study, we induced the formation of NETs in vitro using IFN-I and observed that baicalin significantly reduced the formation of IFN-I-induced NETs. Furthermore, baicalin inhibited the production of pro-inflammatory cytokines, specifically interleukin-1 beta (IL-1β) and interleukin-6 (IL-6), as well as the generation of reactive oxygen species (ROS) and chemotactic responses. Our findings further indicated that baicalin could inhibit both lactic acid and ATP levels in IFN-I-induced neutrophils, as well as the expression of glycolytic-related proteins, including HK2, HK3, PKM2, and LDHA. Moreover, following the administration of glycolytic agonists insulin, it was observed that heightened glycolytic activity significantly augmented NETs formation and the release of inflammatory cytokines, potentially regulated by PKC/Raf/MEK/ERK and PI3K/AKT signaling pathways. In conclusion, our findings indicated that baicalin may exert inhibitory effects on IFN-I-induced NETs formation and inflammatory cytokine production by modulating glycolysis, thereby providing further evidence for the potential clinical application of baicalin in the treatment of IFN-I-related inflammatory diseases.
Collapse
Affiliation(s)
- Hong Wei
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Dongni Xia
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Li Li
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Linpan Liang
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Lijun Ning
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Cuiliu Gan
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Ying Wu
- Liuzhou Key laboratory of Infection Disease and Immunology, Research Center of Medical Sciences, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| |
Collapse
|
22
|
Kato KT, Ferreira GCS, Fonseca DLM, Moretti EH, Trzan IFL, Filgueiras IS, Nobile AL, Adri AS, Fonseca MT, Souza RN, Matos CM, Sales MOR, Lino CA, Teramoto MM, Muxel SM, Cabral-Marques O, Steiner AA. The greater splanchnic nerve preferentially regulates neutrophils over macrophages in a rat model of septic peritonitis. Brain Behav Immun 2025; 129:30-41. [PMID: 40389038 DOI: 10.1016/j.bbi.2025.05.015] [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: 10/08/2024] [Revised: 05/01/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025] Open
Abstract
The sympathetic splanchnic nerve is a major player in immunoregulation, but its specific roles during infection have yet to be elucidated. Here, we evaluated how bilateral ablation of the greater splanchnic nerve (SplancX) impacts bacterial burden and immune function in a rat model of E. coli-induced septic peritonitis. SplancX had a major effect on bacterial burden within 24 h, reducing it to 4 % in the peritoneum and to 8 % in the spleen of what was found in the sham-operated controls. Such a major effect was not explained by gross changes in the infiltration of these sites with innate immune cells (neutrophils and macrophages), as assessed by flow cytometry. Single-cell RNA sequencing was then employed to evaluate the cellular activation programs of leukocyte subsets. Of the nine cellular clusters identified in the peritoneum of the infected rats, three of them had a transcriptional signature of activated neutrophils and two of them corresponded to quiescent neutrophils with an immunosuppressive signature. SplancX shifted the balance between these neutrophil subsets in a way consistent with heightened immunity, i.e., the activated neutrophils were augmented whereas the quiescent neutrophils were reduced in the SplancX group. The remainder of the clusters consisted of macrophages and erythrocytes, none of which changed in a way that could account for the observed effects on bacterial clearance. Confirming that SplancX resulted in heightened neutrophil activation, protein markers of neutrophil degranulation and NETosis were found to be elevated in the peritoneal lavage of the SplancX group. Taken together, the data show that the splanchnic nerve exerts a major effect on bacterial clearance in the acute phase of infection, presumably owing to selective changes in the balance between microbicidal and quiescent subsets of neutrophils.
Collapse
Affiliation(s)
- Kathia T Kato
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Gabriela C S Ferreira
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Dennyson L M Fonseca
- Programa Interunidades de Pos-graduação em Bioinformática, Instituto de Matematica e Estatistica, Universidade de Sao Paulo, Sao Paulo, SP 05508-090, Brazil
| | - Eduardo H Moretti
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Isis F L Trzan
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Igor Salerno Filgueiras
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Adriel L Nobile
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciencias Farmaceuticas, Universidade de São Paulo, Sao Paulo, SP 05505-000, Brazil
| | - Anny S Adri
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciencias Farmaceuticas, Universidade de São Paulo, Sao Paulo, SP 05505-000, Brazil
| | - Monique T Fonseca
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Rayssa N Souza
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Caroline M Matos
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Manoela O R Sales
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Caroline A Lino
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Mariana M Teramoto
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Sandra M Muxel
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Otávio Cabral-Marques
- Programa Interunidades de Pos-graduação em Bioinformática, Instituto de Matematica e Estatistica, Universidade de Sao Paulo, Sao Paulo, SP 05508-090, Brazil; Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciencias Farmaceuticas, Universidade de São Paulo, Sao Paulo, SP 05505-000, Brazil; Departamento de Clinica Medica, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP 01246-903, Brazil; Instituto D'OR de Pesquisa e Ensino, Rio de Janeiro, RJ 22281-100, Brazil
| | - Alexandre A Steiner
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil.
| |
Collapse
|
23
|
Li S, Zhou Y, Hu T, Li M, Luo Y, Chen J. Migrasomes: key players in immune regulation and promising medical applications. Front Immunol 2025; 16:1592314. [PMID: 40443653 PMCID: PMC12119291 DOI: 10.3389/fimmu.2025.1592314] [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: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Migrasomes are newly discovered extracellular organelles released by migrating cells, such as immune cells, tumor cells, and other special functional cells like podocytes and embryonic cells. They contain a diverse array of constituents, including proteins, lipids, and RNA which can be released to the designated location to activate surrounding cells, thereby facilitating intercellular communication and signal transduction. Since then, our understanding of the mechanism and function of the migrasomes has expanded exponentially, with recent evidence indicating they are involved in various physiological and pathological processes, particularly in immune regulation. Furthermore, methods and techniques for extracting, detecting, and characterizing migrasomes are constantly advancing. Herein, we summarize the current understanding of migrasomes and their key roles in modulating immune responses, as well as the prospective challenges surrounding their clinical application, aiming to provide novel insights into the emerging organelles.
Collapse
Affiliation(s)
| | | | | | | | - Yao Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Abdelmawgood IA, Mohamed AS, Mahana NA, Abdel Wahab AHA, Badr AM, Abdelkader AE. Chrysin-loaded PLGA nanoparticle attenuates ferroptosis in lipopolysaccharide-induced indirect acute lung injury by upregulating Nrf2-dependent antioxidant responses. Respir Physiol Neurobiol 2025; 336:104451. [PMID: 40379234 DOI: 10.1016/j.resp.2025.104451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Chrysin (CHR) is the principal active compound in honey, propolis and plants. Its pharmacological effects include anti-inflammatory, antiallergic, and antioxidant capabilities. However, its poor solubility and bioavailability constitute a limitation. In this study, Poly-lactic-co-glycolic acid (PLGA) was used as a nanocarrier to enhance the stability, bioavailability, and effectiveness of CHR to protect mice from indirect acute lung injury (ALI) caused by lipopolysaccharide (LPS). CHR-loaded PLGA nanoparticle (CHR-NP) was prepared and characterized using techniques such as FTIR, zeta potential analysis, DLS, in vitro drug release assessment, encapsulation efficiency measurement, and TEM. Prior to the intraperitoneal injection of LPS (10mg/kg), C57BL/6 mice were orally administered CHR (50mg/kg), PLGA (50mg/kg), CHR-NP (50mg/kg), and dexamethasone (Dexa) (5mg/kg) for a duration of six days. Following 24hours of LPS or normal saline (control) injection, the mice were anesthetized. CHR-NP increased catalase, glutathione, and glutathione peroxidase while decreasing malondialdehyde, myeloperoxidase, nitric oxide, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-12, and interferon (IFN)-γ. Moreover, treatment with CHR-NP augmented the gene and protein expression of the Keap1/Nrf2/ARE signaling pathway utilizing quantitative real-time PCR (RT-PCR), western blotting, and immunohistochemistry. Additionally, CHR-NP reduced histological alterations, pulmonary edema, damage, and iron deposition. Our findings indicate that CHR-NP significantly mitigated indirect ALI, primarily through the suppression of inflammation, oxidative stress, and ferroptosis via the activation of the Keap1/Nrf2/ARE signaling pathways.
Collapse
Affiliation(s)
| | - Ayman Saber Mohamed
- Zoology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Noha A Mahana
- Zoology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | | | - Abeer Mahmoud Badr
- Zoology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | | |
Collapse
|
25
|
Su JH, Lin DY, Liu XH, Zhang JL, Li ZG, Tao EX, Huang KX. Identifying potential co-expressed genes and molecular mechanisms linking post-COVID-19 and Guillain-Barre syndrome through neutrophil extracellular trap-related genes. Front Neurol 2025; 16:1447725. [PMID: 40433617 PMCID: PMC12109036 DOI: 10.3389/fneur.2025.1447725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/31/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Neutrophil extracellular traps (NETs) play a pivotal role in immunity and autoinflammatory disease, leading us to hypothesize that NETs are crucial in Guillain-Barre Syndrome (GBS) after SARS-CoV-2 infection. Methods By collecting six Gene Expression Omnibus (GEO) datasets from the GEO database and dividing them into discovery and validation sets, we screened differentially expressed genes (DEGs) within the discovery set, with further analyses using functional enrichment analysis. Using single-sample gene set enrichment analysis (ssGSEA), we assessed immune cell infiltration in both coronavirus disease 2019 (COVID-19) and GBS datasets. NETs-related genes (NETRGs) were identified through a protein-protein interaction (PPI) network and NETs gene datasets. Finally, candidate drugs were screened using Connectivity Map. Results In this study, a total of 3254 DEGs were identified from the COVID-19 dataset, and 692 DEGs were obtained from the GBS dataset. Among these, 145 co-expressed DEGs were obtained. Bioinformatics functional analysis indicated that co-expressed DEGs were predominantly gathered in immune-related and inflammatory response pathways. Employing various algorithms, we identified MMP9, CAMP, and CASP1 as NETRGs, demonstrating good discriminatory capacity in COVID-19 and GBS. Notably, neutrophils and macrophages were identified as co-upregulated differential immune infiltrating cells significantly associated with both COVID-19 and GBS. Moreover, we identified 10 candidate drugs for patients with post-COVID-19 GBS. Conclusion In conclusion, MMP9, CASP1, and CAMP were identified as promising biomarkers and potential targets for therapy of post-COVID-19 GBS.
Collapse
Affiliation(s)
- Jie-Hua Su
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dan-Yu Lin
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Huan Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie-Li Zhang
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhong-Gui Li
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - En-Xiang Tao
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Kai-Xun Huang
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
26
|
Gao F, Zhou R, He Y, Zhang Y, Bao C, Feng G. Bio-Mimicking Nanoparticle System Facilitates Sonodynamic-Mediated Clearance of Extensively Drug-Resistant Bacteria. ACS Biomater Sci Eng 2025; 11:2988-3002. [PMID: 40294106 DOI: 10.1021/acsbiomaterials.4c02455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The increasing prevalence of carbapenem-resistant and extensively drug-resistant Acinetobacter baumannii (XDR-Ab) poses a critical challenge in treating hospital-acquired pulmonary infections. In this study, we developed a biomimetic neutrophil membrane-coated nanoparticle system, NM@PCN-TIG, for the targeted delivery of tigecycline (TIG). The system utilizes the porphyrin-based metal-organic framework (MOF) PCN-224 as the core of the nanoparticle, encapsulating TIG and coated with a neutrophil membrane (NM) to enhance immune evasion and targeting of infection sites. Its loading efficiency, controlled release properties, cytotoxicity, and bactericidal activity under ultrasound mediation were systematically evaluated in vitro and in vivo. Our results demonstrated that NM@PCN-TIG significantly enhanced the bactericidal efficacy of TIG, increased reactive oxygen species (ROS) production, and promoted macrophage polarization toward an anti-inflammatory phenotype. This innovative biomimetic TIG nanosystem shows great potential as a platform for addressing XDR-Ab-induced pneumonia.
Collapse
Affiliation(s)
- Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Runlu Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Cui Bao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
27
|
Cheng M, Hou Y, Chen Q, Ge S, Chen C, Zheng X, Zhang C. Exploring the wound-healing mechanism of Cayratia japonica extract: A combined experimental and network pharmacology study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119810. [PMID: 40239878 DOI: 10.1016/j.jep.2025.119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wound healing is a complex biological process and remains a significant challenge due to the lack of effective therapeutic drugs. Cayratia japonica (CJG), a traditional folk medicine, has been widely used for its anti-inflammatory and efficacy in treating traumatic injuries. AIM OF THE STUDY This study aimed to investigate the wound-healing effects of CJG and elucidate its underlying mechanism. METHODS First, the phytochemical composition of CJG was identified using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS), and its potential wound-healing mechanisms were predicated via network pharmacology. Next, in vivo experiments were conducted by dividing subjects into control, CJG (1.5-6 mg/cm2), and bFGF (150 IU/cm2) groups to assess its therapeutic efficacy. Finally, the mechanism of CJG and its key bioactive component, luteolin-7-O-glucoside (LUT-7G), were explained through polymerase chain reaction (PCR), Western blotting, histopathology, immunofluorescence, plasmid transfection, colony formation unit assays, and cellular thermal shift assay (CETSA). RESULTS LC-MS/MS identified 15 major constituents of CJG and 102 potential wound healing-related targets. Network pharmacology analysis revealed key enriched pathways, including AMPK, TNF, and metabolic pathways. In vivo, CJG significantly accelerated wound-healing by inhibiting inflammatory responses, promoting angiogenesis, and modulating collagen deposition. In vitro, LUT-7G treatment markedly enhanced the proliferation and migration of HaCaT and HSF cells. Mechanistically, LUT-7G exerted its wound-healing effects by activating the AMPK/CTHRC1/TGF-β1 signaling pathway in HaCaT cells. In conclusion, CJG significantly promotes wound healing by regulating AMPK signaling pathways, indicating its promising clinical application prospects.
Collapse
Affiliation(s)
- Mengqin Cheng
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Hou
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China
| | - Qi Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Shanchun Ge
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Ce Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Xueping Zheng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China.
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
28
|
Achary ST, Gupta P, Rajput A, Sohkhia W, Bonam SR, Sahu BD. Phytochemicals Targeting Inflammatory Pathways in Alcohol-Induced Liver Disease: A Mechanistic Review. Pharmaceuticals (Basel) 2025; 18:710. [PMID: 40430529 PMCID: PMC12115344 DOI: 10.3390/ph18050710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Alcoholic beverages play a significant role in social engagement worldwide. Excessive alcohol causes a variety of health complications. Alcohol-induced liver disease (ALD) is responsible for the bulk of linked fatalities. The activation of immune mechanisms has a crucial role in developing ALD. No effective medication promotes liver function, shields the liver from harm, or aids in hepatic cell regeneration. Alcohol withdrawal is one of the most beneficial therapies for ALD patients, which improves the patient's chances of survival. There is a crucial demand for safe and reasonably priced approaches to treating it. Exploring naturally derived phytochemicals has been a fascinating path, and it has drawn attention in recent years to modulators of inflammatory pathways for the prevention and management of ALD. In this review, we have discussed the roles of various immune mechanisms in ALD, highlighting the importance of intestinal barrier integrity and gut microbiota, as well as the roles of immune cells and hepatic inflammation, and other pathways, including cGAS-STING, NLRP3, MAPK, JAK-STAT, and NF-kB. Further, this review also outlines the possible role of phytochemicals in targeting these inflammatory pathways to safeguard the liver from alcohol-induced injury. We highlighted that targeting immunological mechanisms using phytochemicals or herbal medicine may find a place to counteract ALD. Preclinical in vitro and in vivo investigations have shown promising results; nonetheless, more extensive work is required to properly understand these compounds' mechanisms of action. Clinical investigations are very crucial in transferring laboratory knowledge into effective patient therapy.
Collapse
Affiliation(s)
- Swati Tirunal Achary
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India
| | - Prerna Gupta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India
| | - Apoorva Rajput
- Vaccine Immunology Laboratory, Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Wanphidabet Sohkhia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India
| | - Srinivasa Reddy Bonam
- Vaccine Immunology Laboratory, Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bidya Dhar Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India
| |
Collapse
|
29
|
Yu Q, Zhang Q, Zhu J, Pan F, Zhang H, Chen L, Shen J, Liu Y, Ji Z, Zhu Y, Chen Q, Yang Y. Inhalable neutrophil-mimicking nanoparticles for chronic obstructive pulmonary disease treatment. J Control Release 2025; 381:113648. [PMID: 40118116 DOI: 10.1016/j.jconrel.2025.113648] [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: 11/29/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is an intractable disease with thick mucus layer in bronchi and alveoli, frequently accompanied by bacterial infection. Anti-bacterial drugs with mucus penetrating are urgently needed for efficient COPD treatment. Here, a neutrophil-mimicking nanovehicle was developed by coating neutrophil membrane onto poly(lactic-co-glycolic acid) (PLGA) nanoparticles containing antibiotics levofloxacin (LVX). Neutrophil membrane coated nanoparticles (LVX@PLGA@Mem) reserved most of the membrane proteins and related membrane functions of neutrophil, exhibiting pro-inflammatory cytokines neutralization, inflammation inhibition, successfully delivering LVX through the mucus layer and achieving satisfactory anti-infection effects. Thus, LVX@PLGA@Mem after inhalation could remarkably reduce inflammation and infection in the lung with COPD. Therefore, neutrophil mimicking nanovehicles may be a feasible and desirable drug carrier for lung-related disease treatment in further clinic.
Collapse
Affiliation(s)
- Qifan Yu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Qiang Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China; School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Jiafei Zhu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Feng Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Han Zhang
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Linfu Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jingjing Shen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yanbin Liu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhaoxin Ji
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yuming Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Materials Science and Engineering, Tongji University, Shanghai 201804, China.
| |
Collapse
|
30
|
Wong CHY, Jenne CN, Kolaczkowska E. Editorial: Community series in intravital microscopy imaging of leukocytes, volume II. Front Immunol 2025; 16:1615392. [PMID: 40416958 PMCID: PMC12098065 DOI: 10.3389/fimmu.2025.1615392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Affiliation(s)
- Connie H. Y. Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Craig N. Jenne
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Elzbieta Kolaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
31
|
Zhou Y, Wang X, Zhang D, Cui H, Tian X, Du W, Yang Z, Wan D, Qiu Z, Liu C, Yang Z, Zhang L, Yang Q, Xu X, Li W, Wang D, Huang H, Wu W. Precision-Guided Stealth Missiles in Biomedicine: Biological Carrier-Mediated Nanomedicine Hitchhiking Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504672. [PMID: 40345158 DOI: 10.1002/advs.202504672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/14/2025] [Indexed: 05/11/2025]
Abstract
Nanodrug delivery systems (NDDS) have demonstrated broad application prospects in disease treatment, prevention, and diagnosis due to several advantages, including functionalization capability, high drug-loading capacity, drug stability protection, and the enhanced permeability and retention (EPR) effect. However, their clinical translation still faces multiple challenges, including rapid clearance by the reticuloendothelial system (RES), poor targeting specificity, and insufficient efficiency in crossing biological barriers. To address these limitations, researchers have developed the biological carrier-mediated nanomedicine hitchhiking strategy (BCM-NHS), which leverages circulating cells, proteins, or bacteria as natural "mobile carriers" to enhance drug delivery. This approach enables nanocarriers to inherit the intrinsic biological properties, endowing them with immune evasion, prolonged circulation, dynamic targeting, biocompatibility, biodegradability, and naturally optimized biological interfaces. Here, a systematic overview of the BCM-NHS is provided. First, the review delves into the methods of nanoparticles (NPs) binding and immobilization, encompassing both the surface-attachment-mediated "backpack" strategy and the encapsulation-based "Trojan horse" strategy. Second, the classification of biological carriers, including both cell-based and non-cell-based carriers, is elucidated. Third, the physical properties and release mechanisms of these nanomaterials are thoroughly described. Finally, the latest applications of BCM-NHS in therapeutic and diagnostic contexts across various disease models including tumor, ischemic stroke, and pneumonia are highlighted.
Collapse
Affiliation(s)
- Yuyan Zhou
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Xinyue Wang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Deyu Zhang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Hanxiao Cui
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Xiaorong Tian
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wei Du
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhenghui Yang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Dongling Wan
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhiwei Qiu
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Chao Liu
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhicheng Yang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Lizhihong Zhang
- Department of Stomatology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong Province, 519041, China
| | - Qiusheng Yang
- Department of Infectious Diseases, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xuefeng Xu
- Department of Gastroenterology, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Wenhao Li
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| | - Dong Wang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| | - Haojie Huang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| |
Collapse
|
32
|
Wang Q, Zhang X, Han C, Lv Z, Zheng Y, Liu X, Du Z, Liu T, Xue D, Li T, Wang L. Immunodynamic axis of fibroblast-driven neutrophil infiltration in acute pancreatitis: NF-κB-HIF-1α-CXCL1. Cell Mol Biol Lett 2025; 30:57. [PMID: 40335899 PMCID: PMC12060353 DOI: 10.1186/s11658-025-00734-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a sterile inflammation, and 10-20% of cases can progress to severe acute pancreatitis (SAP), which seriously threatens human life and health. Neutrophils and their extracellular traps (NETs) play an important role in the progression of AP. However, the immunodynamic factors between the excessive infiltration of neutrophils during the occurrence of AP have not been fully elucidated. METHODS Adult male C57BL/6 J mice were selected. An AP model was induced by cerulein, and a control group was set up. Single-cell sequencing technology was used to reveal the cell atlas of AP pancreatitis tissue. In vivo, the model mice were treated with anti-Ly6G antibody, DNase I, SC75741, PX-478, and SRT3109 respectively. In vitro, human pancreatic stellate cells were treated with hypoxia, H2O2, NAC, and JSH-2, and co-cultured with neutrophils in Transwell chambers. The severity of inflammation was evaluated, and the molecular mechanism by which fibroblasts exacerbate AP was revealed through techniques such as cell colony formation assay, cell migration assay, cell transfection, immunofluorescence, flow cytometry, Western blot, reverse-transcription quantitative polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (co-IP). RESULTS The study showed that the elimination of neutrophils and NETs could significantly improve AP. Single-cell RNA sequencing (scRNA-seq) indicated that both neutrophils and fibroblasts in pancreatic tissue exhibited heterogeneity during AP. Among them, neutrophils highly expressed CXCR2, and fibroblasts highly expressed CXCL1. Further experimental results demonstrated that the infiltration of neutrophils in the early stage of AP was related to the activation of fibroblasts. The activation of fibroblasts depended on the nuclear factor kappa B (NF-κB) signaling pathway induced by hypoxia. NF-κB enhanced the activation of pancreatic stellate cells (PSCs) and the secretion of CXCL1 by directly promoting the transcription of HIF-1α and indirectly inhibiting PHD2, resulting in the accumulation of HIF-1α protein. The NF-κB-HIF-1α signal promoted the secretion of CXCL1 by fibroblasts through glycolysis and induced the infiltration of neutrophils. Finally, blocking the NF-κB-HIF-1α-CXCL1 signaling axis in vivo reduced the infiltration of neutrophils and improved AP. CONCLUSIONS This study, for the first time, demonstrated that activation of fibroblasts is one of the immunological driving factors for neutrophil infiltration and elucidated that glycolysis driven by the NF-κB-HIF-1α pathway is the intrinsic molecular mechanism by which fibroblasts secrete CXCL1 to chemotactically attract neutrophils. This finding provides a highly promising target for the treatment of AP.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chenglong Han
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenyi Lv
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiwei Du
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianming Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Liyi Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
33
|
Krémer V, Rambault M, Schmutz S, Montcuquet N, Bruhns P, de Chaisemartin L, Jönsson F. Deep phenotyping of human neutrophils in whole blood using a 33-color spectral flow cytometry panel. J Leukoc Biol 2025; 117:qiaf049. [PMID: 40244916 DOI: 10.1093/jleuko/qiaf049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/31/2025] [Accepted: 04/16/2025] [Indexed: 04/19/2025] Open
Abstract
Neutrophils are the most abundant leukocytes in the circulation and critical players in host defense and inflammation. They respond rapidly to numerous biological, chemical, and physical stimuli, making it challenging to characterize their steady-state phenotypes, activation states, and subsets in an unbiased and precise manner. To address this problem, we designed a 33-color spectral flow cytometry panel for the deep profiling of unprocessed neutrophils in human blood. This panel allows the profiling of neutrophil phenotypes related to activation, immune modulation, granule release, ontogeny, phagocytic capacity, and migration, in addition to monitoring all major human leukocyte populations. We validated the panel using whole blood stimulations that induce distinct phenotypic shifts in the neutrophil population. This optimized spectral flow cytometry panel allows comprehensive immune profiling of the functional heterogeneity of human blood neutrophils and is suitable for longitudinal or exploratory analysis of neutrophil dynamics and activation states in clinical cohorts.
Collapse
Affiliation(s)
- Vanessa Krémer
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 25-28 rue du docteur Roux, 75015 Paris, France
| | - Marion Rambault
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 25-28 rue du docteur Roux, 75015 Paris, France
| | - Sandrine Schmutz
- Cytometry Platform, Institut Pasteur, Université Paris Cité, 25-28 rue du docteur Roux, 75015 Paris, France
| | - Nicolas Montcuquet
- Sony Biotechnology Europe, Sony Europe B.V, The Heights, Brooklands, Weybridge, Surrey, KT13 0XW, United Kingdom
| | - Pierre Bruhns
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 25-28 rue du docteur Roux, 75015 Paris, France
| | - Luc de Chaisemartin
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 25-28 rue du docteur Roux, 75015 Paris, France
- Immunology Department, Assistance Publique-Hôpitaux de Paris (APHP), Bichat Hospital, 46 rue Henri Huchard, 75018 Paris, France
- Université Paris Cité, INSERM UMR1149, Centre de Recherche sur l'Inflammation, 16 Rue Henri Huchard, 75018 Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 25-28 rue du docteur Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 3 rue Michel-Ange, 75016 Paris, France
| |
Collapse
|
34
|
Wang KC, Lin CL, Lin CC, Lee YT, Hsu LY, Chien KL, Yeh TL. Association between neutrophil count and the risk of cardiovascular disease: A community-based cohort study in Taiwan. PLoS One 2025; 20:e0322645. [PMID: 40333826 PMCID: PMC12057848 DOI: 10.1371/journal.pone.0322645] [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: 11/04/2024] [Accepted: 03/25/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Neutrophil count is associated with atherosclerotic plaque formation and cardiovascular diseases (CVD). As previous studies have been predominantly conducted in Caucasians, the significance of neutrophil count as a clinical factor in CVD in other ethnicities remain unclear. METHODS A total of 2,955 participants from the Chin-Shan Community Cardiovascular Study(CCCC), who had no established CVD diagnosis and missing data, were enrolled in this study and followed from 1990-1991-2013. We use Cox regression models to calculate hazard ratio (HR) and 95% confidence interval (CI) to evaluate the association between neutrophil count and CVD risk. Subgroup analyses were performed based on sex and age, while sensitivity analyses were conducted by excluding participants with extreme values. RESULTS Over a median follow-up period of 22 years, 400 cases of new-onset CVD were recorded. Cox proportional hazards regression analysis revealed that a higher neutrophil count was independently associated with CVD incidence in Taiwanese adults, with an HR of 1.42 (95% CI 1.03-1.94) after adjusting for multiple covariates. This association remained consistent in both the subgroup and sensitivity analyses. CONCLUSION Our study demonstrated that, in the Taiwanese population, a higher neutrophil count was associated with a higher incidence of CVD over an average 22-year follow-up in individuals without preexisting CVD.
Collapse
Affiliation(s)
- Kuang-Chung Wang
- Department of Family Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Chu-Lin Lin
- Department of General Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chun-Chieh Lin
- Department of General Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yun-Tzu Lee
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan
| | - Le-Yin Hsu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Graduate Program of Data Science, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Population Health Research Center, National Taiwan University, Taipei, Taiwan
| | - Tzu-Lin Yeh
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan
- Department of Family Medicine. Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| |
Collapse
|
35
|
Ma S, Ni J, Ye D, Kuang Y, Wang Z, Yang L. Human umbilical cord mesenchymal stem cells improve the survial of flaps by promoting angiogenesis in mice. Eur J Med Res 2025; 30:356. [PMID: 40312717 PMCID: PMC12046903 DOI: 10.1186/s40001-025-02602-7] [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: 12/01/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Flap necrosis post-operation disturbs surgeons during plastic and reconstructive surgery. This is caused by hypoperfusion and subsequent ischemia-reperfusion injury, where restricted blood flow followed by restored circulation paradoxically exacerbates tissue damage. Mesenchymal stem cells, which show multidirectional differentiation, provide hematopoietic support and are involved in immune regulation and anti-fibrosis, have inspired research on improving the blood supply of flaps. METHODS Primary human umbilical cord mesenchymal stem cells (HuMSCs), were obtained and subcultured for expansion. The cells of the third generation were incubated in a gelatin sponge. Thirty Kunming mice were randomly divided into three groups, and saline, HuMSCs, and HuMSCs-CM were injected preoperatively into the skin of the back. The vessel density was assessed on the tenth day. Forty-eight Kunming mice were divided into two groups. Group A was subdivided into the saline group, HuMSCs, and HuMSCs-CM groups and pretreated as described above. In Group B, the intervention was changed from injection to subcutaneous embedding. Random flaps were made on the back in both groups on the tenth day after pretreatment. The survival rate of the flap was calculated on the seventh day. RESULTS HuMSCs-CM significantly increased the microvessel density on the tenth day after pretreatment. The flap survival rate was higher in the cell and CM groups, rising from approximately 13% to 60% in Group A, and to about 75% in Group B. Moreover, subcutaneous embedding of cell-carrying gelatin sponges improved flap survival compared to other interventions. CONCLUSION Improved cell incubation conditions can enhance its utility. The application of HuMSCs and their conditioned medium promoted the survival of the flap by inducing neovasculogenesis.
Collapse
Affiliation(s)
- Siyi Ma
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jintao Ni
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Danyan Ye
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuping Kuang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhixia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lujun Yang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
36
|
Ou Z, Li L, Ren P, Zhou T, He F, Chen J, Cai H, Han X, Wu Y, Li J, Li X, Tan Q, Li W, Chen Q, Zhang N, He X, Chen W, Zhao Y, Sun J, Zhang Q, Wu Y, Liang Y, You J, Hu G, Tian X, Liao S, Fu B, Chen A, Cai X, Yang H, Wang J, Jin X, Xu X, Jia W, Li J, Yan H. Spatiotemporal Transcriptomic Profiling Reveals the Dynamic Immunological Landscape of Alveolar Echinococcosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405914. [PMID: 39985260 PMCID: PMC12079354 DOI: 10.1002/advs.202405914] [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: 05/29/2024] [Revised: 10/03/2024] [Indexed: 02/24/2025]
Abstract
Alveolar echinococcosis (AE) is caused by the chronic infection of E. multilocularis, whose tumor-like growth can lead to high fatality if improperly treated. The early diagnosis of infection and the treatment of advanced AE remain challenging. Herein, bulk RNA-seq, scRNA-seq, and spatial transcriptomics technologies are integrated, to reveal the host immune response mechanism against E. multilocularis both spatially and chronologically, collecting mouse liver samples at multiple timepoints up to 15 months post infection. These results unveil an unprecedented high-resolution spatial atlas of the E. multilocularis infection foci and the functional roles of neutrophils, Spp1+ macrophages, and fibroblasts during disease progression. The heterogeneity of neutrophil and macrophage subpopulations are critical in both parasite-killing and the occurrence of immunosuppression during AE progression. These findings indicate the transition of parasite control strategy from "active killing" to "negative segregation" by the host, providing instructive insights into the treatment strategy for echinococcosis.
Collapse
Affiliation(s)
- Zhihua Ou
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
| | - Li Li
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Peidi Ren
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
| | - Ting‐Ting Zhou
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Fan He
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Jialing Chen
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Huimin Cai
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- BGI ResearchShenzhen518083China
| | - Xiumin Han
- Qinghai Provincial People's HospitalClinical Research Institute of Hydatid DiseaseXining810007China
| | - Yao‐Dong Wu
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Jiandong Li
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- BGI ResearchShenzhen518083China
| | - Xiu‐Rong Li
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Qiming Tan
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- Laboratory of Bioinformatics and Genome Biology, Faculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityGronostajowa 7Kraków30‐387Poland
| | - Wenhui Li
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Qi Chen
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Nian‐Zhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Xiuju He
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- BGI ResearchShenzhen518083China
| | - Wei‐Gang Chen
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Yanping Zhao
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- BGI ResearchShenzhen518083China
| | - Jiwen Sun
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Qian Zhang
- BGI ResearchBeijing102601China
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yan‐Tao Wu
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Yingan Liang
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- Department of Immunology and MicrobiologyZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Jie You
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Guohai Hu
- China National GeneBankBGI ResearchShenzhen518120China
| | - Xue‐Qi Tian
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | | | - Bao‐Quan Fu
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Ao Chen
- BGI ResearchChongqing401329China
- JFL‐BGI STOmics CenterJinfeng LaboratoryChongqing401329China
| | - Xue‐Peng Cai
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | | | - Jian Wang
- BGI ResearchShenzhen518083China
- China National GeneBankBGI ResearchShenzhen518120China
| | - Xin Jin
- BGI ResearchShenzhen518083China
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- Shenzhen Key Laboratory of Transomics BiotechnologiesBGI ResearchShenzhen518083China
| | - Xun Xu
- BGI ResearchShenzhen518083China
- Guangdong Provincial Key Laboratory of Genome Read and WriteBGI ResearchShenzhen518083China
| | - Wan‐Zhong Jia
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| | - Junhua Li
- Shenzhen Key Laboratory of Unknown Pathogen IdentificationBGI ResearchShenzhen518083China
- BGI ResearchBelgrade11000Serbia
| | - Hong‐Bin Yan
- State Key Laboratory for Animal Disease Control and Prevention/College of Veterinary Medicine, Lanzhou University/Gansu Province Research Center for Basic Disciplines of Pathogen Biology/Key Laboratory of Veterinary Parasitology of Gansu Province/Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs/National Para‐reference Laboratory for Animal Echinococcosis/Lanzhou Veterinary Research InstituteChinese Academy of Agricultural SciencesLanzhou730046China
| |
Collapse
|
37
|
Zhang T, Sun Y, Xia J, Fan H, Shi D, Wu Q, Huang M, Hou XY. Targeting HPK1 inhibits neutrophil responses to mitigate post-stroke lung and cerebral injuries. EMBO Mol Med 2025; 17:1018-1040. [PMID: 40169896 DOI: 10.1038/s44321-025-00220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
Circulating neutrophils are responsible for poor neurological outcomes and have been implicated in respiratory morbidity after acute ischemic stroke (AIS). However, the molecular mechanisms regulating neutrophil responses and their pathological relevance in post-stroke complications remain unclear. In this study, we investigated the involvement of hematopoietic progenitor kinase 1 (HPK1) in neutrophil responses and mobilization, as well as subsequent lung and cerebral injuries following AIS. We found that lipopolysaccharide treatment triggered neutrophil activation in an HPK1-dependent manner. HPK1 enhanced intrinsic NF-κB/STAT3/p38-MAPK pathways and gasdermin D cleavage, leading to neutrophil hyperactivation. Following AIS, HPK1 promoted the mobilization of CXCR2high bone marrow neutrophils. HPK1 loss inhibited peripheral neutrophil hyperactivation, neutrophil infiltration, and aggregation of neutrophil extracellular traps, progressively alleviating systemic inflammation and impairments in mouse pulmonary and neurological functions. Furthermore, HPK1 pharmacological inhibition attenuated post-stroke pulmonary and neurological impairments in mice. Our findings revealed that HPK1 upregulates neutrophil mobilization and various responses, promoting post-stroke systemic inflammation and tissue injury. This study highlights HPK1 as a therapeutic target for improving pulmonary and neurological functions after AIS.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jing Xia
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Hongye Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Dingfang Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Qian Wu
- The Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ming Huang
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Xiao-Yu Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
38
|
Le PM, Pal-Ghosh S, Stepp MA, Menko AS. Shared Phenotypes of Immune Cells Recruited to the Cornea and the Surface of the Lens in Response to Formation of Corneal Erosions. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:960-981. [PMID: 39889825 PMCID: PMC12016862 DOI: 10.1016/j.ajpath.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Injuries to the cornea can lead to recurrent corneal erosions, compromising its barrier function and increasing the risk of infection. Vital as corneal integrity is to the eye's optical power and homeostasis, the immune response to corneal erosions remains poorly understood. It is also unknown whether there is coordinated immune activation between the cornea and other regions of the anterior segment to protect against microbial invasion and limit the spread of inflammation when corneal erosions occur. Herein, a corneal debridement wounding model was used to characterize the immune cell phenotypes populating the cornea in response to erosion formation, and whether and which immune cells are concurrently recruited to the surface of the lens was investigated. The formation of corneal erosions induced an influx of myeloid lineage phenotypes, both M2 macrophages associated with tissue healing and wound repair, and Ly6G+ Ly6C+ myeloperoxidase+ cells resembling neutrophils/polymorphonuclear-myeloid-derived suppressor cells (PMN-MDSCs), with few regulatory T cells, into the corneal stroma under erosion sites. This leukocyte migration into the cornea when erosions develop was paralleled by the recruitment of immune cells, predominantly neutrophils/PMN-MDSCs, to the anterior, cornea-facing lens capsule. Both cornea-infiltrating and lens capsule-associated neutrophil/PMN-MDSC-like immune cells produce the anti-inflammatory cytokine IL-10. These findings suggest a collaborative role for the lens capsule-associated immune cells in preventing infections, controlling inflammation, and maintaining homeostasis of the anterior segment during recurrent corneal erosions.
Collapse
Affiliation(s)
- Phuong M Le
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - A Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
39
|
Varughese A, Balnadupete A, Ramesh P, Prasad TSK, Nidha AB, Bhandary Y. Guardians Turned Culprits: NETosis and Its Influence on Pulmonary Fibrosis Development. Mol Biotechnol 2025; 67:1752-1764. [PMID: 38717537 DOI: 10.1007/s12033-024-01171-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating, life-threatening irreversible lung disease characterized by the excessive accumulation of fibrotic tissue in the lungs, impairing their function. The exact mechanisms underlying Pulmonary fibrosis (PF) are multifaceted and not yet fully understood. Reports show that during COVID-19 pandemic, PF was dramatically increased due to the hyperactivation of the immune system. Neutrophils and macrophages are the patrolling immune cells that keep the microenvironment balanced. Neutrophil extracellular traps (NETs) are a normal protective mechanism of neutrophils. The chief components of the NETs include DNA, citrullinated histones, and anti-microbial peptides which are released by the activated neutrophils. However, it is becoming increasingly evident that hyperactivation of immune cells can also turn into criminals when it comes to pathological state. Dysregulated NETosis may contribute to sustained inflammation, overactivation of fibroblasts, and ultimately promoting collagen deposition which is the characteristic feature of PF. The role of NETs along with inflammation is attaining greater attention. However, seldom researches are related to the relationship between NETs causing PF. This review highlights the cellular mechanism of NETs-induced pulmonary fibrosis, which could give a better understanding of molecular targets which may be helpful for treating NETs-induced PF.
Collapse
Affiliation(s)
- Aleena Varughese
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | - Akarsha Balnadupete
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | - Poornima Ramesh
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | | | | | - Yashodhar Bhandary
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India.
| |
Collapse
|
40
|
Wang S, Zheng H, Zhao J, Xie J. Role of lysine lactylation in neoplastic and inflammatory pulmonary diseases (Review). Int J Mol Med 2025; 55:71. [PMID: 40052587 PMCID: PMC11913435 DOI: 10.3892/ijmm.2025.5512] [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: 12/11/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025] Open
Abstract
Protein lysine lactylation is a ubiquitous and post‑translational modification of lysine residues that involves the addition of a lactyl group on both histone and non‑histone proteins. This process plays a pivotal role in human health and disease and was first discovered in 2019. This epigenetic modification regulates gene transcription from chromatin or directly influences non‑histone proteins by modulating protein‑DNA/protein interactions, activity and stability. The dual functions of lactylation in both histone and non‑histone proteins establish it as a crucial mechanism involved in various cellular processes, such as cell proliferation, differentiation, immune and inflammatory responses and metabolism. Specific enzymes, referred to as 'writers' and 'erasers', catalyze the addition or removal of lactyl groups at designated lysine sites, thereby dynamically modulating lactylation through alterations in their enzymatic activities. The respiratory system has a remarkably intricate metabolic profile. Numerous pulmonary diseases feature an atypical transition towards glycolytic metabolism, which is linked to an overproduction of lactate, a possible substrate for lactylation. However, there has yet to be a comprehensive review elucidating the full impact of lactylation on the onset, progression and potential treatment of neoplastic and inflammatory pulmonary diseases. In the present review, an extensive overview of the discovery of lactylation and advancements in research on the existing lactylation sites were discussed. Furthermore, the review particularly investigated the potential roles and mechanisms of histone and non‑histone lactylation in various neoplastic and inflammatory pulmonary diseases, including non‑small cell lung cancers, malignant pleural effusion, pulmonary fibrosis, acute lung injury and asthma, to excavate the new therapeutic effects of post‑translational modification on various pulmonary diseases.
Collapse
Affiliation(s)
| | | | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
41
|
Stephan JK, Knerr T, Wells CK, Gu Z, Johnson S, Jobe TK, Isaacs WS, Hill BG, Wysoczynski M. G-CSF-Induced Emergency Granulopoiesis Modulates Neutrophil Effector Function in Mice. Stem Cell Rev Rep 2025; 21:1113-1126. [PMID: 40299198 DOI: 10.1007/s12015-025-10885-w] [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] [Accepted: 04/10/2025] [Indexed: 04/30/2025]
Abstract
Neutrophils function as first responders of the immune system by deploying cytotoxic armaments and orchestrating local inflammation. Their functionality is programmed during daily production in the bone marrow through granulopoiesis. During severe inflammation, increased neutrophil demand is met through activation of emergency granulopoiesis. The effect of emergency granulopoiesis on neutrophil functionality remains cryptic. In the present study, we assessed neutrophil function in mice injected with G-CSF (100 µg/kg/d for 3 days) to activate emergency granulopoiesis. We found that emergency granulopoiesis neutrophils exhibit impaired ROS production (n = 6, P = 0.003) and NETosis (n = 5, P < 0.01), but increase neutrophil elastase secretion (n = 9, P < 0.0001) and LPS-induced Tnfa, Il1b, Il1a, Il12a, and Ccl2 expression (n = 13, P < 0.01). To test the impact of emergency granulopoiesis neutrophils on the inflammatory response in vivo, we pre-treated mice with G-CSF and challenged them with zymosan to induce peritonitis. At 4 h post-zymosan injection, peritoneal neutrophils from G-CSF treated mice exhibit increased expression of Ccl2 (n = 3, P < 0.05). Subsequently, we observed enhanced peritoneal macrophage accumulation at 48 h post-zymosan administration in G-CSF-treated mice (n = 5, P < 0.05). These data indicate that emergency granulopoiesis programs neutrophils to have an enhanced immunomodulatory function that orchestrates the subsequent macrophage response in local tissue inflammation.
Collapse
Affiliation(s)
- Jonah K Stephan
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Taylor Knerr
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Collin K Wells
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Zhen Gu
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Sidney Johnson
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Tyler K Jobe
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - William S Isaacs
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Bradford G Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA
| | - Marcin Wysoczynski
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, 580 South Preston Street - Rm 204B, Louisville, KY, USA.
| |
Collapse
|
42
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025; 9:618-637. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [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: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
43
|
Huang Z, Raby RBN, Yin W, Zhang M, Li Z. Development of ROS-responsive collagen-based hemostatic sponges for the repair of MRSA-infected wounds. Int J Biol Macromol 2025; 305:140990. [PMID: 39954906 DOI: 10.1016/j.ijbiomac.2025.140990] [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] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Uncontrolled bleeding and infections, particularly from drug-resistant bacteria like Methicillin-Resistant Staphylococcus aureus (MRSA), pose significant challenges in clinical wound management, delaying healing, increasing patient discomfort, and elevating healthcare costs. This study introduces a novel reactive oxygen species (ROS)-responsive collagen-based hemostatic sponge designed to enhance wound healing and minimize blood loss, especially in MRSA-infected wounds. By chemically modifying the carboxyl groups of collagen with amino-rich oligomers, the primary amino content was increased, enhancing drug loading capacity-particularly for vancomycin-while also improving the sponge's mechanical properties, hemostatic performance, and biological stability. The ROS-responsive covalent bonding of vancomycin facilitated controlled vancomycin release in response to ROS, offering superior antibacterial efficacy and specifically targeting MRSA more effectively than conventional non-ROS-responsive approaches. In MRSA-infected full-thickness skin repair models, the ROS-responsive vancomycin-loaded sponge significantly enhanced wound healing and skin regeneration compared to both the physical adsorption group and the non-ROS-responsive release group. These results underscore the potential of the ROS-responsive collagen composite as an advanced hemostatic material with enhanced antibacterial capabilities, providing rapid hemostasis and improved healing outcomes for complex or infected wounds.
Collapse
Affiliation(s)
- Zhi Huang
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha 410083, China
| | - Randy Bachelard Nziengui Raby
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha 410083, China
| | - Wang Yin
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha 410083, China
| | - Minghua Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Zhexuan Li
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410083, China.
| |
Collapse
|
44
|
Parolini C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem Pharmacol 2025; 235:116844. [PMID: 40044049 DOI: 10.1016/j.bcp.2025.116844] [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: 11/28/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Osteoporosis is the most common skeletal disease worldwide, characterized by low bone mineral density, resulting in weaker bones, and an increased risk of fragility fractures. The maintenance of bone mass relies on the precise balance between bone synthesis and resorption. The close relationship between the immune and skeletal systems, called "osteoimmunology", was coined to identify these overlapping "scientific worlds", and its function resides in the evaluation of the mutual effects of the skeletal and immune systems at the molecular and cellular levels, in both physiological and pathological states. Lipids play an essential role in skeletal metabolism and bone health. Indeed, bone marrow and its skeletal components demand a dramatic amount of daily energy to control hematopoietic turnover, acquire and maintain bone mass, and actively being involved in whole-body metabolism. Statins, the main therapeutic agents in lowering plasma cholesterol levels, are able to promote osteoblastogenesis and inhibit osteoclastogenesis. This review is meant to provide an updated overview of the pathophysiology of bone remodelling cycle, focusing on the interplay between bone, immune system and lipids. Novel therapeutic strategies for the management of osteoporosis are also discussed.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti', via Balzaretti 9 - Università degli Studi di Milano 20133 Milano, Italy.
| |
Collapse
|
45
|
Zvida‐Bloch T, Muchtar E, Dispenzieri A, Shpilberg O, Hershkovitz‐Rokah O. The molecular landscape of AL amyloidosis. Br J Haematol 2025; 206:1297-1311. [PMID: 40211787 PMCID: PMC12078870 DOI: 10.1111/bjh.20070] [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: 01/24/2025] [Accepted: 03/25/2025] [Indexed: 05/16/2025]
Abstract
Amyloid light-chain (AL) amyloidosis is a systemic clonal plasma cell disorder characterized by the production and deposition of misfolded immunoglobulin light chains (LCs), resulting in multiorgan dysfunction. Due to its intricate molecular mechanisms and diverse organ involvement, the disease poses significant diagnostic and therapeutic challenges. This review explores the molecular landscape of AL amyloidosis, emphasizing genetic, transcriptomic and proteomic alterations. Key findings include chromosomal abnormalities, somatic mutations, aberrant gene expression, disrupted protein folding pathways and the role of cytokine and chemokine secretion. These factors collectively drive the overproduction and destabilization of amyloidogenic LCs, leading to organ-specific amyloid deposition, clinical heterogeneity and variable patient outcomes. Despite therapeutic advancements, the disease's complexity challenges the development of effective biological models. Progressing towards personalized therapies requires the development of preclinical models and the identification of biomarkers and molecular data to design targeted interventions. This review highlights the importance of integrating DNA, RNA and protein-level analyses to deepen the understanding of AL amyloidosis pathogenesis. Such insights are pivotal for improving diagnostics, prognostics and therapeutic strategies, ultimately advancing precision medicine for this challenging disease.
Collapse
Affiliation(s)
- Tal Zvida‐Bloch
- Department of Molecular Biology, Faculty of Natural SciencesAriel UniversityArielIsrael
- Translational Research LabAssuta Medical CentersTel‐AvivIsrael
| | - Eli Muchtar
- Division of Hematology, Department of Internal MedicineMayo ClinicRochesterMinnesotaUSA
| | - Angela Dispenzieri
- Division of Hematology, Department of Internal MedicineMayo ClinicRochesterMinnesotaUSA
| | - Ofer Shpilberg
- Adelson School of MedicineAriel UniversityArielIsrael
- Institute of Hematology, Assuta Medical CentersTel‐AvivIsrael
| | - Oshrat Hershkovitz‐Rokah
- Department of Molecular Biology, Faculty of Natural SciencesAriel UniversityArielIsrael
- Translational Research LabAssuta Medical CentersTel‐AvivIsrael
| |
Collapse
|
46
|
Li J, Xuan M, Yang L, Liu Y, Lou N, Fu L, Shi Q, Xue C. Comprehensive single-cell analysis deciphered the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. J Adv Res 2025; 71:457-470. [PMID: 39956402 DOI: 10.1016/j.jare.2025.02.018] [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: 11/05/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025] Open
Abstract
INTRODUCTION Sepsis-related acute liver injury involves complex immune dysfunctions. Epoxyeicosatrienoic acids (EETs), bioactive molecules derived from arachidonic acid (AA) via cytochrome P450 (CYP450) and rapidly hydrolyzed by soluble epoxide hydrolase (sEH), possess anti-inflammatory properties. Nevertheless, the impact of the sEH inhibitor TPPU on sepsis-related acute liver injury remains uncertain. OBJECTIVES This study utilized comprehensive single-cell analysis to investigate the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. METHODS Hepatic bulk RNA sequencing and proteomics analyses were employed to investigate the mechanisms underlying sepsis-related acute liver injury induced by cecal ligation and puncture in mice. Cytometry by time-of-flight and single-cell RNA sequencing were conducted to thoroughly examine the immunoregulatory role of TPPU at single-cell resolution. RESULTS Downregulation of AA metabolism and the CYP450 pathway was observed during sepsis-related acute liver injury, and TPPU treatment reduced inflammatory cytokine production and mitigated sepsis-related hepatic inflammatory injury. Comprehensive single-cell analysis revealed that TPPU promotes the expansion of anti-inflammatory CD206+CD73+ M2-like macrophages and PDL1-CD39-CCR2+ neutrophils, reprogramming liver neutrophils to an anti-inflammatory CAMP+NGP+CD177+ phenotype. Additionally, TPPU inhibits the CCL6-CCR1 signaling mediated by M2-like macrophages and CAMP+NGP+CD177+ neutrophils, altering intercellular communication within the septic liver immune microenvironment. CONCLUSION This study demonstrated TPPU's protective efficacy against sepsis-related acute liver injury, underscoring its vital role in modulating liver macrophages and neutrophils and enhancing prospects for personalized immunomodulatory therapy.
Collapse
Affiliation(s)
- Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingru Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingmiao Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
47
|
Keefe JA, Wang J, Song J, Ni L, Wehrens XHT. Immune cells and arrhythmias. Cardiovasc Res 2025; 121:382-395. [PMID: 39937651 PMCID: PMC12038251 DOI: 10.1093/cvr/cvaf017] [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: 11/06/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 02/14/2025] Open
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide. Emerging evidence has demonstrated that resident and infiltrating cardiac immune cells play direct, mechanistic roles in arrhythmia onset and progression. In this review, we provide a comprehensive summary and expert commentary on the role of each immune cell subtype in the pathogenesis of atrial and ventricular arrhythmias.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, BCM335, One Baylor Plaza, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1095 Jiefang Avenue, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, 1021 Dongmen Rd N, Luohu District, Shenzhen, Guangdong Province, 518001, China
| | - Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1095 Jiefang Avenue, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, BCM335, One Baylor Plaza, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
48
|
Wu D, Yang X, Xu Q, Yao Z, Shan C, Xu H, Ma C. In vitro and in vivo anti-osteoporotic effects of different fractions from Xanthoceras sorbifolium wood. Nat Prod Res 2025:1-7. [PMID: 40296820 DOI: 10.1080/14786419.2025.2497449] [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: 11/29/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
The wood of Xanthoceras sorbifolium (XS) is used in ethnomedicine to treat bone pain. Osteoporosis is among the causes that may result in bone pain. XS was extracted with supercritical CO2 to obtain a fraction CDF and XS residue; The XS residue was extracted by refluxing with ethanol, and the ethanol extract was concentrated to obtain a supernatant fraction (ES) and a precipitate fraction (EP); the major chemical components the fractions were identified by high-performance liquid chromatography-mass spectrometry. ES and EP effectively counteracted the dexa-methasone-induced delay in the calcification of zebrafish and promoted the differentiation of MC3T3-E1 cells. ES upregulated oestrogen receptor 1 and osteocalcin, while EP downregulated myc proto-oncogene, matrix metalloproteinases 9, preproinsulin, and interleukin 6, promoting bone formation. ES and CDF decreased the abnormal aggregation of neutrophils. ES and EP's primary components also exhibited anti-osteoporotic activity, with myricetin demonstrating the strongest anti-osteoporotic effect.
Collapse
Affiliation(s)
- Dandan Wu
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xinyue Yang
- Wuhai Municipal Health Commission, City Administrative Center, Wuhai, China
| | - Qianqian Xu
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zhiwei Yao
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Chengbin Shan
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Haiyan Xu
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Chaomei Ma
- Key Laboratory of Herbage & Endemic Crop Biology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
49
|
Yao Y, Yin Y, Shuai F, Lam W, Zhou T, Xie Y, He X, Han X. M2 Macrophage-Derived Extracellular Vesicles Reprogram Immature Neutrophils into Anxa1 hi Neutrophils to Enhance Inflamed Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416159. [PMID: 40277454 DOI: 10.1002/advs.202416159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/22/2025] [Indexed: 04/26/2025]
Abstract
Periodontitis is a microbiome-related inflammation that can lead to irreversible bone reduction and even tooth loss. This study reveals that macrophage polarization states significantly influence periodontal homeostasis, with M2 macrophage-derived extracellular vesicles (M2-EVs) playing a pivotal role in mitigating periodontitis-induced bone loss. Single-cell RNA sequencing of periodontal tissues treated with M2-EVs uncovered a unique Anxa1hi neutrophil subpopulation exhibiting pro-reparative properties. This subpopulation is characterized by immaturity and demonstrated osteogenic and angiogenic capabilities in vivo, partially mediated through the secretion of oncostatin M (OSM) signals. The findings suggest that this functional heterogeneity arises from M2-EVs disrupting the neutrophil maturation trajectory, with pivotal reprogramming genes, such as Acvrl1 and Fpr2, driving the differentiation of the Anxa1hi reparative subpopulation. This work underscores the potential of targeting M2 macrophage-neutrophil interactions to promote the regeneration of inflamed bone tissues.
Collapse
Affiliation(s)
- Yufei Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yijia Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fangyuan Shuai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Waishan Lam
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tao Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yaxin Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuesong He
- The ADA Forsyth Institute, 100 Chestnut Street, Somerville, MA, 02143, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
50
|
Temnik M, Rudyk M, Balakin A, Gurin S, Dovbynchuk T, Byshovets R, Dzubenko N, Tolstanova G, Skivka L. Anti-inflammatory effects of 64Zn-aspartate is accompanied by cognitive improvements in rats with Aβ 1-40-induced alzheimer disease. Sci Rep 2025; 15:14272. [PMID: 40274975 PMCID: PMC12022080 DOI: 10.1038/s41598-025-97830-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Alzheimer disease (AD) is a debilitating progressive dementia, whose pathophysiology is not fully understood. Chronic inflammation is now widely accepted as one of the key features of AD pathogenesis. Because of this, anti-inflammatory preparations are considered as putative disease modifying agents. A new compound of zinc aspartate with enriched light atoms 64Zn (64Zn-asp) was evaluated as a possible anti-AD agent using Aβ1-40-induced AD model. Intrahippocampal Aβ1-40 injection resulted in pronounced neuroinflammation, as was evidenced by increased phagocytic activity, augmented reactive oxygen species generation, and up-regulated CD86 and CD206 expression by microglia. In rats with Aβ1-40-induced AD, persistent systemic inflammation was also registered, as was ascertained by significantly increased white blood cell-based inflammatory indices and development of anemia of inflammation. Neuro- and systemic inflammation in rats was accompanied by hippocampal dopamine neuron loss, as well as by impairment of short-term and remote spatial memory and cognitive flexibility. Intravenous 64Zn-asp administration rats with AD was associated with returning all microglia indicators to normal range. All aforementioned features of systemic inflammation were not observed in these animals. Anti-inflammatory 64Zn-asp effect was strongly correlated with improvement of short-term spatial memory and cognitive flexibility, and moderately-with betterment of remote spatial memory. These results demonstrated that i.v. 64Zn-asp administration could reverse the inflammatory and, as a result, cognitive effects of intra-hippocampal Aβ1-40 in rats. Therefore, its use may be a viable approach in the complex therapeutic strategy for AD.
Collapse
Affiliation(s)
- Max Temnik
- Physical Chemistry, Vector Vitale, North Miami Beach, USA
| | - Mariia Rudyk
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkova Avenue, Kyiv, 03022, Ukraine.
| | | | - Sergey Gurin
- Physical Chemistry, Vector Vitale, North Miami Beach, USA
| | - Taisa Dovbynchuk
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkova Avenue, Kyiv, 03022, Ukraine
| | - Roman Byshovets
- Department of Internal Diseases, Bogomolets National Medical University, 13, Shevchenko Blvd., Kyiv, 01004, Ukraine
| | - Nataliia Dzubenko
- Educational and Scientific Institute of High Technologies, Taras Shevchenko National University of Kyiv, 4g, Hlushkova Avenue, Kyiv, 03022, Ukraine
| | - Ganna Tolstanova
- Educational and Scientific Institute of High Technologies, Taras Shevchenko National University of Kyiv, 4g, Hlushkova Avenue, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkova Avenue, Kyiv, 03022, Ukraine
| |
Collapse
|