1
|
Emami S, Westerlund E, Rojas Converso T, Johansson-Lindbom B, Persson JJ. Protection acquired upon intraperitoneal group a Streptococcus immunization is independent of concurrent adaptive immune responses but relies on macrophages and IFN-γ. Virulence 2025; 16:2457957. [PMID: 39921669 PMCID: PMC11810095 DOI: 10.1080/21505594.2025.2457957] [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: 08/09/2024] [Revised: 11/09/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Group A Streptococcus (GAS; Streptococcus pyogenes) is an important bacterial pathogen causing over 700 million superficial infections and around 500.000 deaths due to invasive disease or severe post-infection sequelae yearly. In spite of this major impact on society, there is currently no vaccine available against this bacterium. GAS strains can be separated into >250 distinct emm (M)-types, and protective immunity against GAS is believed to in part be dependent on type-specific antibodies. Here, we analyse the nature of protective immunity generated against GAS in a model of intraperitoneal immunization in mice. We demonstrate that multiple immunizations are required for the ability to survive a subsequent lethal challenge, and although significant levels of GAS-specific antibodies are produced, these are redundant for protection. Instead, our data show that the immunization-dependent protection in this model is induced in the absence of B and T cells and is accompanied by the induction of an altered acute cytokine profile upon subsequent infection, noticeable e.g. by the absence of classical pro-inflammatory cytokines and increased IFN-γ production. Further, the ability of immunized mice to survive a lethal infection is dependent on macrophages and the macrophage-activating cytokine IFN-γ. To our knowledge these findings are the first to suggest that GAS may have the ability to induce forms of trained innate immunity. Taken together, the current study proposes a novel role for the innate immune system in response to GAS infections that potentially could be leveraged for future development of effective vaccines.
Collapse
Affiliation(s)
- Shiva Emami
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elsa Westerlund
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Jenny J Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
2
|
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
|
3
|
Guillemaud M, Chavez M, Kobeissy F, Vezzani A, Jimenez AD, Basha MM, Batra A, Demeret S, Eka O, Eschbach K, Foreman B, Gaspard N, Gerard EE, Gofton TE, Haider HA, Hantus ST, Howe CL, Jongeling A, Kalkach-Aparicio M, Kandula P, Kazazian K, Kim M, Lai YC, Marois C, Mellor A, Mohamed W, Morales M, Pimentel CM, Ramirez AM, Steriade C, Struck AF, Taraschenko O, Torcida Sedano N, Wainwright MS, Yoo JY, Wang KKW, Navarro V, Hirsch LJ, Hanin A. Identification of Distinct Biological Groups of Patients With Cryptogenic NORSE via Inflammatory Profiling. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200403. [PMID: 40334176 PMCID: PMC12063244 DOI: 10.1212/nxi.0000000000200403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/11/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND AND OBJECTIVES Emerging evidence suggests that immune dysregulation plays a pivotal role in triggering cryptogenic new-onset refractory status epilepticus (c-NORSE), prompting a consensus on early initiation of immunotherapy. However, despite similar timing of administration, responses to immunotherapies have been varied and unpredictable, suggesting the presence of heterogeneous underlying mechanisms. The aim of this study was to identify distinct inflammatory response subtypes in patients with c-NORSE by analyzing their cytokine profiles. Insights into underlying mechanisms were sought to understand the pathophysiology and guide personalized therapies to improve patient outcomes. METHODS Sixty-two patients with c-NORSE were included. A comprehensive panel of 96 cytokines was analyzed in serum samples. Patients were clustered based on their cytokine profiles using the Louvain algorithm, an unsupervised graph-based clustering method. The identified clusters of patients were compared regarding cytokine levels and clinical features. Protein pathway analysis was used to explore the biological relevance of the inflammatory markers within each cluster. Patients with c-NORSE were compared with control patients (n = 18) and patients with other forms of refractory SE (n = 45). RESULTS Compared with controls, patients with c-NORSE exhibited significant differences in 33 cytokines. Pathway analysis revealed dysregulations in chemotaxis and neutrophil recruitment and migration, highlighting the importance of innate immunity in patients with c-NORSE. Within the c-NORSE cohort, 3 clusters of patients emerged: cluster A, lacking specific inflammatory markers; cluster B, with a much stronger innate-immunity cytokine-driven inflammatory response compared with clusters A and C; and cluster C, defined by dysregulated autoimmune processes. Notably, patients in cluster B showed a statistically significant elevation of innate immune-related proinflammatory cytokines associated with leukocyte recruitment and degranulation. By contrast, those in cluster C showed activation of Janus kinase signal transducer and activator of transcription (JAK-STAT) pathways, suggesting autoimmune mechanisms. Patients in clusters B and C demonstrated varied responses to immunotherapies, with cluster C patients showing favorable outcomes after multiple immunotherapies. DISCUSSION The identification of distinct inflammatory subgroups in c-NORSE suggests that variations in the underlying immune mechanisms contribute to differential treatment responses. These findings underscore the importance of personalized therapeutic strategies, potentially targeting specific inflammatory pathways, to optimize clinical outcomes in this challenging condition.
Collapse
Affiliation(s)
- Martin Guillemaud
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, INRIA, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, France
| | - Mario Chavez
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, INRIA, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, France
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, GA
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research, IRCCS, Milan, Italy
| | - Anthony D Jimenez
- Department of Neurology, Comprehensive Epilepsy Center, Yale University School of Medicine, New Haven, CT
| | | | - Ayush Batra
- Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Sophie Demeret
- Neuro-Intensive Care Unit, AP-HP, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Onome Eka
- Icahn School of Medicine at Mount Sinai, New York City
| | - Krista Eschbach
- Department of Pediatrics, Section of Neurology, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| | - Brandon Foreman
- Division of Neurocritical Care, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH
| | - Nicolas Gaspard
- Department of Neurology, Comprehensive Epilepsy Center, Yale University School of Medicine, New Haven, CT
- Department of Neurology, Hôpital Universitaire de Bruxelles-Hôpital Erasme, Université Libre de Bruxelles, Belgium
| | | | - Teneille Emma Gofton
- Schulich School of Medicine and Dentistry, Western University, London Health Sciences Center, Ontario, Canada
| | - Hiba A Haider
- Department of Neurology, University of Chicago, IL
- Epilepsy Center, Emory University School of Medicine, Atlanta, GA
| | | | | | - Amy Jongeling
- Department of Neurology, NYU Langone Medical Center, New York City
| | | | - Padmaja Kandula
- Weill Cornell Medicine, Department of Neurology, New York City
| | - Karnig Kazazian
- Schulich School of Medicine and Dentistry, Western University, London Health Sciences Center, Ontario, Canada
| | - Minjee Kim
- Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Yi-Chen Lai
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Clémence Marois
- Neuro-Intensive Care Unit, AP-HP, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Andrew Mellor
- Department of Pediatrics, Section of Neurology, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| | - Wazim Mohamed
- Wayne State University School of Medicine, Detroit, MI
| | - Mikaela Morales
- Divison of Pediatric Neurology, Seattle Children's Hospital, University of Washington, Seattle
| | | | - Alexandra M Ramirez
- Division of Neurocritical Care, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH
| | - Claude Steriade
- Department of Neurology, NYU Langone Medical Center, New York City
| | - Aaron F Struck
- University of Wisconsin, Department of Neurology, Madison, WI
| | - Olga Taraschenko
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE
| | - Nathan Torcida Sedano
- Department of Neurology, Hôpital Universitaire de Bruxelles-Hôpital Erasme, Université Libre de Bruxelles, Belgium
| | - Mark S Wainwright
- Divison of Pediatric Neurology, Seattle Children's Hospital, University of Washington, Seattle
| | - Ji Yeoun Yoo
- Icahn School of Medicine at Mount Sinai, New York City
| | - Kevin K W Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, GA
| | - Vincent Navarro
- Epilepsy Unit and Department of Clinical Neurophysiology, AP-HP, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, ERN-Epicare, Paris, France
| | - Lawrence J Hirsch
- Department of Neurology, Comprehensive Epilepsy Center, Yale University School of Medicine, New Haven, CT
| | - Aurélie Hanin
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, INRIA, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, France
- Department of Neurology, Comprehensive Epilepsy Center, Yale University School of Medicine, New Haven, CT
- Department of Metabolic Biochemistry, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; and
- Universite Paris Cité, Faculté de Pharmacie, France
| |
Collapse
|
4
|
Gao H, Sun F, Zhang X, Qiao X, Guo Y. The role and application of Coronin family in human tumorigenesis and immunomodulation. Biochim Biophys Acta Rev Cancer 2025; 1880:189304. [PMID: 40154644 DOI: 10.1016/j.bbcan.2025.189304] [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: 10/16/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The Coronin family, a class of actin-binding proteins involved in the formation and maintenance of cytoskeleton structural stability, is aberrantly expressed in various tumors, including lung, gastric and head and neck cancers. They can regulate tumor cell metabolism and proliferation through RAC-1 and Wnt/β-Catenin signaling pathways and regulate invasion by influencing the PI3K, PAK4, and MT1-MMP signaling pathways and impacting the actin-network dynamics. In recent years, an increasing number of studies have highlighted the crucial roles of the cytoskeleton and immune modulation in the occurrence and development of tumors. The article delves into the Coronin family's pivotal role in tumor immune evasion, highlighting its modulation of neutrophil, T cell, and vesicular transport functions, as well as its interactions with tumorigenesis related organelles such as the endoplasmic reticulum, Golgi apparatus, mitochondria, and lysosomes. It also summarizes the potential therapeutic applications of the Coronin family in oncology. This review provides valuable insights into the mechanisms through which the Coronin family is implicated in the onset and progression of tumors. It also provides more theoretical foundation for tumor immunotherapy and combination drug therapy.
Collapse
Affiliation(s)
- Huimeng Gao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Fuli Sun
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Diseases, Shenyang, Liaoning 110002, China
| | - Xuanyu Zhang
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Xue Qiao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| | - Yan Guo
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| |
Collapse
|
5
|
Roy M, Bagchi A, Karmakar C, Chatterjee M. Transdifferentiation of neutrophils facilitates the establishment of infection by Leishmania donovani parasites. Infect Immun 2025; 93:e0040924. [PMID: 40340446 DOI: 10.1128/iai.00409-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/10/2025] Open
Abstract
Neutrophil transdifferentiation involves the acquisition of dendritic cell-like properties, challenging the traditional view of neutrophils being solely phagocytes. The presence of transdifferentiated neutrophils is established in Visceral Leishmaniasis, but not in its dermal sequel, Post Kala-azar Dermal Leishmaniasis. Accordingly, this study investigated the altered functionalities of neutrophils focusing on the acquisition of dendritic cell-like properties and its impact on infection establishment. In PKDL cases, immunophenotyping of neutrophil-dendritic cells (N-DC hybrids) was performed using flow cytometry, along with studying the status of N-DC hybrid inducing cytokines (TNF-α, IFN-γ) and growth factor (GM-CSF). Ex vivo infection of neutrophils with L. donovani was monitored by droplet digital PCR, employing A2; additionally, their frequency of transdifferentiation, oxidative and phagocytic status, as well as apoptosis potential were quantified by flow cytometry. Compared with healthy controls, neutrophils from PKDL cases demonstrated a significant upregulation of CD83 positivity, but the frequency of co-stimulation (HLA-DR, CD80/86) was unaltered. PKDL cases demonstrated raised levels of TNF-α and IFN-γ, but GM-CSF remained unchanged. Following ex vivo infection of neutrophils, infection was evident at 2 h and was accompanied by CD83 positivity. Furthermore, the CD66b+/CD83 vis-à-vis CD66b+/CD83- subset exhibited heightened generation of reactive oxygen species (ROS), enhanced phagocytosis, and increased apoptosis. Taken together, neutrophils from PKDL cases demonstrated transdifferentiation with the absence of antigen-presenting function. Virulent Leishmania induced transdifferentiation in neutrophils, altering their functionalities and facilitating parasite uptake, along with heightened generation of intra-neutrophilic ROS and enhanced apoptosis, which possibly facilitated their engulfment by macrophages, thereby bolstering the "Trojan horse" mechanism of parasite transfer.
Collapse
Affiliation(s)
- Madhurima Roy
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Aniruddha Bagchi
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Chaitali Karmakar
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
6
|
Forsman H, Li W, Levin NK, Karlsson R, Karlsson A, Dahlgren C, Sundqvist M. Activation and signaling characteristics of the hydroxy-carboxylic acid 3 receptor identified in human neutrophils through a microfluidic flow cell technique. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119950. [PMID: 40194600 DOI: 10.1016/j.bbamcr.2025.119950] [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: 12/17/2024] [Revised: 03/05/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025]
Abstract
Human neutrophils express numerous G protein-coupled receptors (GPCRs) of importance for immune regulation. However, several functionally characterized neutrophil GPCRs, are not included within the human neutrophil proteome. To identify GPCRs not previously demonstrated to be expressed in human neutrophils, we utilized a microfluidic flow cell technique in conjunction with subcellular granule fractionation and liquid chromatography-tandem mass spectrometry (LC-MS/MS). This approach led to the identification of hydroxy-carboxylic acid 3 receptor (HCA3R, also known as GPR109B) as a novel component of the human neutrophil proteome. The β-oxidation intermediate 3-hydroxy-octanoic acid (3-OH-C8) is the primary endogenous agonist of the HCA3R and expressed at high levels in adipocytes where it exerts anti-lipolytic effects. However, literature describing the role and function of HCA3R in human neutrophils is scarce. We show that 3-OH-C8, as well as the synthetic HCA3R agonist IBC 293, activate human neutrophils determined as an increase in the intracellular concentration of free calcium ions ([Ca2+]i) and activation of the NADPH oxidase. However, in contrast to the rise in [Ca2+]i, which could be triggered in naïve neutrophils, pre-treatment of neutrophils was required for the HCA3R agonists to activate the NADPH oxidase. That is, the HCA3R-mediated NADPH oxidase activation occurred only in neutrophils pre-treated with either an actin cytoskeleton disrupter or an allosteric modulator targeting the GPCR termed free fatty acid receptor 2 (FFA2R). Our findings demonstrate that HCA3R is not only a new member of the human neutrophil proteome but also exhibits functional activity with complex signaling pathways when stimulated with endogenous and synthetic HCA3R agonists.
Collapse
Affiliation(s)
- Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Wenyan Li
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Neele K Levin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | | | - Claes Dahlgren
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
7
|
Wang C, Huang M, Guo B, Zhou X, Cui Z, Xu Y, Ren Y. Severe Enterovirus A71 infection is associated with dysfunction of T cell immune response and alleviated by Astragaloside A. Virol Sin 2025:S1995-820X(25)00068-9. [PMID: 40449890 DOI: 10.1016/j.virs.2025.05.010] [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: 01/25/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025] Open
Abstract
Enterovirus A71 (EV-A71) is the major causative pathogen for severe hand-foot-mouth disease (HFMD), a predominantly childhood-associated communicable disease. The mechanisms that children manifest severe disease progression while adults typically exhibit milder or asymptomatic infections remain incompletely characterized, which hinders the development of effective therapy against this disease. Herein, using the newborn mouse model of EV-A71 infection, we uncovered that the underdevelopment of T cells closely associated with the severity of EV-A71 infection, and EV-A71 infection dramatically impaired T-cell immune response. Moreover, the dysfunction of T-cell immunity contributes to the pathogenesis of EV-A71 infection, as the loss of T cells made neonatal mice highly vulnerable to EV-A71 infection. To further assess the relationship between T-cell immunity and HFMD, we enrolled a cohort of 145 pediatric patients with laboratory-confirmed EV-A71 infection and found that the compromised T-cell immune response is associated with the severity of EV-A71-caused HFMD in these children. Furthermore, we found that the treatment of newborn mice with Astragaloside A, a saponin from the medicinal herb Astragalus membranaceus, showed potent in vivo therapeutic efficacy against EV-A71 infection in a T-cell-dependent manner. In conclusion, these findings uncover the interaction between EV-A71 infection and T-cell immunity, provide novel insights onto the physiological impacts of T cells on the pathogenesis of EV-A71 infection and HFMD, and find a promising immunotherapeutic strategy to treat this viral disease.
Collapse
Affiliation(s)
- Chong Wang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Muhan Huang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bingyu Guo
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xi Zhou
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zongqiang Cui
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yi Xu
- Department of Infectious Disease, Women and Children's Medical Center, Affiliated to Guangzhou Medical University, Guangzhou 510623, China.
| | - Yujie Ren
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
8
|
Li Y, Wang H, Zhao X, Deng W, Song C, Wen J, Zhu S, Shen W. Prognostic value of immunotrophic inflammatory markers in ESCC undergoing chemoradiotherapy combined with immunotherapy. Sci Rep 2025; 15:18258. [PMID: 40414935 DOI: 10.1038/s41598-025-02454-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
This study aimed to investigate the role of immunotrophic inflammatory markers in assessing the prognosis and treatment-related toxicity in patients with esophageal squamous cell carcinoma (ESCC) undergoing first-line radiotherapy and chemotherapy combined with immunotherapy. We retrospectively enrolled 67 patients with ESCC and determined the optimal cutoff values for prognostic nutrition index (PNI), neutrophil to lymphocyte ratio (NLR), and platelet to lymphocyte ratio (PLR) using receiver operating characteristic (ROC) curve analysis. Statistical analysis was performed using SPSS 25.0. The one-, two-, and three-year overall survival (OS) rates were 88.1%, 62.3%, and 57.6%, respectively. The overall effective rate was 82.1% (55/67). ROC curve analysis revealed optimal cutoff values for PNI, NLR, and PLR as 48.35, 3.84, and 150.49, respectively. Patients in the high PNI group, low NLR group and PLR group exhibited significantly higher mOS and mPFS time compared to the control group. Notably, the incidence of grade 2 toxicity and side effects in the PNI ≥ 48.35 group was significantly lower than that in the PNI < 48.35 group. Our findings suggest that pretreatment values of PNI, NLR, and PLR can serve as valuable biomarkers for evaluating the prognosis of ESCC patients undergoing first-line radiotherapy and chemotherapy combined with immunotherapy. Further studies with larger cohorts are warranted to validate these results.
Collapse
Affiliation(s)
- Youmei Li
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Hesong Wang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Xiaohan Zhao
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Wenzhao Deng
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Chunyang Song
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Jingyuan Wen
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Shuchai Zhu
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Wenbin Shen
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China.
| |
Collapse
|
9
|
Lucarini E, Schettino A, Marigliano N, Ciampi C, Smimmo M, Romano F, Paolillo A, Izzo L, Begum J, Mansour AA, Iaccarino N, Randazzo A, Greco KV, Scarpa R, Caso F, Iqbal AJ, Bucci M, Ghelardini C, Mannelli LDC, Saviano A, Maione F. Exploring the dual role of Mangifera indica L. in regulating immune response and pain persistence in inflammatory bowel disease. Pharmacol Res 2025:107773. [PMID: 40389041 DOI: 10.1016/j.phrs.2025.107773] [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: 03/19/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/21/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis and Crohn's disease, is characterized by chronic intestinal inflammation and immune dysregulation, driven mainly by Th1 and Th17 cells and sustained by pro-inflammatory cyto-chemokines. This inflammatory milieu is associated with visceral pain, a key symptom affecting patient quality of life. Addressing both gut inflammation/immunity and visceral pain is crucial for improving IBD therapy. This study assessed the therapeutic potential of Mangifera indica L. extract (MIE), a mangiferin-rich formulation, in a DNBS-induced colitis model in rats. MIE treatment administered either simultaneously or post-DNBS induction, significantly reduced pathogenic Th1 and Th17 cell infiltration, along with pro-inflammatory cytokines (IL-1β, TNF-α) and chemokines (CXCL1, CXCL2), though histopathology showed no significant improvements in tissue healing. Additionally, MIE restored microbiota-derived short-chain fatty acids (acetate and butyrate) in colon and faecal samples. Importantly, MIE alleviated post-inflammatory visceral hypersensitivity, reducing the abdominal withdrawal reflex (AWR) to colorectal distension (CRD), after either acute or repeated treatment. These findings suggest that MIE, in the context of nutraceuticals and functional foods, shows promise as a dual-action therapeutic strategy for complementary and/or adjuvant therapy in IBD.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Anna Schettino
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Martina Smimmo
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesca Romano
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Antonio Paolillo
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Jenefa Begum
- Department of Cardiovascular Sciences, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia
| | - Nunzia Iaccarino
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Karin Vicente Greco
- University College London (UCL), Division of Surgery and Interventional Science, Royal Free Hospital Campus, UK; Department of Engineering of Materials and of Bioprocesses, School of Chemical Engineering, University of Campinas (UNICAMP), Av. Albert Einstein 500, CEP 13083-852 Campinas, SP, Brazil
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Asif Jilani Iqbal
- Department of Cardiovascular Sciences, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
| | - Mariarosaria Bucci
- Department of Pharmacy, University of Naples Federico II, School of Medicine and Surgery, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy; Nutraceuticals and Functional Foods Task Force, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
10
|
Amer H, Flanagan KL, Kampan NC, Itsiopoulos C, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Is a Crucial Factor in Shaping the Inflammatory Tumor Microenvironment in Ovarian Cancer and Determining Its Hot or Cold Nature with Diagnostic and Prognostic Utilities. Cancers (Basel) 2025; 17:1691. [PMID: 40427188 PMCID: PMC12109964 DOI: 10.3390/cancers17101691] [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: 03/17/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Ovarian cancer (OC) remains the leading cause of cancer-related deaths among women, often diagnosed at advanced stages due to the lack of effective early diagnostic procedures. To reduce the high mortality rates in OC, reliable biomarkers are urgently needed, especially to detect OC at its earliest stage, predict specific drug responses, and monitor patients. The cytokine interleukin-6 (IL6) is associated with low survival rates, treatment resistance, and recurrence. In this review, we summarize the role of IL6 in inflammation and how IL6 contributes to ovarian tumorigenesis within the tumor microenvironment, influencing whether the tumor is subsequently classified as "hot" or "cold". We further dissect the molecular and cellular mechanisms through which IL6 production and downstream signaling are regulated, to enhance our understanding of its involvement in OC development, as well as OC resistance to treatment. We highlight the potential of IL6 to be used as a reliable diagnostic biomarker to help detect OC at its earliest stage, and as a part of predictive and prognostic signatures to improve OC management. We further discuss ways to leverage artificial intelligence and machine learning to integrate IL6 into diverse biomarker-based strategies.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
- School of Medicine and Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | | | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| |
Collapse
|
11
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
Cao M, Pan C, Zhao Z, Ye S, Bai L, Zhang T. Novel immune-related prognostic models for patients with hepatocellular carcinoma after curative resection. Hepatol Int 2025:10.1007/s12072-025-10839-x. [PMID: 40374839 DOI: 10.1007/s12072-025-10839-x] [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: 01/15/2025] [Accepted: 04/27/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND The patterns of postoperative recurrence vary among hepatocellular carcinoma (HCC) patients and infiltration of immune cells is correlated with patients prognosis. The present study aimed to develop and assess novel nomogram models for postsurgical recurrence and survival in HCC patients by combination of immune cell scores and clinicopathological features. METHODS A total of 233 patients with curative hepatic resection and complete clinicopathologic information were enrolled. The infiltration of CD8 + T lymphocytes, CD15 + neutrophils and CD68 + macrophages in the tumor microenvironment was assessed by immunohistochemistry in tissue microarray. Two prognostic nomogram models for disease-free survival (DFS) and overall survival (OS) were developed and multi-dimensionally evaluated to predict postsurgical HCC outcomes. RESULTS The DFS nomogram was developed using AFP, GGT, tumor differentiation, Ki-67, and the densities of intratumoral CD15 + neutrophils and CD68 + macrophages. The OS nomogram was established based on gender, AFP, tumor differentiation, number of tumor nodules, microvascular vascular tumor thrombus (MVTT), Ki-67, microvessel density (MVD), the densities of intratumoral CD15 + neutrophils and CD68 + macrophages. The C-indexes for the DFS and OS nomogram were 0.708 (95% CI, 0.675-0.741) and 0.723 (95% CI, 0.688 to 0.758), respectively. The AUC values of the models for 1-, 2- or 5-year DFS were 0.832, 0.807 and 0.783, and for 1-, 2- or 5-year OS were 0.745, 0.794 and 0.842. CONCLUSION The present study proposed two nomogram models integrating infiltrating immune cells with clinicopathological risks and showed relatively good predictive performance of recurrence and survival, which may be beneficial to the clinical practice of HCC stratification. Further multicenter studies are needed to assess its general applicability.
Collapse
Affiliation(s)
- Mingnan Cao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Chen Pan
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Sisi Ye
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China
| | - Li Bai
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China.
| | - Tingting Zhang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China.
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University &National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
13
|
Blair KM, Bohinc DJ, Bane KL, Warnock M, Abuaita B, Gura C, Grinsztejn E, Marshall SH, Wilson BM, Bonomo RA, Tambralli A, Knight JS, O'Riordan MX, Lawrence DA, Stavrou EX, Sandkvist M. Acinetobacter Baumannii Secreted Protease CpaA Inhibits Factor XII-Mediated Bradykinin Generation and Neutrophil Activation. Circ Res 2025. [PMID: 40357548 DOI: 10.1161/circresaha.124.324764] [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: 04/19/2024] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND FXII (coagulation factor XII) is best known for its roles in the contact and kallikrein-kinin pathways. FXII is converted to its active enzyme (FXIIa [activated factor XII]) by PKa (plasma kallikrein) or its unique ability to autoactivate on bacterial or other biologic surfaces. In vivo, FXIIa initiates the intrinsic coagulation pathway and promotes inflammation by reciprocal activation of prekallikrein, which cleaves HK (high-molecular-weight kininogen) to liberate bradykinin. CpaA (coagulation targeting metallo-endopeptidase of A baumannii) is a secreted metalloprotease identified in a human clinical isolate of Acinetobacter baumannii that cleaves FXII at O-linked glycosylated sites, inhibiting contact activation. While CpaA facilitates a modest in vivo fitness advantage in mice, the role of CpaA in human infection remains unclear. As such, the objectives of this study were to characterize the structural details of the interaction between CpaA, FXII, and the KKSs (kallikrein-kinin systems) and to determine the downstream consequences on thromboinflammatory responses. METHODS The effect of purified CpaA on the coagulant activity of FXII and the generation of bradykinin was characterized. Neutrophil signaling, flow cytometry, and functional assays were performed to define how CpaA-mediated cleavage of FXII affects innate immune functions. Bacterial killing by human neutrophils was performed with wild-type and mutant A baumannii strains lacking CpaA. RESULTS We found that CpaA cleaves both FXII zymogen and FXIIa but not beta Factor XII. However, cleavage of FXIIa by CpaA does not significantly inhibit its clotting activity, demonstrating that CpaA does not inactivate FXIIa, but rather prevents activation of zymogen FXII. CpaA also cleaves HK, resulting in reduced kallikrein activation and bradykinin generation. We previously identified that zymogen FXII interacts with the urokinase receptor on neutrophils and upregulates neutrophil activation. Here, we demonstrate that CpaA cleaves neutrophil FXII, resulting in reduced Akt2 phosphorylation, chemotaxis, oxidative burst, and neutrophil extracellular trap formation. Importantly, CpaA decreases the human neutrophil killing efficiency of A baumannii in culture. CONCLUSIONS These data identify a role for FXII in responding to bacterial infection and suggest that by inhibiting the contact and KKSs and impairing neutrophil activation, CpaA may blunt the innate immune response and help prevent the elimination of A baumannii from the human host.
Collapse
Affiliation(s)
- Kris M Blair
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Now with Fred Hutch/University of Washington/Seattle Children's Cancer Consortium (K.M.B.)
| | - Dillon J Bohinc
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Kara L Bane
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Mark Warnock
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Basel Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge (B.A.)
| | - Colby Gura
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Eduarda Grinsztejn
- Hematology and Oncology Division, Department of Medicine, University Hospitals Cleveland Medical Center, OH (E.G.)
| | - Steven H Marshall
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Brigid M Wilson
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Robert A Bonomo
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
- Clinician Scientist Investigator, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH.(R.A.B.)
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), OH (R.A.B.)
| | - Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Evi X Stavrou
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
- Medicine Service, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH (E.X.S.)
| | - Maria Sandkvist
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| |
Collapse
|
14
|
Chen B, Fan H, Pang X, Shen Z, Gao R, Wang H, Yu Z, Li T, Li M, Tang Y, Liang X. Single-cell and spatial transcriptomics reveals an anti-tumor neutrophil subgroup in microwave thermochemotherapy-treated lip cancer. Int J Oral Sci 2025; 17:40. [PMID: 40360503 PMCID: PMC12075663 DOI: 10.1038/s41368-025-00366-8] [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: 03/03/2024] [Revised: 12/29/2024] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
Microwave thermochemotherapy (MTC) has been applied to treat lip squamous cell carcinoma (LSCC), but a deeper understanding of its therapeutic mechanisms and molecular biology is needed. To address this, we used single-cell transcriptomics (scRNA-seq) and spatial transcriptomics (ST) to highlight the pivotal role of tumor-associated neutrophils (TANs) among tumor-infiltrating immune cells and their therapeutic response to MTC. MNDA+ TANs with anti-tumor activity (N1-phenotype) are found to be abundantly infiltrated by MTC with benefit of increased blood perfusion, and these TANs are characterized by enhanced cytotoxicity, ameliorated hypoxia, and upregulated IL1B, activating T&NK cells and fibroblasts via IL1B-IL1R. In this highly anti-tumor immunogenic and hypoxia-reversed microenvironment under MTC, fibroblasts accumulated in the tumor front (TF) can recruit N1-TANs via CXCL2-CXCR2 and clear N2-TANs (pro-tumor phenotype) via CXCL12-CXCR4, which results in the aggregation of N1-TANs and extracellular matrix (ECM) deposition. In addition, we construct an N1-TANs marker, MX2, which positively correlates with better prognosis in LSCC patients, and employ deep learning techniques to predict expression of MX2 from hematoxylin-eosin (H&E)-stained images so as to conveniently guide decision making in clinical practice. Collectively, our findings demonstrate that the N1-TANs/fibroblasts defense wall formed in response to MTC effectively combat LSCC.
Collapse
Affiliation(s)
- Bingjun Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huayang Fan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zeliang Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Gao
- University of Electronic Science and Technology of China, Chengdu, China
| | - Haofan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenwei Yu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianjiao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaling Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xinhua Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Teng Y, Wu J, Cai X, Zhang W, Jiang K, Zhou H, Guo Z, Liu J, Wang Y, Liu F, Lan S, Meng H, Ji X, Xiang M, Li Y, Wu D. Poor prognosis of early-stage acral melanoma with a history of trauma: a multicenter analysis of 468 patients. Oncologist 2025; 30:oyaf086. [PMID: 40349138 PMCID: PMC12065939 DOI: 10.1093/oncolo/oyaf086] [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: 08/28/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Previous studies have suggested that trauma may be a risk factor for the development and prognosis of acral melanoma (AM). METHODS This population-based retrospective cohort study included patients with AM and a confirmed history of trauma who received treatment at 5 melanoma treatment centers in China. Factors associated with recurrence and survival were analyzed using univariate and multivariate Cox regression analyses. RESULTS Totally 468 AM cases were included in this study, with 101 patients in the trauma group and 367 in the non-trauma group. The trauma group had more patients with ulceration (P = .027), mitotic rate ≥ 1 (P = .036), and Clark level IV-V (P = .009) than the non-trauma group. Among stage I-II postoperative AM patients, the median recurrence-free survival (RFS) was 33.3 months (95% CI: 18.8-47.8) and 115.6 months (95% CI: 96.3-135.0) in the trauma and non-trauma groups, respectively (P < .001). Similarly, the median overall survival (OS) was 64.6 (95% CI: 54.8-74.4) and not reached (95% CI: NR), respectively (P = .002). Comparatively, no significant differences were observed in RFS or OS between the trauma and non-trauma groups in patients with stage III and IV AM. Multifactorial analysis showed that trauma was an independent risk factor for RFS and OS only in patients with stage I-II AM patients. CONCLUSIONS Postoperative stage I-II patients in the trauma group had significantly worse RFS and OS compared to those in the non-trauma group.
Collapse
Affiliation(s)
- Yi Teng
- Department of Oncology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jin Wu
- Department of Head and Neck Genitourinary Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Cai
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Weizhen Zhang
- Department of Medical Oncology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Kui Jiang
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hongfeng Zhou
- Department of Head and Neck Genitourinary Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhen Guo
- Department of Oncology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yan Wang
- Department of Medical Oncology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Fang Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shijie Lan
- Department of Oncology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiang Ji
- Dalian Medical University, Dalian, China
| | - Mei Xiang
- Department of Medical Oncology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Yongqi Li
- Department of Oncology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Di Wu
- Department of Oncology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Chen Y, Deng Q, Fu T, Huang Y, Li H, Xie J, Liao F, Zeng F, Fang X, Li R, Chen Z. Comprehensive analysis of potential biomarkers for the diagnosis and prognosis of Cervical squamous cell carcinoma - based on GEO and TCGA databases. Front Oncol 2025; 15:1524225. [PMID: 40406253 PMCID: PMC12094959 DOI: 10.3389/fonc.2025.1524225] [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: 11/07/2024] [Accepted: 04/08/2025] [Indexed: 05/26/2025] Open
Abstract
Background Cervical squamous cell carcinoma (CESC) constitutes a substantial global health burden, especially in resource-limited regions. The identification of reliable biomarkers is critical for developing a clinically applicable nomogram to predict survival outcomes and evaluate immune infiltration in CESC patients. Methods This study integrated RNA-seq data from GEO and TCGA databases to identify key genes associated with CESC through differential expression analysis and machine learning techniques. Prognostic models were constructed and validated, with additional analyses exploring immune cell infiltration and gene function via GSEA and clinical correlation. Finally, key genes were validated via qRT-PCR in CESC tissues. Results A total of 112 differentially expressed genes (DEGs) were identified through differential analysis of the GEO and TCGA datasets. EFNA1, CXCL8, and PPP1R14A emerged as prognostic biomarkers for CESC, showing significant associations with survival, tumor stage, and immune infiltration. EFNA1 may drive tumor progression via the MAPK signaling pathway, CXCL8 could influence immune evasion through NOD-like receptor signaling, and PPP1R14A may contribute to tumor invasion by modulating extracellular matrix remodeling. A nomogram integrating these genes demonstrated high predictive accuracy for overall survival (AUC>0.75) and calibration plots. Decision curve analysis (DCA) was performed to assess the nomogram's clinical utility and net benefit for application in clinical practice. Additionally, it was validated by qRT-PCR, showing elevated expression in tumors versus normal tissues (P<0.05). Conclusion EFNA1, CXCL8, and PPP1R14A are promising biomarkers for CESC prognosis and immune regulation. The nomogram model provides a practical tool for personalized survival prediction, enhancing clinical decision-making for immunotherapy and risk stratification.
Collapse
Affiliation(s)
- Yufen Chen
- Department of Obstetrics and Gynecology, The First Affiliate Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Qinghua Deng
- Department of Gynaecology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Tengyue Fu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuxiang Huang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Houlin Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Jingmu Xie
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Feng Liao
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Feimiao Zeng
- Department of Gynaecology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xinyi Fang
- Reproductive Medical Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliate Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhuming Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
He T, Wang ZY, Xu B, Zhong CJ, Wang LN, Shi HC, Yang ZY, Zhou SQ, Li H, Hu B, Zhu XD, Shen YH, Zhou J, Fan J, Sun HC, Huang C. CXCL6 Reshapes Lipid Metabolism and Induces Neutrophil Extracellular Trap Formation in Cholangiocarcinoma Progression and Immunotherapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503009. [PMID: 40305734 DOI: 10.1002/advs.202503009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/31/2025] [Indexed: 05/02/2025]
Abstract
The chemokine CXCL6 is identified as a pivotal regulator of biological processes across multiple malignancies. However, its function in cholangiocarcinoma (CCA) is underexplored. Tumor profiling for CXCL6 is performed using a public database. Both in vitro and in vivo experiments are utilized to evaluate the oncogenic effects of CXCL6 on CCA. Additionally, RNA-Seq is employed to detect transcriptomic changes related to CXCL6 expression in CCA cells and neutrophils. Molecular docking, fluorescence colocalization, and Co-IP are used to elucidate a direct interaction between JAKs and CXCR1/2. Additionally, LC-MS lipidomics and explored the impact of CXCL6 on immunotherapy in vivo. CXCL6 is upregulated in CCA tissues and promoted the proliferation and metastasis of CCA. Mechanistically, CXCL6 regulated the CXCR1/2-JAK-STAT/PI3K axis in CCA via autocrine signaling, leading to lipid metabolic reprogramming, and promoted neutrophil extracellular traps (NETs) formation by activating the RAS/MAPK pathway in neutrophils. Eventually, NETs formation induced immunotherapy resistance in CCA by blocking CD8+T cell infiltration. CXCL6 modulates CCA progression through the CXCR1/2-JAK-STAT/PI3K axis and reshaping its lipid metabolism. CXCL6 also mediates immunotherapy resistance through NETs, which may be a potential therapeutic target and biomarker for CCA.
Collapse
Affiliation(s)
- Tian He
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zi-Yi Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bin Xu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng-Jie Zhong
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lu-Na Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huan-Chen Shi
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zi-Yue Yang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shi-Qi Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hui Li
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bo Hu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Dong Zhu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ying-Hao Shen
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia Fan
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hui-Chuan Sun
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Huang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
18
|
Zhang N, Zhang H, Yu L, Fu Q. Advances in anti-inflammatory treatment of sepsis-associated acute respiratory distress syndrome. Inflamm Res 2025; 74:74. [PMID: 40298991 DOI: 10.1007/s00011-025-02043-2] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Sepsis is characterized by a dysregulated host response to infection, leading to organ dysfunction and associated with significant morbidity and mortality, posing a critical challenge to global public health. Among its complications, sepsis frequently causes acute respiratory distress syndrome (ARDS), which has a high incidence and mortality rate, particularly in intensive care units (ICUs). Currently, the management of sepsis-induced ARDS is largely limited to supportive care, as no specific pharmacological treatments are available. The progression of sepsis to ARDS is driven by severe inflammation and cytokine storms, highlighting the importance of anti-inflammatory therapies as a primary treatment focus. We summarize conventional drugs and emerging treatments targeting excessive inflammatory responses in sepsis-associated ARDS, reviewing progress in basic research and clinical trials. Additionally, we discuss current research challenges to propose future directions for anti-inflammatory treatments, aiming to develop highly effective drugs with better clinical translation potential.
Collapse
Affiliation(s)
- Nana Zhang
- The Fourth Central Clinical School, Tianjin Medical University, 300140, Tianjin, China
| | - Hewei Zhang
- Department of Critical Care Medicine, Tianjin Fourth Central Hospital, 300140, Tianjin, China
| | - Li Yu
- Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Qiang Fu
- The Fourth Central Clinical School, Tianjin Medical University, 300140, Tianjin, China.
| |
Collapse
|
19
|
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
|
20
|
Lamb ER, Glomski IJ, Harper TA, Solga MD, Criss AK. High-dimensional spectral flow cytometry of activation and phagocytosis by peripheral human polymorphonuclear leukocytes. J Leukoc Biol 2025; 117:qiaf025. [PMID: 40036255 PMCID: PMC12031652 DOI: 10.1093/jleuko/qiaf025] [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: 10/31/2024] [Revised: 01/16/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
Polymorphonuclear leukocytes (PMNs) are terminally differentiated phagocytes with pivotal roles in infection, inflammation, tissue injury, and resolution. PMNs display a breadth of responses to diverse endogenous and exogenous stimuli, making understanding of these innate immune responders vital yet challenging to achieve. Here, we report a 22-color spectral flow cytometry panel to profile primary human PMNs for surface marker expression of activation, degranulation, phagocytosis, migration, chemotaxis, and interaction with fluorescently labeled cargo. We demonstrate the surface marker response of PMNs to phorbol ester stimulation compared with untreated controls in an adherent PMN model with additional analysis of intra- and inter-subject variability. PMNs challenged with the Gram-negative bacterial pathogen Neisseria gonorrhoeae revealed infectious dose-dependent changes in surface marker expression in bulk, population-level analysis. Imaging flow cytometry complemented spectral cytometry, demonstrating that fluorescence signal from labeled bacteria corresponded with bacterial burden on a per-cell basis. Spectral flow cytometry subsequently identified surface markers, which varied with direct PMN-bacterium association as well as those which varied in the presence of bacteria but without phagocytosis. This spectral panel protocol highlights best practices for efficient customization and is compatible with downstream approaches such as spectral cell sorting and single-cell RNA-sequencing for applicability to diverse research questions in the field of PMN biology.
Collapse
Affiliation(s)
- Evan R Lamb
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Box 800734, Charlottesville, VA 22908-0734, United States
| | - Ian J Glomski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Box 800734, Charlottesville, VA 22908-0734, United States
| | - Taylor A Harper
- Flow Cytometry Core Facility, University of Virginia School of Medicine, Box 800741, Charlottesville, VA 22908-0741, United States
| | - Michael D Solga
- Flow Cytometry Core Facility, University of Virginia School of Medicine, Box 800741, Charlottesville, VA 22908-0741, United States
| | - Alison K Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Box 800734, Charlottesville, VA 22908-0734, United States
| |
Collapse
|
21
|
Salamanna F, Tedesco G, Sartori M, Contartese D, Asunis E, Cini C, Veronesi F, Martikos K, Fini M, Giavaresi G, Gasbarrini A. Clinical Outcomes and Inflammatory Response to the Enhanced Recovery After Surgery (ERAS) Protocol in Adolescent Idiopathic Scoliosis Surgery: An Observational Study. Int J Mol Sci 2025; 26:3723. [PMID: 40332332 PMCID: PMC12027706 DOI: 10.3390/ijms26083723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Limited data exist on the clinical outcomes and inflammatory response of Enhanced Recovery After Surgery (ERAS) protocols in adolescent idiopathic scoliosis (AIS) patients undergoing posterior spinal fusion (PSF). This study evaluates ERAS's impact by analyzing blood parameters and serum protein, surgical, and radiological parameters to assess safety, feasibility, and clinical outcomes. Between July 2021 and July 2022, 30 AIS patients underwent PSF with standardized pain management, high-dose tranexamic acid, early mobilization, and reduced opioid use. Blood tests and serum markers (IL-6, IL-1β, IL-1α, IL-10, TNF-α, IL-8, PGE2) were measured preoperatively and on postoperative day 2. Pain levels (VAS) were recorded preoperatively, on postoperative days 1 and 2, and at discharge. Results showed increased postoperative white blood cell counts, reduced hemoglobin and hematocrit, and elevated C-reactive protein levels. IL-6 was the only inflammatory marker significantly elevated, indicating a controlled inflammatory response. Pain peaked on day 1 but significantly decreased by discharge, confirming the effectiveness of multimodal analgesia. The average hospital stay was 6.97 ± 2.03 days, with low rehospitalization (6.66%) and manageable complications (20%). In conclusion, ERAS effectively optimizes AIS patient recovery, stabilizing pain, reducing complications, and improving perioperative care.
Collapse
Affiliation(s)
- Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (M.S.); (F.V.); (G.G.)
| | - Giuseppe Tedesco
- Department of Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (G.T.); (E.A.); (C.C.); (K.M.); (A.G.)
| | - Maria Sartori
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (M.S.); (F.V.); (G.G.)
| | - Deyanira Contartese
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (M.S.); (F.V.); (G.G.)
| | - Emanuela Asunis
- Department of Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (G.T.); (E.A.); (C.C.); (K.M.); (A.G.)
| | - Chiara Cini
- Department of Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (G.T.); (E.A.); (C.C.); (K.M.); (A.G.)
| | - Francesca Veronesi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (M.S.); (F.V.); (G.G.)
| | - Konstantinos Martikos
- Department of Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (G.T.); (E.A.); (C.C.); (K.M.); (A.G.)
| | - Milena Fini
- Scientific Direction, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (M.S.); (F.V.); (G.G.)
| | - Alessandro Gasbarrini
- Department of Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136 Bologna, Italy; (G.T.); (E.A.); (C.C.); (K.M.); (A.G.)
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
22
|
Tursi NJ, Tiwari S, Bedanova N, Kannan T, Parzych E, Okba N, Liaw K, Sárközy A, Livingston C, Trullen MI, Gary EN, Vadovics M, Laenger N, Londregan J, Khan MS, Omo-Lamai S, Muramatsu H, Blatney K, Hojecki C, Machado V, Maricic I, Smith TRF, Humeau LM, Patel A, Kossenkov A, Brenner JS, Allman D, Krammer F, Pardi N, Weiner DB. Modulation of lipid nanoparticle-formulated plasmid DNA drives innate immune activation promoting adaptive immunity. Cell Rep Med 2025; 6:102035. [PMID: 40120578 PMCID: PMC12047470 DOI: 10.1016/j.xcrm.2025.102035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/20/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Nucleic acid vaccines have grown in importance over the past several years, with the development of new approaches remaining a focus. We describe a lipid nanoparticle-formulated DNA (DNA-LNP) formulation which induces robust innate and adaptive immunity with similar serological potency to mRNA-LNPs and adjuvanted protein. Using an influenza hemagglutinin (HA)-encoding construct, we show that priming with our HA DNA-LNP demonstrated stimulator of interferon genes (STING)-dependent upregulation and activation of migratory dendritic cell (DC) subpopulations. HA DNA-LNP induced superior antigen-specific CD8+ T cell responses relative to mRNA-LNPs or adjuvanted protein, with memory responses persisting beyond one year. In rabbits immunized with HA DNA-LNP, we observed immune responses comparable or superior to mRNA-LNPs at the same dose. In an additional model, a SARS-CoV-2 spike-encoding DNA-LNP elicited protective efficacy comparable to spike mRNA-LNPs. Our study identifies a platform-specific priming mechanism for DNA-LNPs divergent from mRNA-LNPs or adjuvanted protein, suggesting avenues for this approach in prophylactic and therapeutic vaccine development.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sachchidanand Tiwari
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Bedanova
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Toshitha Kannan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Elizabeth Parzych
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nisreen Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - András Sárközy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cory Livingston
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Maria Ibanez Trullen
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ebony N Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Niklas Laenger
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Biology Department, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer Londregan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Suhail Khan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Serena Omo-Lamai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kerry Blatney
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Casey Hojecki
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Igor Maricic
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | | | | | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew Kossenkov
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
DuBois EM, Herrema KE, Simkulet MG, Hassan LF, O’Connor PR, Sen R, O’Shea TM. Thioether-Functionalized Cellulose for the Fabrication of Oxidation-Responsive Biomaterial Coatings and Films. Adv Healthc Mater 2025; 14:e2403021. [PMID: 39604609 PMCID: PMC12031653 DOI: 10.1002/adhm.202403021] [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: 08/13/2024] [Revised: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Biomaterial coatings and films can prevent premature failure and enhance the performance of chronically implanted medical devices. However, current hydrophilic polymer coatings and films have significant drawbacks, including swelling and delamination. To address these issues, hydroxyethyl cellulose is modified with thioether groups to generate an oxidation-responsive polymer, HECMTP. HECMTP readily dissolves in green solvents and can be fabricated as coatings or films with tunable thicknesses. HECMTP coatings effectively scavenge hydrogen peroxide, resulting in the conversion of thioether groups to sulfoxide groups on the polymer chain. Oxidation-driven, hydrophobic-to-hydrophilic transitions that are isolated to the surface of HECMTP coatings under physiologically relevant conditions increase wettability, decrease stiffness, and reduce protein adsorption to generate a non-fouling interface with minimal coating delamination or swelling. HECMTP can be used in diverse optical applications and permits oxidation-responsive, controlled drug release. HECMTP films are non-resorbable in vivo and evoke minimal foreign body responses. These results highlight the versatility of HECMTP and support its incorporation into chronically implanted medical devices.
Collapse
Affiliation(s)
- Eric M. DuBois
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| | - Kate E. Herrema
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| | - Matthew G. Simkulet
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| | - Laboni F. Hassan
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| | - Payton R. O’Connor
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Riya Sen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Timothy M. O’Shea
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| |
Collapse
|
24
|
Song J, Zhu J, Jiang Y, Guo Y, Liu S, Qiao Y, Du Y, Li J. Advancements in immunotherapy for gastric cancer: Unveiling the potential of immune checkpoint inhibitors and emerging strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189277. [PMID: 39938663 DOI: 10.1016/j.bbcan.2025.189277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Gastric cancer (GC) is linked to high morbidity and mortality rates. Approximately two-thirds of GC patients are diagnosed at an advanced or metastatic stage. Conventional treatments for GC, including surgery, radiotherapy, and chemotherapy, offer limited prognostic improvement. Recently, immunotherapy has gained attention for its promising therapeutic effects in various tumors. Immunotherapy functions by activating and regulating the patient's immune cells to target and eliminate tumor cells, thereby reducing the tumor burden in the body. Among immunotherapies, immune checkpoint inhibitors (ICIs) are the most advanced. ICIs disrupt the inhibitory protein-small molecule (PD-L1, CTLA4, VISTA, TIM-3 and LAG3) interactions produced by immune cells, reactivating these cells to recognize and attack tumor cells. However, adverse reactions and resistance to ICIs hinder their further clinical and experimental development. Therefore, a comprehensive understanding of the advancements in ICIs for GC is crucial. This article discusses the latest developments in clinical trials of ICIs for GC and examines combination therapies involving ICIs (targeted therapy, chemotherapy, radiotherapy), alongside ongoing clinical trials. Additionally, the review investigates the tumor immune microenvironment and its role in non-responsiveness to ICIs, highlighting the function of tumor immune cells in ICI efficacy. Finally, the article explores the prospects and limitations of new immunotherapy-related technologies, such as tumor vaccines, nanotechnologies, and emerging therapeutic strategies, aiming to advance research into personalized and optimized immunotherapy for patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
- Jiawei Song
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China
| | - Jun Zhu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yu Jiang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yajie Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Shuai Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yihuan Qiao
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yongtao Du
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Jipeng Li
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China.
| |
Collapse
|
25
|
Chalmers JD, Metersky M, Aliberti S, Morgan L, Fucile S, Lauterio M, McDonald PP. Neutrophilic inflammation in bronchiectasis. Eur Respir Rev 2025; 34:240179. [PMID: 40174958 PMCID: PMC11962982 DOI: 10.1183/16000617.0179-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/11/2025] [Indexed: 04/04/2025] Open
Abstract
Noncystic fibrosis bronchiectasis, hereafter referred to as bronchiectasis, is a chronic, progressive lung disease that can affect people of all ages. Patients with clinically significant bronchiectasis have chronic cough and sputum production, as well as recurrent respiratory infections, fatigue and impaired health-related quality of life. The pathophysiology of bronchiectasis has been described as a vicious vortex of chronic inflammation, recurring airway infection, impaired mucociliary clearance and progressive lung damage that promotes the development and progression of the disease. This review describes the pivotal role of neutrophil-driven inflammation in the pathogenesis and progression of bronchiectasis. Delayed neutrophil apoptosis and increased necrosis enhance dysregulated inflammation in bronchiectasis and failure to resolve this contributes to chronic, sustained inflammation. The excessive release of neutrophil serine proteases, such as neutrophil elastase, cathepsin G and proteinase 3, promotes a protease-antiprotease imbalance that correlates with increased inflammation in bronchiectasis and contributes to disease progression. While there are currently no licensed therapies to treat bronchiectasis, this review will explore the evolving evidence for neutrophilic inflammation as a novel treatment target with meaningful clinical benefits.
Collapse
Affiliation(s)
- James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Mark Metersky
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - Lucy Morgan
- Department of Respiratory Medicine, Concord Clinical School, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
26
|
Iliakis CS, Crotta S, Wack A. The Interplay Between Innate Immunity and Nonimmune Cells in Lung Damage, Inflammation, and Repair. Annu Rev Immunol 2025; 43:395-422. [PMID: 40036704 DOI: 10.1146/annurev-immunol-082323-031852] [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: 03/06/2025]
Abstract
As the site of gas exchange, the lung is critical for organismal survival. It is also subject to continual environmental insults inflicted by pathogens, particles, and toxins. Sometimes, these insults result in structural damage and the initiation of an innate immune response. Operating in parallel, the immune response aims to eliminate the threat, while the repair process ensures continual physiological function of the lung. The inflammatory response and repair processes are thus inextricably linked in time and space but are often studied in isolation. Here, we review the interplay of innate immune cells and nonimmune cells during lung insult and repair. We highlight how cellular cross talk can fine-tune the circuitry of the immune response, how innate immune cells can facilitate or antagonize proper organ repair, and the prolonged changes to lung immunity and physiology that can result from acute immune responses and repair processes.
Collapse
Affiliation(s)
- Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
27
|
Hou R, Wu X, Wang C, Fan H, Zhang Y, Wu H, Wang H, Ding J, Jiang H, Xu J. Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review). Int J Oncol 2025; 66:28. [PMID: 40017131 PMCID: PMC11900975 DOI: 10.3892/ijo.2025.5734] [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: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Cancer is the second leading cause of death among humans worldwide. Despite remarkable improvements in cancer therapies, drug resistance remains a significant challenge. The tumor microenvironment (TME) is intimately associated with therapeutic resistance. Tumor‑associated neutrophils (TANs) are a crucial component of the TME, which, along with other immune cells, play a role in tumorigenesis, development and metastasis. In the current review, the roles of TANs in the TME, as well as the mechanisms of neutrophil‑mediated resistance to cancer therapy, including immunotherapy, chemotherapy, radiotherapy and targeted therapy, were summarized. Furthermore, strategies for neutrophil therapy were discussed and TANs were explored as potential targets for cancer treatment. In conclusion, the need to explore the precise roles, recruitment pathways and mechanisms of action of TANs was highlighted for the purpose of developing therapies that precisely target TANs and reverse drug resistance.
Collapse
Affiliation(s)
- Rui Hou
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Xi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Cenzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanfang Fan
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Yuhan Zhang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanchi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huiyu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junli Ding
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huning Jiang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junying Xu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| |
Collapse
|
28
|
Ribeiro TF, Soares Ferreira R, Amaral C, Bastos Gonçalves F, Ferreira ME. The Impact of Neutrophil-to-Lymphocyte Ratio on Short- and Long-Term Prognosis Following Elective Infrarenal EVAR. Ann Vasc Surg 2025; 113:195-204. [PMID: 39880285 DOI: 10.1016/j.avsg.2025.01.033] [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: 11/20/2024] [Revised: 01/02/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Neutrophil-to-lymphocyte ratio (NLR) is a readily available parameter, associated with long-term outcomes in cardiovascular conditions. This study aims to analyze the predictors of NLR and its impact on prognosis and disease-specific outcomes following endovascular aneurysm repair (EVAR). METHODS Single-center retrospective cohort study. Consecutive patients who underwent elective EVAR (2011-2023) were considered. Primary outcome is short-term major adverse events (MAE) and long-term mortality. Secondary outcomes were freedom-from EVAR failure, aortic reintervention, and NLR predictors. Multivariable logistic regression analyses were performed for binary outcomes. Survival outcomes were analyzed through Kaplan-Meier and Cox regression analyses. RESULTS Overall, 434 patients were included. A 2.4 NLR cutoff was a fair discriminator for long-term-mortality (area under the curve, 0.62), and groups were dichotomized according to this premise. Increasing age (adjusted odds ratio [aOR]: 1.06; 1.03-1.09, per 1-year increase) and a pulmonary comorbidity (aOR: 1.91; 1.24-2.96) associated to NLR ≥ 2.4. No significant association between comorbidity burden and NLR was observed. MAE occurred more often if NLR ≥ 2.4 (6.2 vs. 11.6% high-NLR, P = 0.049; NLR ≥ 2.4 aOR: 2.10; 1.01-4.36). At 8-years follow-up, survival estimates favored NLR < 2.4 (55.7% vs. 33.7% high-NLR, P < 0.001, NLR ≥ 2.4 adjusted hazard ratio [aHR]: 1.07; 1.05-1.98), without differences in freedom-from EVAR failure (70.6 vs. 68.2% high-NLR, P = 0.27, NLR ≥ 2.4 aHR: 1.26; 0.82-1.94). Conversely, NLR ≥ 2.4 associated with lower freedom from aortic reinterventions (80.0% vs. 70.2% high-NLR, P = 0.01, NLR ≥ 2.4 aHR: 1.80; 1.08-3.01). CONCLUSION NLR appears as a prognostic marker with reduced impact of comorbidity burden. Following EVAR, it independently predicts MAE and mortality. Over time, elevated NLR appears associated with increased aortic reinterventions, although rates and mode of failure seem similar across groups.
Collapse
Affiliation(s)
- Tiago F Ribeiro
- Hospital de Santa Marta, Unidade Local de Saúde São José, Lisboa, Portugal.
| | - Rita Soares Ferreira
- Hospital de Santa Marta, Unidade Local de Saúde São José, Lisboa, Portugal; NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Carlos Amaral
- Hospital de Santa Marta, Unidade Local de Saúde São José, Lisboa, Portugal
| | - Frederico Bastos Gonçalves
- Hospital de Santa Marta, Unidade Local de Saúde São José, Lisboa, Portugal; NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Hospital CUF Tejo, Lisboa, Portugal
| | | |
Collapse
|
29
|
Masset C, Drillaud N, Ternisien C, Degauque N, Gerard N, Bruneau S, Branchereau J, Blancho G, Mesnard B, Brouard S, Giral M, Cantarovich D, Dantal J. The concept of immunothrombosis in pancreas transplantation. Am J Transplant 2025; 25:650-668. [PMID: 39709128 DOI: 10.1016/j.ajt.2024.11.025] [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: 08/09/2024] [Revised: 11/06/2024] [Accepted: 11/23/2024] [Indexed: 12/23/2024]
Abstract
Early failure of a pancreatic allograft due to complete thrombosis has an incidence of approximately 10% and is the main cause of comorbidity in pancreas transplantation. Although several risk factors have been identified, the exact mechanisms leading to this serious complication are still unclear. In this review, we define the roles of the individual components involved during sterile immunothrombosis-namely endothelial cells, platelets, and innate immune cells. Further, we review the published evidence linking the main risk factors for pancreatic thrombosis to cellular activation and vascular modifications. We also explore the unique features of the pancreas itself: the vessel endothelium, specific vascularization, and relationship to other organs-notably the spleen and adipose tissue. Finally, we summarize the therapeutic possibilities for the prevention of pancreatic thrombosis depending on the different mechanisms such as anticoagulation, anti-inflammatory molecules, endothelium protectors, antagonism of damage-associated molecular patterns, and use of machine perfusion.
Collapse
Affiliation(s)
- Christophe Masset
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| | - Nicolas Drillaud
- Laboratory of Hemostasis, Nantes University Hospital, Nantes, France
| | | | - Nicolas Degauque
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Nathalie Gerard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sarah Bruneau
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Julien Branchereau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Gilles Blancho
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Benoit Mesnard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sophie Brouard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Magali Giral
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Diego Cantarovich
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jacques Dantal
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
30
|
Huang H, Li M, Liu Y, Chen Y, Xie Z, Luo M, Li D, Liu H, Xi L. Absence of neutrophils impairs the host defense in murine footpad model of chromoblastomycosis. PLoS Negl Trop Dis 2025; 19:e0012986. [PMID: 40267064 PMCID: PMC12017585 DOI: 10.1371/journal.pntd.0012986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Chromoblastomycosis (CBM), a chronic subcutaneous infection caused by black fungi such as Fonsecaea monophora (F. monophora), is characterized by a low cure rate, high recurrence rate, and prolonged treatment duration. Neutrophils, one of the most important innate immune cells, play complex roles in the prevention of fungal infections. This study investigated the function of neutrophils in host defense against F. monophora using a neutrophil-depleted mouse model and in vitro co-culture conditions. Fungal burden, histopathological changes, and cytokine profiles were compared between neutrophil-depleted mice and isotype control mice. Our findings demonstrated that neutrophil depletion in mice led to impaired fungal clearance, prolonged inflammation in F. monophora infected footpad tissues, highlighting the critical role of neutrophils in controlling F. monophora infection. Histopathological analysis revealed extensive inflammatory cell infiltration, especially macrophages, accompanied by elevated levels of pro-inflammatory cytokines such as IL-1β, CCL3, IL-6, and TNF-α. Besides, we observed that neutrophils play a key role in inhibiting the morphological transition of F. monophora from conidia to hyphae and sclerotic-like cells. Notably, the F. monophora morphology was also associated with the formation of neutrophil extracellular traps (NETs) in in vitro experiment. These findings underscore the importance of neutrophil-mediate immune responses in early fungal clearance and their ability to influence F.monophora morphological transition. The study provides novel insights into the immune mechanisms underlying CBM and highlights the potential therapeutic implications of targeting neutrophil-mediated responses in CBM infections.
Collapse
Affiliation(s)
- Huan Huang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Minying Li
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yinghui Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yangxia Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Zhenmou Xie
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Mingfen Luo
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Dongmei Li
- Department of Microbiology/Immunology, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Hongfang Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Liyan Xi
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
31
|
Fatima M, Al-Keridis LA, Adnan M, Alshammari N, Sulieman AME, Khan MR. Jasminum humile extract mitigates carrageenan-induced paw oedema in rats by modulating inflammatory and antioxidant signalling pathways. Inflammopharmacology 2025; 33:1907-1920. [PMID: 40042724 DOI: 10.1007/s10787-025-01692-3] [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: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Jasminum humile is widely used in traditional medicines to treat hard lumps, mouth inflammation, ringworms, and other infections. Leaf decoction of the plant is known to be effective in treating various skin conditions. In addition, root juice is traditionally utilized as a remedy for ringworm infections. Studies have reported that J. humile contains various antioxidant metabolites with analgesic and anti-inflammatory properties. In this study, J. humile chloroform extract (JHC) was investigated for anti-inflammatory effects against carrageenan-induced paw oedema in rat models. METHODS High-performance liquid chromatography was used to examine phenolic compounds present in JHC. The in-vivo anti-inflammatory activities were investigated using carrageenan-induced paw oedema rat models, while indomethacin was referred to as positive control. Therapeutic properties of JHC were examined by assessing paw volumes, motility score, and inflammatory proteins in serum. The anti-inflammatory nature of JHC was further investigated by biochemical and hematological profiles along with genetic expression of inflammatory and antioxidant genes through qRT-PCR analysis. RESULTS Indomethacin at 10 mg/kg and JHC at 100, 200, and 300 mg/kg doses decreased the concentration of C-reactive protein (CRP) while upregulating the concentration of albumin and myeloperoxidase (MPO). Moreover, JHC administration reduced the expression levels of inflammatory markers, cyclooxygenase-2 (COX2), and inducible nitric oxide synthase (iNOS) compared to the Carr-treated control. However, a significant rise was induced in nuclear factor erythroid 2-related factor 2 (Nrf-2) and heme oxygenase 1 (HO-1) levels after JHC treatment as compared to Carr-treated rats. CONCLUSION These results showed significant anti-inflammatory potential of J. humile by increasing the activity levels of enzymatic antioxidants and lowering inflammatory markers. These results confirm the beneficial use of natural plants in the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Mehreen Fatima
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, P.O. Box 45320, Islamabad, Pakistan.
| | - Lamya Ahmed Al-Keridis
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | | | - Muhammad Rashid Khan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, P.O. Box 45320, Islamabad, Pakistan
| |
Collapse
|
32
|
Wang Z, Jiao Y, Diao W, Shi T, Geng Q, Wen C, Xu J, Deng T, Li X, Zhao L, Gu J, Deng T, Xiao C. Neutrophils: a Central Point of Interaction Between Immune Cells and Nonimmune Cells in Rheumatoid Arthritis. Clin Rev Allergy Immunol 2025; 68:34. [PMID: 40148714 DOI: 10.1007/s12016-025-09044-3] [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] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease involving activation of the immune system and the infiltration of immune cells. As the first immune cells to reach the site of inflammation, neutrophils perform their biological functions by releasing many active substances and forming neutrophil extracellular traps (NETs). The overactivated neutrophils in patients with RA not only directly damage tissues but also, more importantly, interact with various other immune cells and broadly activate innate and adaptive immunity, leading to irreversible joint damage. However, owing to the pivotal role and complex influence of neutrophils in maintaining homoeostasis, the treatment of RA by targeting neutrophils is very difficult. Therefore, a comprehensive understanding of the interaction pathways between neutrophils and various other immune cells is crucial for the development of neutrophils as a new therapeutic target for RA. In this study, the important role of neutrophils in the pathogenesis of RA through their crosstalk with various other immune cells and nonimmune cells is highlighted. The potential of epigenetic modification of neutrophils for exploring the pathogenesis of RA and developing therapeutic approaches is also discussed. In addition, several models for studying cell‒cell interactions are summarized to support further studies of neutrophils in the context of RA.
Collapse
Affiliation(s)
- Zhaoran Wang
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yi Jiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenya Diao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tong Shi
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qishun Geng
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Chaoying Wen
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiahe Xu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Tiantian Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoya Li
- The Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100193, China
| | - Lu Zhao
- China-Japan Friendship Clinical Medical College, Capital Medical University, Beijing, 100029, China
| | - Jienan Gu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tingting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Cheng Xiao
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China.
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
33
|
Liang C, Wang Z, Mai Y, Li J, Dai Q, Yuan Y, Wang M, Liu Y, Zhang W, Li Y, Lu X, Lin Z, Mao T. Mendelian randomization study of circulating leukocytes counts reveals causal associations with inflammatory bowel disease. Medicine (Baltimore) 2025; 104:e41969. [PMID: 40153772 PMCID: PMC11957634 DOI: 10.1097/md.0000000000041969] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/07/2025] [Indexed: 03/30/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent IBD, whose cause involves the interaction between genetic and environmental factors. Although there is a recognized link between immune response and IBD, the causal relationship between circulating immune cell counts and IBD remains controversial. This study aimed to elucidate the causal relationship between genetically predicted circulating immune cell counts and IBD. We conducted a bidirectional 2-sample Mendelian randomization (MR) study using aggregated statistics from genome-wide association studies. The causal relationship between 5 circulating leukocytes cells (monocytes, lymphocytes, eosinophils, basophils and neutrophils) counts and IBD, including ulcerative colitis (UC) and Crohn disease (CD) was analyzed. Horizontal pleiotropy test and heterogeneity test were used to ensure the stability of the results. Our findings indicated that monocytes, lymphocytes, eosinophils, and basophils count were not significantly associated with IBD, however, elevated circulating neutrophils count was significantly associated with higher risk of IBD [odds ratio (OR) = 1.0017; 95% confidence interval (CI) = 1.0004-1.003; P = .009] and UC [OR = 2.465; 95% CI = 1.236-4.916; P = .01]. In addition, we also found that IBD [OR: 12.07; 95% CI = 1.909-76.316; P = .008] and CD [OR = 1.014; 95% CI = 1.004-1.023; P = .005] were significantly associated with higher circulating neutrophils count in reverse MR. This MR study provides genetic evidence for the causal relationship between the genetically predicted increase in circulating neutrophils count and the risk of IBD (UC and CD). This finding stresses the need for further exploring physiological functions of neutrophils in order to develop effective strategies against IBD.
Collapse
Affiliation(s)
- Chengtao Liang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Zhibin Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yuhe Mai
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Qiuhong Dai
- Qinhuangdao Hospital of Traditional Chinese Medicine, Qinhuangdao, PR China
| | - Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Muyuan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yuyue Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yitong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Xinyu Lu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Zhengdao Lin
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| |
Collapse
|
34
|
Steindl D, Schroeder T, Krannich A, Nee J. Hemoadsorption in the Management of Septic Shock: A Systematic Review and Meta-Analysis. J Clin Med 2025; 14:2285. [PMID: 40217734 PMCID: PMC11989519 DOI: 10.3390/jcm14072285] [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: 02/15/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Septic shock remains a significant clinical challenge with consistently high mortality rates. Recent investigations have focused on the efficacy of CytoSorb® (CytoSorbents Corporation, Monmouth Junction, NJ, USA), an extracorporeal cytokine adsorber, and how it impacts outcomes in sepsis. The current meta-analysis reports on the impact of CytoSorb® on survival, specifically in septic shock patients. Methods: We conducted a comprehensive systematic search across the PubMed and COCHRANE databases for studies published up to 10 June 2024. The analysis prioritized randomized controlled trials and observational studies with control groups involving septic shock patients while excluding case reports and case series. Nine studies were finally included in our meta-analysis following the initial screening of 115 articles after excluding duplicates and irrelevant entries. Results: The meta-analysis was performed on 744 critically ill patients with septic shock from one RCT and eight observational studies. Of these, 449 patients received treatment with CytoSorb® in addition to standard care. Our data indicate that CytoSorb® use is associated with reduced in-hospital mortality, evidenced by an odds ratio (OR) of 0.64 [0.42; 0.97] and a p-value of 0.036. For 28-30-day mortality, the findings were more pronounced with an OR of 0.49 [0.28; 0.83] and a p-value of 0.003. The analysis of the longest observed mortality showed a trend for improved survival within the CytoSorb group; however, it did not reach statistical significance. Additionally, there was a significant improvement in hemodynamic stability as a secondary endpoint, as evidenced by notable reductions in vasopressor requirements in the hemoadsorption group. Conclusions: The current meta-analysis suggests that the use of CytoSorb® alongside standard of care management may be linked to improved short-term survival in patients with septic shock; however, these findings should be interpreted with caution in light of the heterogeneity and the modest quality of the studies included. Prospective studies are needed to better determine the impact of hemoadsorption on shock reversal and survival in these critically ill patients.
Collapse
Affiliation(s)
- David Steindl
- Charité Poison Control Center, Charité—Universitätsmedizin Berlin, 1220 Berlin, Germany;
| | - Tim Schroeder
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (T.S.); (J.N.)
| | | | - Jens Nee
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (T.S.); (J.N.)
| |
Collapse
|
35
|
Ji H, Wang Y, Cao X, Liu Y, Xu M, Zhao X, Chen M. Neutrophil percentage to albumin ratio predicts cardiovascular and all-cause mortality in diabetes and pre diabetes patients. Sci Rep 2025; 15:10075. [PMID: 40128326 PMCID: PMC11933700 DOI: 10.1038/s41598-025-93558-5] [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: 11/09/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
The association between Neutrophil-Percentage-to-Albumin Ratio (NPAR) and mortality in cardiovascular disease (CVD) patients with diabetes or pre-diabetes is not well understood. This study investigates the relationship between baseline NPAR levels and all-cause and cardiovascular mortality among American adults with CVD and diabetes or pre-diabetes. This study enrolled 6,080 patients with diabetes or prediabetes from the National Health and Nutrition Examination Survey (2001-2018). Mortality outcomes were determined by linkage to the National Death Index (NDI) records through December 31, 2019. Multivariate Cox proportional hazards models were used to explore associations between NPAR and mortality. Non-linear correlations were assessed with restricted cubic splines, and segmented Cox proportional hazards models were used to evaluate threshold effects. Receiver operating characteristic (ROC) curves were used to evaluate NPAR's predictive ability for all-cause mortality. Weighted Kaplan-Meier curves with log-rank tests assessed cumulative survival differences across NPAR levels. In this cohort study, with a total follow-up of 53,217 person-years, 1,378 deaths from all causes and 476 deaths from CVD were recorded. Restricted cubic spline analysis revealed a J-shaped association between NPAR and both all-cause and cardiovascular mortality. Threshold effect analysis identified inflection points for NPAR in relation to all-cause mortality at 15.1 and cardiovascular mortality at 14.2. When baseline NPAR exceeded these inflection points, a positive correlation was observed with all-cause mortality (HR: 1.55, 95% CI: 1.08-2.16) and cardiovascular mortality (HR: 1.25, 95% CI: 1.09-1.86). ROC curves for 3-year, 5-year, and 10-year survival rates for all-cause mortality had areas under the curve (AUC) of 0.83, 0.83, and 0.81, respectively. For cardiovascular mortality, the AUC values were 0.86, 0.87, and 0.84. Increased NPAR is significantly associated with increased all-cause and cardiovascular mortality in individuals with diabetes or prediabetes, suggesting its potential role as a prognostic marker.
Collapse
Affiliation(s)
- Hua Ji
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Yongqi Wang
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Xinyi Cao
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Yichang Liu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Murong Xu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China.
| | - Mingwei Chen
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei City, Anhui Province, People's Republic of China.
| |
Collapse
|
36
|
Lian Y, Lai X, Wu C, Wang L, Shang J, Zhang H, Jia S, Xing W, Liu H. The roles of neutrophils in cardiovascular diseases. Front Cardiovasc Med 2025; 12:1526170. [PMID: 40176832 PMCID: PMC11961988 DOI: 10.3389/fcvm.2025.1526170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
The immune response plays a vital role in the development of cardiovascular diseases (CVDs). As a crucial component of the innate immune system, neutrophils are involved in the initial inflammatory response following cardiovascular injury, thereby inducing subsequent damage and promoting recovery. Neutrophils exert their functional effects in tissues through various mechanisms, including activation and the formation of neutrophil extracellular traps (NETs). Once activated, neutrophils are recruited to the site of injury, where they release inflammatory mediators and cytokines. This study discusses the main mechanisms associated with neutrophil activity and proposes potential new therapeutic targets. In this review, we systematically summarize the diverse phenotypes of neutrophils in disease regulatory mechanisms, different modes of cell death, and focus on the relevance of neutrophils to various CVDs, including atherosclerosis, acute coronary syndrome, myocardial ischemia/reperfusion injury, hypertension, atrial fibrillation, heart failure, and viral myocarditis. Finally, we also emphasize the preclinical/clinical translational significance of neutrophil-targeted strategies.
Collapse
Affiliation(s)
- Yanjie Lian
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaolei Lai
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Cong Wu
- Beijing Hospital of Traditional Chinese Medicine, Huairou Hospital, Beijing, China
| | - Li Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - JuJu Shang
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Heyi Zhang
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Sihan Jia
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wenlong Xing
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Hongxu Liu
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Zhang X, Cui X, Sun N, Wu X, Pan X, Wang R, Chen Z, Li Y, Hu Y, Liu F, Cao X. Microglial repopulation alleviates surgery-induced neuroinflammation and cognitive impairment in a ZEB1-dependent manner. FASEB J 2025; 39:e70440. [PMID: 40052833 DOI: 10.1096/fj.202402492r] [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: 10/16/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Microglia play a crucial role in postoperative cognitive dysfunction (POCD). This study investigated the effects of microglial depletion and subsequent repopulation on POCD and its underlying mechanisms. An aged mouse model of POCD was induced by partial hepatectomy, and the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 was administered to facilitate microglial depletion and repopulation. Neutrophil involvement was assessed with anti-Ly6G antibodies, while ZEB1 was manipulated through shRNA knockdown and lentiviral overexpression in the BV2 microglial cell line. A TGF-β1 neutralizing antibody was employed to elucidate the relationship between ZEB1 and its downstream pathways. The results indicated that microglial depletion alone did not reverse cognitive impairments. However, microglial repopulation significantly reduced neutrophil infiltration and improved cognitive function post-surgery. This improvement correlated with ZEB1 upregulation in microglia, which decreased CXCL1 production by astrocytes via TGF-β1 signaling, thereby reducing neutrophil migration to the hippocampus. These findings suggest that microglial repopulation, dependent on ZEB1 and TGF-β1 signaling, effectively alleviates neuroinflammation, reduces neutrophil infiltration, and enhances cognitive function, highlighting microglia as a promising target for the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaotong Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Naihui Sun
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyi Wu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xue Pan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renyi Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zitong Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yilong Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Hu
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fang Liu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuezhao Cao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
38
|
Santos AP, Rodrigues LS, Rother N, Mello FCDQ, Magis-Escurra C. The role of neutrophil response in lung damage and post-tuberculosis lung disease: a translational narrative review. Front Immunol 2025; 16:1528074. [PMID: 40124364 PMCID: PMC11925771 DOI: 10.3389/fimmu.2025.1528074] [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: 11/14/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
It is estimated that more than 150 million individuals alive in 2020 had survived tuberculosis (TB). A portion of this large population continues to experience chronic respiratory abnormalities, with or without symptoms, due to previous active pulmonary TB. This condition known as Post-TB Lung Disease (PTLD), involves a complex interaction between pathogen, host and environmental factors. These interactions are believed to drive a hyperinflammatory process in the lungs during active TB, resulting in tissue damage, which may lead to radiological sequelae, impaired pulmonary function, clinical symptoms, such as cough, dyspnea, hemoptysis, and respiratory infections. Such complications impose significant health, financial, and social burdens, which remain poorly understood and inadequately addressed by health care systems. Given the heterogeneity of immune cells and their products infiltrating the airways and the lung parenchyma during acute and chronic inflammation caused by Mycobacterium tuberculosis infection, it is evident that TB immunopathology is multifactorial. Among the various components involved, neutrophils have recently emerged as critical contributors to the deleterious immune response against TB, leading to severe pulmonary damage. In this translational narrative review, we aim to summarize the role of neutrophils and their primary products - proteases (such as elastase), matrix metalloproteinases and neutrophils extracellular traps (NETs) - in pulmonary TB. We highlight new concepts and emerging evidence of neutrophil involvement during the active disease, translating these insights from "bench to bedside" to facilitate dialogue between fundamental researchers and clinical practitioners. Additionally, we present potential targets for future treatment strategies that could mitigate or even prevent PTLD.
Collapse
Affiliation(s)
- Ana Paula Santos
- Pulmonary Diseases Department, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Thoracic Diseases Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luciana Silva Rodrigues
- Department of Pathology and Laboratories, Medical Sciences Faculty, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Cecile Magis-Escurra
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
39
|
Tang L, Tang R, Zheng J, Zhao P, Zhu R, Tang Y, Zhang X, Gong X, Wang F. Dissecting biological heterogeneity in major depressive disorder based on neuroimaging subtypes with multi-omics data. Transl Psychiatry 2025; 15:72. [PMID: 40032862 PMCID: PMC11876359 DOI: 10.1038/s41398-025-03286-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
The heterogeneity of Major Depressive Disorder (MDD) has been increasingly recognized, challenging traditional symptom-based diagnostics and the development of mechanism-targeted therapies. This study aims to identify neuroimaging-based MDD subtypes and dissect their predominant biological characteristics using multi-omics data. A total of 807 participants were included in this study, comprising 327 individuals with MDD and 480 healthy controls (HC). The amplitude of low-frequency fluctuations (ALFF), a functional neuroimaging feature, was extracted for each participant and used to identify MDD subtypes through machine learning clustering. Multi-omics data, including profiles of genetic, epigenetics, metabolomics, and pro-inflammatory cytokines, were obtained. Comparative analyses of multi-omics data were conducted between each MDD subtype and HC to explore the molecular underpinnings involved in each subtype. We identified three neuroimaging-based MDD subtypes, each characterized by unique ALFF pattern alterations compared to HC. Multi-omics analysis showed a strong genetic predisposition for Subtype 1, primarily enriched in neuronal development and synaptic regulation pathways. This subtype also exhibited the most severe depressive symptoms and cognitive decline compared to the other subtypes. Subtype 2 is characterized by immuno-inflammation dysregulation, supported by elevated IL-1 beta levels, altered epigenetic inflammatory measures, and differential metabolites correlated with IL-1 beta levels. No significant biological markers were identified for Subtype 3. Our results identify neuroimaging-based MDD subtypes and delineate the distinct biological features of each subtype. This provides a proof of concept for mechanism-targeted therapy in MDD, highlighting the importance of personalized treatment approaches based on neurobiological and molecular profiles.
Collapse
Affiliation(s)
- Lili Tang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Rui Tang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Pengfei Zhao
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Rongxin Zhu
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Yanqing Tang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xizhe Zhang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China.
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering, MOE key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
- Department of Psychiatry, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
40
|
Forouzanfar F, Sahranavard T, Tsatsakis A, Iranshahi M, Rezaee R. Rutin: a pain-relieving flavonoid. Inflammopharmacology 2025; 33:1289-1301. [PMID: 39961908 DOI: 10.1007/s10787-025-01671-8] [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: 12/21/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025]
Abstract
Rutin (vitamin P or rutoside) is a citrus flavonoid glycoside that has shown beneficial health effects in different organs against various conditions including inflammation and pain. The majority of rutin therapeutic benefits are ascribed to its antioxidant and anti-inflammatory properties. This review article discusses studies that investigated pain-relieving activity of rutin and summarizes the reported mechanisms of action. Rutin pain-relieving effect has been studied in streptozotocin-induced diabetes, chronic constriction injury, and oxaliplatin-, formalin-, acetic acid- and glutamate-induced nociception in mice or rats. Based on the literature, rutin analgesic effects are induced through potentiation of antioxidant arsenal, reduction of inflammatory cytokines (e.g., Tumor necrosis factor alpha (TNF-α) and interleukin-1β) levels, suppression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions and modulation of MAPK, NF-κB and Nrf-2/HO-1 signaling. Preclinical findings on rutin pain-relieving activity are promising, however, its safety profile needs to be more thoroughly investigated and clinical trials should be conducted to assess its analgesic effects in humans.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Toktam Sahranavard
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Division of Morphology, Medical School, University of Crete, Voutes Campus, 71003, Heraklion, Greece
| | - Mehrdad Iranshahi
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ramin Rezaee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Center of Toxicology Science & Research, Division of Morphology, Medical School, University of Crete, Voutes Campus, 71003, Heraklion, Greece.
| |
Collapse
|
41
|
Hong SP, Woo JB, Kim HY. Use of the Neutrophil-to-Lymphocyte Ratio Improves the Accuracy of Outcome Prediction in Patients with Acute Traumatic Subdural Hematoma Undergoing Surgical Treatment. World Neurosurg 2025; 195:123642. [PMID: 39755151 DOI: 10.1016/j.wneu.2024.123642] [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] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Traumatic brain injury is a major cause of disability and mortality worldwide. Acute traumatic subdural hematoma (TSDH) accounts for a large proportion of all traumatic brain injury cases. However, factors to predict postoperative prognosis in patients with acute TSDH are limited. Recently, it has been reported that inflammatory markers increase the accuracy of prognosis in various diseases. The neutrophil-to-lymphocyte ratio (NLR) is a marker for inflammation, which is easy to test, inexpensive, and can be performed quickly. However, the prognostic value of NLR in patients with acute TSDH remains controversial. This study therefore aimed to assess the predictive value of the admission and postoperative NLR in patients with acute TSDH who underwent surgical treatment. METHODS We retrospectively identified patients who underwent surgery for acute TSDH at our institute between April 2010 and August 2023. The NLR was calculated as the ratio of the absolute neutrophil count to the absolute lymphocyte count. Multivariable logistic regression analysis was subsequently applied to assess the independent predictors of 30-day mortality. In logistic regression analysis, multivariate analysis was performed using the backward elimination method for all P value<0.05 in the univariate analysis. Receiver-operating characteristic curve analysis was used to assess the predictive abilities of the postoperative 48-hour NLR and determine the cutoff values. RESULTS A total of 131 patients were enrolled, among whom the mortality within 1 month was 47.3% (62 patients). Initial NLR (P value = 0.905) was not strongly associated with mortality in patients with acute TSDH who underwent surgery. Only the postoperative 48-hour NLR (odds ratio, 1.103; 95% confidence interval, 1.051-1.157; P < 0.001) and Glasgow Coma Scale score at admission (odds ratio, 0.855; 95% confidence interval, 0.756-0.967; P = 0.012) were independent factors for 1-month mortality in the multivariate logistic analysis. The optimal cutoff value of the postoperative 48-hour NLR to distinguish between survival and nonsurvival was 15.786. CONCLUSIONS Initial NLR was not strongly associated with 1-month mortality in patients with acute TSDH who underwent surgery. However, the postoperative 48-hour NLR was associated with 1-month mortality.
Collapse
Affiliation(s)
- Seok Pyo Hong
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Joon Bum Woo
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Hae Yu Kim
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University College of Medicine, Busan, South Korea.
| |
Collapse
|
42
|
Gabashvili AN, Vasiukova AA, Rakitina AS, Garanina AS. The Issue on Dualistic Role of Neutrophils in Carcinogenesis and Their Possible Use for Treatment of Malignant Neoplasms. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:303-320. [PMID: 40367075 DOI: 10.1134/s000629792460368x] [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: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 05/16/2025]
Abstract
Neutrophils are phagocytic leukocytes of the myeloid series, which are the most common myeloid cells in human blood, normally accounting from 65 to 80% of all circulating leukocytes. Over the years of investigation of these cells, more and more evidence has emerged indicating functional plasticity of neutrophils and their ambiguous role in the tumor development. Similarly to the M1/M2 classification of macrophages, the N1/N2 paradigm could be applied to neutrophils, where N1-neutrophils exhibit tumor-suppressive properties, and N2-neutrophils contribute to tumor development and immune suppression. An important natural feature of neutrophils is their mobility and ability to overcome physical barriers, thus these cells, as well as their vesicles and membranes, could be used to deliver therapeutic drugs to tumor cells. In addition, neutrophils themselves could be activated and mobilized to fight the tumor. This review describes current state of research on the role of neutrophils in carcinogenesis, as well as possible approaches of using these cells and their derivatives as systems for targeted delivery of therapeutic drugs for treatment of malignant neoplasms.
Collapse
Affiliation(s)
- Anna N Gabashvili
- Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, 119991, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Anastasiia A Vasiukova
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, 119034, Russia
| | - Aleksandra S Rakitina
- Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, 119991, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | | |
Collapse
|
43
|
Ye S, Cao Q, Ni P, Xiong S, Zhong M, Yuan T, Shan J, Liang J, Fan Y, Zhang X. A ceramic microbridge microfluidic chip to study osteogenic differentiation of mesenchymal stem cells in bioactive ceramic immune microenvironment. Bioact Mater 2025; 45:520-533. [PMID: 39735335 PMCID: PMC11681893 DOI: 10.1016/j.bioactmat.2024.11.005] [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: 06/24/2024] [Revised: 09/13/2024] [Accepted: 11/05/2024] [Indexed: 12/31/2024] Open
Abstract
Bioactive ceramics have been used in bone tissue repair and regeneration. However, because of the complex in vivo osteogenesis process, long cycle, and difficulty of accurately tracking, the mechanism of interaction between materials and cells has yet to be fully understood, hindering its development. The ceramic microbridge microfluidic chip system may solve the problem and provide an in vitro method to simulate the microenvironment in vivo. Nevertheless, the complex microenvironment parameters of the chip system need to be studied in detail. Computer simulation bionics can provide clues for the setting of microenvironment parameters. This study used a computational bionic model to simulate the bone growth process in the presence of immune-related factors. The osteoblast differentiation of mesenchymal stem cells of calcium phosphate ceramics in a macrophage-dominated immune microenvironment was studied using a microfluidic chip system. The computational biomimetic model and microfluidic chip findings were basically consistent with the reported results of the animal experiments. These findings suggest that studying the osteogenic behavior of calcium phosphate ceramics using a microfluidic chip model is feasible. The method model provided in this study can be extended to other biomaterials, providing a viable path for their research and evaluation.
Collapse
Affiliation(s)
- Sheng Ye
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- School of Big Health & Intelligent Engineering, Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Nuclear Industry 416 Hospital, the 2nd Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610057, China
| | - Quanle Cao
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Panxianzhi Ni
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Shuting Xiong
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Meng Zhong
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Tun Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Jing Shan
- Department of Gastroenterology, the 3rd People's Hospital of Chengdu, Southwest Jiaotong University, Chengdu, Sichuan, 610064, China
| | - Jie Liang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Yujiang Fan
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xingdong Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| |
Collapse
|
44
|
Li Y, Xu X, Wu X, Li J, Chen S, Chen D, Li G, Tang Z. Cell polarization in ischemic stroke: molecular mechanisms and advances. Neural Regen Res 2025; 20:632-645. [PMID: 38886930 PMCID: PMC11433909 DOI: 10.4103/nrr.nrr-d-23-01336] [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: 08/10/2023] [Revised: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 06/20/2024] Open
Abstract
Ischemic stroke is a cerebrovascular disease associated with high mortality and disability rates. Since the inflammation and immune response play a central role in driving ischemic damage, it becomes essential to modulate excessive inflammatory reactions to promote cell survival and facilitate tissue repair around the injury site. Various cell types are involved in the inflammatory response, including microglia, astrocytes, and neutrophils, each exhibiting distinct phenotypic profiles upon stimulation. They display either proinflammatory or anti-inflammatory states, a phenomenon known as 'cell polarization.' There are two cell polarization therapy strategies. The first involves inducing cells into a neuroprotective phenotype in vitro, then reintroducing them autologously. The second approach utilizes small molecular substances to directly affect cells in vivo. In this review, we elucidate the polarization dynamics of the three reactive cell populations (microglia, astrocytes, and neutrophils) in the context of ischemic stroke, and provide a comprehensive summary of the molecular mechanisms involved in their phenotypic switching. By unraveling the complexity of cell polarization, we hope to offer insights for future research on neuroinflammation and novel therapeutic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
45
|
Zhao X, Wu F, Zhao S, Chen W, Si W, Li Y, Zhang D, Wang J, Wang N, Sun L, Sun Z, Chang H, Du G. The clinical value of the neutrophil-to-lymphocyte ratio, systemic immune-inflammation index, monocyte-to-lymphocyte ratio and platelet-to-lymphocyte ratio for predicting the severity of patients with autoimmune encephalitis. Front Neurol 2025; 16:1498007. [PMID: 40093740 PMCID: PMC11906310 DOI: 10.3389/fneur.2025.1498007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Background The systemic inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR) and platelet-to-lymphocyte ratio (PLR) are inflammatory markers in peripheral blood, which have been proven to be associated with some central nervous system diseases. We aimed to evaluate the association of SII, NLR MLR and PLR with the severity of autoimmune encephalitis (AE) and to compare the predictive value of those biomarkers in the early identification of ICU admission. Methods This retrospective study was conducted in three medical centers in China. We included 176 patients diagnosed with AE and 200 age and gender-matched healthy controls and correlated their demographic and clinical data. The SII, NLR, MLR and PLR levels were calculated from the blood routine tests. The severity of the patients was evaluated by the Clinical Assessment Scale for Autoimmune Encephalitis (CASE) and the modified Rankin Scale (mRS) at admission, and the patients were divided into two groups according to the ICU admission. Results The SII, NLR, MLR and PLR were significantly higher in AE patients than that in HCs (<0.001 for all). The SII and NLR were positively correlated with the CASE score (r = 0.243, p = 0.001; r = 0.237, p = 0.002) and the mRS score (r = 0.185, p = 0.014; r = 0.185, p = 0.014) in AE patients. The MLR and PLR were only positively correlated with the CASE score (r = 0.242, p = 0.001; r = 0.158, p = 0.036). The SII and NLR of the ICU group were significantly higher than that of the non-ICU group. The result of receiver operating characteristic (ROC) analysis showed that NLR was the best predictor of ICU admission for AE patients (AUC = 0.701). NLR and MLR had similar predictive ability (AUC = 0.654; AUC = 0.608) and were superior to PLR. The optimal NLR cut-off value for the incidence of ICU was 3.906. Conclusion Increased SII, NLR, MLR and PLR at admission are positively correlated with the CASE score of AE patients. Among the four indexes, the NLR is the best predictor of ICU admission, which may be helpful for clinicians to monitor disease progression and identify potentially severe patients of AE.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Fen Wu
- Department of Clinical Laboratory, Liaocheng Third People’s Hospital, Liaocheng, China
| | - Shunfeng Zhao
- Department of Clinical Laboratory, Liaocheng Third People’s Hospital, Liaocheng, China
| | - Wenna Chen
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Wei Si
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yuanrui Li
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Dengke Zhang
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jing Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ningning Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Sun
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiyu Sun
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Haoxiao Chang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
46
|
Zhou HJ, Mu BX, Wen MC, Zhao Q, Li Y, Zhao WX, Yin HY, Ren S, Zhou JY, Chen M. Yiqi Huayu Jiedu Decoction reduces colorectal cancer liver metastasis by promoting N1 neutrophil chemotaxis. Front Immunol 2025; 16:1530053. [PMID: 40083557 PMCID: PMC11903724 DOI: 10.3389/fimmu.2025.1530053] [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: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Objective To observe the inhibitory effect and potential mechanism of Yiqi Huayu Jiedu Decoction (YHJD) on liver metastasis of colorectal cancer (CRC). Methods We compared the changes of liver weight and liver index before and after YHJD treatment in CRC liver metastasis mouse models. HE staining was employed to observe the pathological changes in mouse liver tissue sections. Flow cytometry was used to analyze the number and marker of neutrophils treated with YHJD. Transcriptomics, proteomics, and multiplex cytokine array analyses were conducted to further verify the role of YHJD on CXCL1. Differential gene analysis was performed to further explore the mechanism by which YHJD inhibits liver metastasis of CRC. Results Animal studies demonstrated that YHJD reduces liver metastases. Flow cytometry results revealed that YHJD promotes N1 neutrophils in liver. Combining multi-omics and multiple cytokine arrays, we observed a significant increase in the expression of CXCL1 in the liver and plasma. GO and KEGG enrichment analyses indicated that YHJD may regulate the chemotaxis of neutrophils to inhibit the liver metastasis of CRC by participating in the regulation of cell adhesion molecule binding, adhesion protein binding, and multiple metabolic pathways. Conclusions YHJD inhibits CRC liver metastasis by upregulating CXCL1, thereby promoting N1 neutrophil chemotaxis towards the liver, and concurrently raising the expression of N1 neutrophil markers.
Collapse
Affiliation(s)
- Hua-Jian Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Bai-Xiang Mu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Meng-Chao Wen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qi Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuanxiang Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wen-Xuan Zhao
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hong-Ye Yin
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin-Yong Zhou
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
47
|
Pesenti L, de Oliveira Formiga R, Tamassia N, Gardiman E, Chable de la Héronnière F, Gasperini S, Chicher J, Kuhn L, Hammann P, Le Gall M, Saraceni-Tasso G, Martin C, Hosmalin A, Breckler M, Hervé R, Decker P, Ladjemi MZ, Pène F, Burgel PR, Cassatella MA, Witko-Sarsat V. Neutrophils Display Novel Partners of Cytosolic Proliferating Cell Nuclear Antigen Involved in Interferon Response in COVID-19 Patients. J Innate Immun 2025; 17:154-175. [PMID: 40015257 PMCID: PMC11867639 DOI: 10.1159/000543633] [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: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Neutrophils are key players in the hyperinflammatory response during SARS-CoV-2 infection. The cytosolic proliferating cell nuclear antigen (PCNA) is a scaffolding protein highly dependent on the microenvironment status and known to interact with numerous proteins that regulate neutrophil functions. This study aimed to examine the cytosolic protein content and PCNA interactome in neutrophils from COVID-19 patients. METHODS Proteomic analyses were performed on neutrophil cytosols from healthy donors and patients with severe or critical COVID-19. In vitro approaches were used to explore the biological significance of the COVID-19-specific PCNA interactome. RESULTS Neutrophil cytosol analysis revealed a strong interferon (IFN) protein signature, with variations according to disease severity. Interactome analysis identified associations of PCNA with proteins involved in interferon signaling, cytoskeletal organization, and neutrophil extracellular trap (NET) formation, such as protein arginine deiminase type-4 (PADI4) and histone H3, particularly in critical patients. Functional studies of interferon signaling showed that T2AA, a PCNA scaffold inhibitor, downregulated IFN-related genes, including STAT1, MX1, IFIT1, and IFIT2 in neutrophils. Additionally, T2AA specifically inhibited the secretion of CXCL10, an IFN-dependent cytokine. PCNA was also found to interact with key effector proteins implicated in NET formation, such as histone H3, especially in critical COVID-19 cases. CONCLUSION The analysis of the PCNA interactome has unveiled new protein partners that enhance the interferon pathway, thereby modulating immune responses and contributing to hyperinflammation in COVID-19. These findings provide valuable insights into interferon dysregulation in other immune-related conditions. INTRODUCTION Neutrophils are key players in the hyperinflammatory response during SARS-CoV-2 infection. The cytosolic proliferating cell nuclear antigen (PCNA) is a scaffolding protein highly dependent on the microenvironment status and known to interact with numerous proteins that regulate neutrophil functions. This study aimed to examine the cytosolic protein content and PCNA interactome in neutrophils from COVID-19 patients. METHODS Proteomic analyses were performed on neutrophil cytosols from healthy donors and patients with severe or critical COVID-19. In vitro approaches were used to explore the biological significance of the COVID-19-specific PCNA interactome. RESULTS Neutrophil cytosol analysis revealed a strong interferon (IFN) protein signature, with variations according to disease severity. Interactome analysis identified associations of PCNA with proteins involved in interferon signaling, cytoskeletal organization, and neutrophil extracellular trap (NET) formation, such as protein arginine deiminase type-4 (PADI4) and histone H3, particularly in critical patients. Functional studies of interferon signaling showed that T2AA, a PCNA scaffold inhibitor, downregulated IFN-related genes, including STAT1, MX1, IFIT1, and IFIT2 in neutrophils. Additionally, T2AA specifically inhibited the secretion of CXCL10, an IFN-dependent cytokine. PCNA was also found to interact with key effector proteins implicated in NET formation, such as histone H3, especially in critical COVID-19 cases. CONCLUSION The analysis of the PCNA interactome has unveiled new protein partners that enhance the interferon pathway, thereby modulating immune responses and contributing to hyperinflammation in COVID-19. These findings provide valuable insights into interferon dysregulation in other immune-related conditions.
Collapse
Affiliation(s)
- Lucie Pesenti
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | | | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Johana Chicher
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Lauriane Kuhn
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Philippe Hammann
- Strasbourg-Esplanade Proteomics Platform, CNRS UAR1589, Molecular and Cellular Biology Institute, University of Strasbourg, Strasbourg, France
| | - Morgane Le Gall
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | | | - Clémence Martin
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Respiratory Medicine, AP-HP, Cochin Hospital, Paris, France
| | - Anne Hosmalin
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | - Magali Breckler
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Roxane Hervé
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Patrice Decker
- INSERM UMR 1125, Bobigny, France
- UFR SMBH, Li2P, Université Sorbonne Paris Nord, Bobigny, France
| | - Maha Zohra Ladjemi
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
| | - Frédéric Pène
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Intensive Medicine and Reanimation, AP-HP, Cochin Hospital, Paris, France
| | - Pierre-Régis Burgel
- INSERM U1016, Institut Cochin, CNRS 8104, Université Paris Cité, Paris, France
- Department of Respiratory Medicine, AP-HP, Cochin Hospital, Paris, France
| | - Marco A. Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | |
Collapse
|
48
|
Li H, Shan W, Zhao X, Sun W. Neutrophils: Linking Inflammation to Thrombosis and Unlocking New Treatment Horizons. Int J Mol Sci 2025; 26:1965. [PMID: 40076593 PMCID: PMC11901051 DOI: 10.3390/ijms26051965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophils play a key role in inflammatory responses and thrombosis, but their complex interactions in disease pathogenesis are not fully understood. This review examines the multifaceted roles of neutrophils, focusing on their activation, cytokine release, and formation of neutrophil extracellular traps (NETs), which contribute to host defense and thrombosis. We discuss the interaction between inflammation and coagulation, the direct effect of neutrophils on thrombus stability, and their involvement in pathological thrombotic diseases. The therapeutic potential of neutrophil drug loading in the treatment of thrombosis, as well as the clinical implications and future research directions, are highlighted. The aim of this review is to gain insight into the critical neutrophil-inflammation-thrombus axis and its potential as a therapeutic target for thrombotic diseases and to suggest possible directions for neutrophil-loaded drug therapy for thrombosis.
Collapse
Affiliation(s)
| | | | | | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.L.); (W.S.); (X.Z.)
| |
Collapse
|
49
|
Ciurus S, Elewa MAF, Palmer MA, Wolf A, Hector M, Fuhrmann DC, Thomas D, Gurke R, Schwalm MP, Berger L, Zech TJ, Burgers LD, Marschalek R, Geisslinger G, Knapp S, Langmann T, Bracher F, Weigert A, Fürst R. Inhibition of DYRK1B BY C81 impedes inflammatory processes in leukocytes by reducing STAT3 activity. Cell Mol Life Sci 2025; 82:85. [PMID: 39985685 PMCID: PMC11846820 DOI: 10.1007/s00018-025-05579-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 10/15/2024] [Accepted: 01/05/2025] [Indexed: 02/24/2025]
Abstract
Chronic inflammatory diseases are a significant global burden and are associated with dysregulated resolution of inflammation. Therefore, promoting the process of resolution is a promising therapeutic approach. This study presents the potent anti-inflammatory and pro-resolving effects of a natural product-derived compound called C81. Administration of C81 in a therapeutic window resolved inflammation in the murine imiquimod-induced psoriasis model, and reduced microglial infiltration in a laser-induced choroidal neovascularisation model. Investigations into the underlying mechanisms of C81 identified the DYRK1B/STAT3 axis as a new regulator of inflammatory processes in leukocytes. The inhibition of DYRK1B by C81 resulted in attenuated STAT3 phosphorylation. The depletion of STAT3-regulated gene expression led to the inhibition of leukocyte adhesion and migration due to reduced integrin activation, and in addition to the inhibition of the release of pro-inflammatory mediators such as cytokines and eicosanoids. Importantly, the pro-resolving effects of C81 included the cell type-specific induction of apoptosis in neutrophils and a subsequent increase in efferocytosis. In conclusion, we report the DYRK1B/STAT3 axis as a novel and promising therapeutic target for activating the resolution of inflammation.
Collapse
Affiliation(s)
- Sarah Ciurus
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Mohammed A F Elewa
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
- Department of Biochemistry, Faculty of Pharmacy, Kafr El-Sheikh University, Karf El-Sheikh, Egypt
| | - Megan A Palmer
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Martin P Schwalm
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Lena Berger
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas J Zech
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Luisa D Burgers
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry and Buchmann Institute Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Franz Bracher
- Pharmaceutical Chemistry, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany.
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
50
|
Zhou X, Jin Y, Zhu Y, Luo X, Li S, Shen W, Wu R. The Role of Crosstalk between Nets and Keratinocytes in Skin Immunity. J Invest Dermatol 2025:S0022-202X(25)00012-0. [PMID: 39985552 DOI: 10.1016/j.jid.2024.07.040] [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: 11/15/2023] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 02/24/2025]
Abstract
The skin is the principal barrier against pathogens. Skin-resident cells, especially keratinocytes, play essential roles in skin immunity. Damage to the integrity of the skin barrier triggers the localized release of proinflammatory factors from keratinocytes, which attract neutrophils. These infiltrating neutrophils in turn release cytokines to modulate keratinocyte function, thereby amplifying skin inflammation. In addition, neutrophils produce neutrophil extracellular traps in a process called NETosis. Notably, crosstalk between neutrophils and keratinocytes is a prominent feature of skin infection eradication and autoimmune disorder development. In this paper, we review research progress on neutrophil extracellular traps in cutaneous immunity, with a particular emphasis on their modulation of keratinocytes. Moreover, we discuss the implications of neutrophil heterogeneity for immune defense and disease development and treatment.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Yi Jin
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Yanshan Zhu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Xin Luo
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Siying Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Weiyun Shen
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Ruifang Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China.
| |
Collapse
|