1
|
Givian A, Azizan A, Jamshidi A, Mahmoudi M, Farhadi E. Iron metabolism in rheumatic diseases. J Transl Autoimmun 2025; 10:100267. [PMID: 39867458 PMCID: PMC11763848 DOI: 10.1016/j.jtauto.2025.100267] [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: 11/09/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Iron is a crucial element for living organism in terms of oxygen transport, hematopoiesis, enzymatic activity, mitochondrial respiratory chain function and also immune system function. The human being has evolved a mechanism to regulate body iron. In some rheumatic diseases such as rheumatoid arthritis (RA), systemic lupus erythematous (SLE), systemic sclerosis (SSc), ankylosing spondylitis (AS), and gout, this balanced iron regulation is impaired. Altered iron homeostasis can contribute to disease progression through ROS production, fibrosis, inflammation, abnormal bone homeostasis, NETosis and cell senescence. In this review, we have focused on the iron metabolism in rheumatic disease and its role in disease progression.
Collapse
Affiliation(s)
- Aliakbar Givian
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Amin Azizan
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Ando S, Miyatake JI, Takeda M, Amakawa R, Nakamine H. Langerhans Cell Histiocytosis With Unusual Hexagonal Crystals in Addition to Usual Charcot-Leyden Crystals. Report of a Patient With Possible Process of Crystal Formation and Clinical Significance of a "Necrotic" Change. Int J Surg Pathol 2025; 33:153-159. [PMID: 38646671 DOI: 10.1177/10668969241246456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Langerhans cell histiocytosis is a rare neoplastic disorder characterized by the proliferation of Langerhans cells and often accompanied by eosinophil infiltration. Charcot-Leyden crystals, composed of galectin 10, are occasionally observed in Langerhans cell histiocytosis; however, histological images are rarely reported. We herein present a patient with Langerhans cell histiocytosis with Charcot-Leyden crystals and hexagonal crystals by describing the histologic and immunohistochemical features of a lymph node. A unique distribution of Charcot-Leyden crystals and hexagonal crystals was observed in this patient, shedding light on their possible formation process of the latter. We discuss the biological significance of eosinophilic abscesses in Langerhans cell histiocytosis and propose that these crystals may be linked to extracellular trap-cell death (ETosis). This example challenges the conventional characterization of "necrosis" in Langerhans cell histiocytosis and underscores the importance of recognizing ETosis as a potential mechanism involved in the pathogenesis of Langerhans cell histiocytosis. Further studies are underway to validate significance of these findings in a larger cohort of Langerhans cell histiocytosis patients.
Collapse
Affiliation(s)
- Sayaka Ando
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | | | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | - Ryuichi Amakawa
- Department of Hematology, Japan Baptist Hospital, Kyoto, Japan
| | - Hirokazu Nakamine
- Department of Pathology and Laboratory Medicine, Japan Baptist Hospital, Kyoto, Japan
| |
Collapse
|
3
|
Wu P, Chen J, Li H, Lu H, Li Y, Zhang J. Interactions between ferroptosis and tumour development mechanisms: Implications for gynaecological cancer therapy (Review). Oncol Rep 2025; 53:18. [PMID: 39635847 PMCID: PMC11638741 DOI: 10.3892/or.2024.8851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Ferroptosis is a form of programmed cell death that is distinct from apoptosis. The mechanism involves redox‑active metallic iron and is characterized by an abnormal increase in iron‑dependent lipid reactive oxygen species, which results in high levels of membrane lipid peroxides. The relationship between ferroptosis and gynaecological tumours is complex. Ferroptosis can regulate tumour proliferation, metastasis and chemotherapy resistance, and targeting ferroptosis is a promising antitumour approach. Ferroptosis interacts with mechanisms related to tumorigenesis and development, such as macrophage polarization, the neutrophil trap network, mitochondrial autophagy and cuproptosis. The present review examines recent information on the interaction between the molecular mechanism of ferroptosis and other tumour‑related mechanisms, as well as the involvement of ferroptosis in gynaecological tumours, to identify implications for gynaecological cancer therapy.
Collapse
Affiliation(s)
- Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Jianlin Chen
- Department of Assisted Reproductive Centre, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Haiyuan Lu
- Department of Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Hunan Vigorzoe Biotechnology Co., Ltd., Hunan 417700, P.R. China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| |
Collapse
|
4
|
Xia L, Yan X, Zhang H. Mitochondrial DNA-activated cGAS-STING pathway in cancer: Mechanisms and therapeutic implications. Biochim Biophys Acta Rev Cancer 2025; 1880:189249. [PMID: 39701325 DOI: 10.1016/j.bbcan.2024.189249] [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: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Mitochondrial DNA (mtDNA), a circular double-stranded DNA located within mitochondria, plays a pivotal role in mitochondrial-induced innate immunity, particularly via the cyclic GMP-AMP synthase (cGAS)-STING pathway, which recognizes double-stranded DNA and is crucial for pathogen resistance. Recent studies elucidate the interplay among mtDNA, the cGAS-STING pathway, and neutrophil extracellular traps (NETs) in the context of cancer. mtDNA uptake by recipient cells activates the cGAS-STING pathway, while mtDNA leakage reciprocally regulates NET release, amplifying inflammation and promoting NETosis, a mechanism of tumor cell death. Autophagy modulates these processes by clearing damaged mitochondria and degrading cGAS, thus preventing mtDNA recognition. Tumor microenvironmental factors, such as metabolic reprogramming and lipid accumulation, induce mitochondrial stress, ROS production, and further mtDNA leakage. This review explores strategies in cancer drug development that leverage mtDNA leakage to activate the cGAS-STING pathway, potentially converting 'cold tumors' into 'hot tumors,' while discussing advancements in targeted therapies and proposing new research methodologies.
Collapse
Affiliation(s)
- Lintao Xia
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiuli Yan
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Hui Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
5
|
Verçosa BLA, Muniz-Junqueira MI, Mineiro ALBB, Melo MN, Vasconcelos AC. Enhanced apoptosis and inflammation allied with autophagic and apoptotic Leishmania amastigotes in the seemingly undamaged ear skin of clinically affected dogs with canine visceral Leishmaniasis. Cell Immunol 2025; 408:104909. [PMID: 39701006 DOI: 10.1016/j.cellimm.2024.104909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Programmed cell death plays a relevant role in the pathogenesis of visceral Leishmaniasis. Apoptosis selects suitable parasites, regulating parasite density, whereas autophagy eliminates pathogens. This study aimed to assess the inflammation and apoptosis in inflammatory cells and presents a unique description of the presence of autophagic and apoptotic Leishmania amastigotes in naturally Leishmania-infected dogs. Fragments from seemingly undamaged ear skin of sixteen Leishmania-infected dogs and seven uninfected dogs were evaluated through histomorphometry, ultrastructural, immunohistochemical and transmission electron microscopy (TEM) analyses. Leishmania amastigotes were present on seemingly undamaged ear skin only in clinically affected dogs. Parasite load, morphometrical parameters of inflammation and apoptotic index of inflammatory cells were higher in clinically affected animals and were related to clinical manifestations. Apoptotic index and morphometric parameters of the inflammatory infiltrate in undamaged ear skin were positively correlated with parasite load. Apoptotic and non-apoptotic Leishmania amastigotes were observed within neutrophils and macrophages. Leishmania amastigotes were positive for Bax, a marker for apoptosis, by immunohistochemistry. Morphological characteristics of apoptosis and autophagy in Leishmania amastigotes were observed only in phagocytes of clinically affected dogs. Positive correlations were found between histomorphometry and clinical manifestations. Our results showed that apoptosis and autophagy in Leishmania amastigotes may be related to both the increase in parasite load and apoptotic index in inflammatory cells, and with the intensity of the inflammatory response in clinically affected dogs. Thus, our study suggests that apoptotic and autophagy Leishmania within phagocytes may have facilitate the survival of the parasite and it appears to play an important role in the process of Leishmania infection.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil; Faculdade de Ciências da Saúde Pitágoras, Campus Codó, Codó, Maranhão, Brazil.
| | | | - Ana Lys Bezerra Barradas Mineiro
- Departamento de Clínica e Cirurgia Veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
6
|
Melzer YF, Fergen NL, Mess C, Stadler JC, Geidel G, Schwietzer YA, Kött J, Pantel K, Schneider SW, Utikal J, Wladykowski E, Vidal-Y-Sy S, Bauer AT, Gebhardt C. Evaluation of S100A8/A9 and neutrophils as prognostic markers in metastatic melanoma patients under immune-checkpoint inhibition. Transl Oncol 2025; 52:102224. [PMID: 39700646 PMCID: PMC11718343 DOI: 10.1016/j.tranon.2024.102224] [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: 07/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionized melanoma treatment, yet approximately half of patients do not respond to these therapies. Identifying prognostic biomarkers is crucial for treatment decisions. Our retrospective study assessed liquid biopsies and tumor tissue analyses for two potential biomarkers: danger-associated molecular pattern (DAMP) S100A8/A9 and its source, neutrophils. In 43 metastatic unresected stage III/IV melanoma patients, elevated serum levels of S100A8/A9 and neutrophils before and during ICI treatment correlated with worse outcomes. Furthermore, in 113 melanoma patients, neutrophil expression in the tumor microenvironment (TME) was associated with relapse and reduced survival. Measuring S100A8/A9 and neutrophils could enhance immunotherapy monitoring by predicting impaired clinical outcomes and non-response to ICIs. Serum S100A8/A9 levels and neutrophil counts at baseline (T0) and during treatment (T3) correlated with reduced progression-free survival (PFS). Elevated S100A8/A9 levels at T0 and T3 negatively impacted overall survival (OS). Notably, neutrophil infiltration was more prevalent in primary melanomas than in nevi and metastases, and its presence in primary melanomas was linked to poorer survival. S100A8/A9 serum levels, neutrophil counts, and tumor-associated neutrophil infiltration represent promising biomarkers for predicting treatment response and clinical outcomes in melanoma patients receiving ICIs. SIGNIFICANCE: These findings underscore the critical need for reliable biomarkers in melanoma research, particularly for predicting responses to immune-checkpoint inhibitors (ICIs). Identifying S100A8/A9 levels and neutrophil infiltration as potential indicators of treatment outcomes offers valuable insights for personalized therapy decisions. By enhancing monitoring and prognosis assessment, these biomarkers contribute to refining treatment strategies, ultimately improving patient care and outcomes. This research bridges gaps in understanding melanoma response mechanisms and highlights avenues for further investigation into immune-related markers, fostering advancements in precision medicine for melanoma patients.
Collapse
Affiliation(s)
- Yasmin F Melzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Nadine L Fergen
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Christian Mess
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julia-Christina Stadler
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Glenn Geidel
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Ysabel A Schwietzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julian Kött
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Klaus Pantel
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Ewa Wladykowski
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Alexander T Bauer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
7
|
Ehrens A, Lenz B, Nieto-Pérez C, Latz E, Schmidt FI, Hoerauf A, Hübner MP. Litomosoides sigmodontis microfilariae-induced eosinophil ETosis is dependent on the canonical inflammasome pathway. Cell Rep 2025; 44:115164. [PMID: 39772394 DOI: 10.1016/j.celrep.2024.115164] [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: 01/30/2024] [Revised: 09/03/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Granulocytes exert several effector mechanisms, including the release of DNA traps during ETosis. While bacteria-induced ETosis has been linked to the non-canonical inflammasome pathway, the role of the inflammasome activation during ETosis in response to extracellular pathogens has not been investigated. The current study demonstrates that microfilariae (MF) of the rodent filarial nematode Litomosoides sigmodontis induce eosinophil ETosis via the canonical inflammasome pathway. The absence of key components of the canonical inflammasome, including gasdermin D, caspase-1, the adaptor molecule ASC, or AIM2 (double-stranded DNA sensor) prevents MF-induced DNA release in murine eosinophils. While AIM2 activation is not affecting other effector mechanisms such as reactive oxygen species generation and nuclear membrane collapse, it appears to be critical in mediating the release of DNA from the cell during the later stages of ETosis. Finally, the findings on inflammasome-dependent ETosis in response to MF are confirmed in human eosinophils.
Collapse
Affiliation(s)
- Alexandra Ehrens
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany.
| | - Benjamin Lenz
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Celia Nieto-Pérez
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
8
|
Wang Z, Saxena A, Yan W, Uriarte SM, Siqueira R, Li X. The impact of aging on neutrophil functions and the contribution to periodontitis. Int J Oral Sci 2025; 17:10. [PMID: 39819982 PMCID: PMC11739572 DOI: 10.1038/s41368-024-00332-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/21/2024] [Accepted: 10/21/2024] [Indexed: 01/19/2025] Open
Abstract
The increasing aging population and aging-associated diseases have become a global issue for decades. People over 65 show an increased prevalence and greater severity of periodontitis, which poses threats to overall health. Studies have demonstrated a significant association between aging and the dysfunction of neutrophils, critical cells in the early stages of periodontitis, and their crosstalk with macrophages and T and B lymphocytes to establish the periodontal lesion. Neutrophils differentiate and mature in the bone marrow before entering the circulation; during an infection, they are recruited to infected tissues guided by the signal from chemokines and cytokines to eliminate invading pathogens. Neutrophils are crucial in maintaining a balanced response between host and microbes to prevent periodontal diseases in periodontal tissues. The impacts of aging on neutrophils' chemotaxis, anti-microbial function, cell activation, and lifespan result in impaired neutrophil functions and excessive neutrophil activation, which could influence periodontitis course. We summarize the roles of neutrophils in periodontal diseases and the aging-related impacts on neutrophil functional responses. We also explore the underlying mechanisms that can contribute to periodontitis manifestation in aging. This review could help us better understand the pathogenesis of periodontitis, which could offer novel therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Zi Wang
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anish Saxena
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA
| | - Wenbo Yan
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Rafael Siqueira
- Department of Periodontics, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| | - Xin Li
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA.
- Comprehensive Cancer Center, University of Virginia, Charlottesville, USA.
| |
Collapse
|
9
|
Englert H, Rangaswamy C, Kullik GA, Divivier M, Göbel J, Hermans‐Borgmeyer I, Borgmeyer U, Mowen KA, Beerens M, Frye M, Mailer RK, Gelderblom M, Stavrou EX, Preston RJS, Schneider SW, Fuchs TA, Renné T. Sepsis-induced NET formation requires MYD88 but is independent of GSDMD and PAD4. FASEB J 2025; 39:e70301. [PMID: 39777764 PMCID: PMC11707982 DOI: 10.1096/fj.202402514r] [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: 10/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025]
Abstract
Neutrophils are peripheral blood-circulating leukocytes that play a pivotal role in host defense against bacterial pathogens which upon activation, they release web-like chromatin structures called neutrophil extracellular traps (NETs). Here, we analyzed and compared the importance of myeloid differentiation factor 88 (MYD88), peptidyl arginine deiminase 4 (PAD4), and gasdermin D (GSDMD) for NET formation in vivo following sepsis and neutrophilia challenge. Injection of lipopolysaccharide (LPS)/E. coli or the transgenic expression of granulocyte colony-stimulating factor (G-CSF), each induced NET-mediated lethal vascular occlusions in mice with combined genetic deficiency in Dnase1 and Dnase1l3 (D1/D1l3-/-). In accordance with the signaling of toll-like receptors, Myd88/D1/D1l3-/- animals were protected from the formation of lethal intravascular NETs during septic conditions. However, this protection was not observed during neutrophilia. It was unexpected to find that both Gsdmd/D1/D1l3-/- and Pad4/D1/D1l3-/- mice were fully capable of forming NETs upon LPS/E.coli challenge. Sepsis equally triggered a similar inflammatory response in these mice characterized by formation of DNA-rich thrombi, vessel occlusions, and mortality from pulmonary embolism, compared to D1/D1l3-/- mice. Pharmacologic GSDMD inhibitors did not reduce PMA-stimulated NET formation in ex vivo models either. Similarly, neither Pad4 nor GSDMD deficiency affected intravascular occlusive NET formation upon neutrophilia challenge. The magnitude of NET production, multi-organ damage, and lethality were comparable to those observed in challenged control mice. In conclusion, our data indicate that NET formation during experimental sepsis and neutrophilia is regulated by distinct stimulus-dependent pathways that may be independent of canonical PAD4 and GSDMD.
Collapse
Affiliation(s)
- Hanna Englert
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Giuliano A. Kullik
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mylène Divivier
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Josephine Göbel
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Irm Hermans‐Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Uwe Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Kerri A. Mowen
- Chemical PhysiologyThe Scripps InstituteLa JollaCaliforniaUSA
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Reiner K. Mailer
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mathias Gelderblom
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Evi X. Stavrou
- Medicine Service, Section of Hematology‐OncologyLouis Stokes Veterans Administration Medical CenterClevelandOhioUSA
- Department of Medicine, Hematology and Oncology DivisionCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Roger J. S. Preston
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Stefan W. Schneider
- Department of Dermatology and VenereologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tobias A. Fuchs
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- NeutrolisCambridgeMassachusettsUSA
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
- Center for Thrombosis and Hemostasis (CTH)Johannes Gutenberg University Medical CenterMainzGermany
| |
Collapse
|
10
|
Iba T, Helms J, Maier CL, Ferrer R, Levy JH. Mitochondrial dysfunction is a major cause of thromboinflammation and inflammatory cell death in critical illnesses. Inflamm Res 2025; 74:17. [PMID: 39806233 DOI: 10.1007/s00011-025-01994-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/02/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Mitochondria generate the adenosine triphosphate (ATP) necessary for eukaryotic cells, serving as their primary energy suppliers, and contribute to host defense by producing reactive oxygen species. In many critical illnesses, including sepsis, major trauma, and heatstroke, the vicious cycle between activated coagulation and inflammation results in tissue hypoxia-induced mitochondrial dysfunction, and impaired mitochondrial function contributes to thromboinflammation and cell death. METHODS A computer-based online search was performed using the PubMed and Web of Science databases for published articles concerning sepsis, trauma, critical illnesses, cell death, mitochondria, inflammation, coagulopathy, and organ dysfunction. RESULTS Mitochondrial outer membrane permeabilization triggers apoptosis by releasing cytochrome c and activating caspases. Apoptosis is a non-inflammatory programmed cell death but requires sufficient ATP supply. Therefore, conversion to inflammatory necrosis may occur due to a lack of ATP in critical illness. Severely damaged mitochondria release excess reactive oxygen species and injurious mitochondrial DNA, inducing cell death. Besides non-programmed necrosis, mitochondrial damage can trigger programmed inflammatory cell death, including necroptosis, pyroptosis, and ferroptosis. Additionally, a unique form of DNA-ejecting cell death, known as etosis, occurs in monocytes and granulocytes following external stimuli and mitochondrial damage. The type of cell death chosen remains uncertain but is known to depend on the cell type, the nature of the injury, and the degree of damage. CONCLUSIONS Mitochondria damage is the major contributor to the cell death mechanism that leads to organ damage in critical illnesses. Regulating and restoring mitochondrial function holds promise for developing new therapeutic approaches for mitigating critical diseases.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Julie Helms
- Medical Intensive Care Unit - NHC, Strasbourg University (UNISTRA), Strasbourg University Hospital, INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ricard Ferrer
- Intensive Care Department, Hospital Universitari Vall d'Hebron Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
11
|
Dai SZ, Wu RH, Chen H, Chen MH, Xie W, Zheng WP, Tan GH, Huang FY. Progesterone suppresses rhinovirus-induced airway inflammation by inhibiting neutrophil infiltration and extracellular traps formation. Int Immunopharmacol 2025; 144:113714. [PMID: 39626540 DOI: 10.1016/j.intimp.2024.113714] [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: 07/23/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND The process of NETosis is observed in a range of inflammatory conditions. Progesterone (P4) has been shown to alleviate inflammation caused by viral infections such as influenza and SARS-CoV-2. However, the precise molecular mechanisms responsible for this effect are not yet fully understood. Therefore, the present investigation aims to explore whether P4 can exert its anti-inflammatory properties by inhibiting NETosis and the related molecular pathways. METHODS Airway inflammation caused by rhinovirus serotype-1b (RV-1b) was induced in male BALB/c mice. The inflammation was assessed through histological examination and calculation of inflammatory cells present in the bronchoalveolar lavage fluid. Flow cytometry was used to analyze the inflammatory cells and NETotic neutrophils. Western blotting analysis was conducted to detect proteins associated with NETosis, inflammasome activation, and signaling. Furthermore, confocal microscopy was utilized to observe neutrophil extracellular trap (NET) structures in vivo tissues and in vitro neutrophils, neutrophil infiltration, and inflammasome formation. RESULTS The administration of P4 proved to be an effective treatment for reducing airway inflammation and the production of NETs caused by RV-1b infection. The infection triggered the activation of NLRP3 inflammasomes in neutrophils, which led to the maturation of IL-1β and subsequent activation of both the NF-κB and p38 signaling pathways. The activation of NF-κB signaling resulted in the secretion of downstream chemokines CCL3 and IL-6, which led to an increase in neutrophil infiltration into the lung airways. Moreover, the activation of p38 signaling led to the generation of reactive oxygen species, resulting in NETosis. However, the administration of P4 inhibited the activation of the NLRP3 inflammasome, which subsequently led to the deactivation of both the IL-1β-NF-κB and IL-1β-p38 axes. As a result, there was a reduction in neutrophil infiltration and NETosis. Furthermore, TGF-β-activated kinase 1 (TAK1) was identified as an intermediary enzyme. P4 inhibits both the NF-κB and IL-1β-p38 pathways by suppressing the activity of TAK1. CONCLUSION The capacity of P4 to mitigate rhinovirus-induced airway inflammation is attributed to its ability to impede the infiltration of neutrophils and NETosis. As inflammation mediated by NETosis is widespread in diverse disorders, our findings propose that P4 could potentially function as a universal therapeutic agent in the management of such ailments.
Collapse
Affiliation(s)
- Shu-Zhen Dai
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China; Hainan Academy of Medical Sciences, Hainan Medical University, Hainan 571199, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Hengyu Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ming-Hui Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Weijing Xie
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Wu-Ping Zheng
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Feng-Ying Huang
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
12
|
Mercado-Evans V, Branthoover H, Chew C, Serchejian C, Saltzman AB, Mejia ME, Zulk JJ, Cornax I, Nizet V, Patras KA. Tamm-Horsfall protein augments neutrophil NETosis during urinary tract infection. JCI Insight 2025; 10:e180024. [PMID: 39589812 PMCID: PMC11721310 DOI: 10.1172/jci.insight.180024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Urinary neutrophils are a hallmark of urinary tract infection (UTI), yet the mechanisms governing their activation, function, and efficacy in controlling infection remain incompletely understood. Tamm-Horsfall glycoprotein (THP), the most abundant protein in urine, uses terminal sialic acids to bind an inhibitory receptor and dampen neutrophil inflammatory responses. We hypothesized that neutrophil modulation is an integral part of THP-mediated host protection. In a UTI model, THP-deficient mice showed elevated urinary tract bacterial burdens, increased neutrophil recruitment, and more severe tissue histopathological changes compared with WT mice. Furthermore, THP-deficient mice displayed impaired urinary NETosis during UTI. To investigate the effect of THP on NETosis, we coupled in vitro fluorescence-based NET assays, proteomic analyses, and standard and imaging flow cytometry with peripheral human neutrophils. We found that THP increases proteins involved in respiratory chain, neutrophil granules, and chromatin remodeling pathways; enhances NETosis in an ROS-dependent manner; and drives NET-associated morphologic features including nuclear decondensation. These effects were observed only in the presence of a NETosis stimulus and could not be solely replicated with equivalent levels of sialic acid alone. We conclude that THP is a critical regulator of NETosis in the urinary tract, playing a key role in host defense against UTI.
Collapse
Affiliation(s)
- Vicki Mercado-Evans
- Department of Molecular Virology and Microbiology
- Medical Scientist Training Program
| | | | | | | | - Alexander B. Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine (BCM), Houston, Texas, USA
| | | | | | | | - Victor Nizet
- Department of Pediatrics and
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology
- Department of Pediatrics and
- Alkek Center for Metagenomics and Microbiome Research, BCM, Houston, Texas, USA
| |
Collapse
|
13
|
Liao R, Wang L, Zeng J, Tang X, Huang M, Kantawong F, Huang Q, Mei Q, Huang F, Yang Y, Liao B, Wu A, Wu J. Reactive oxygen species: Orchestrating the delicate dance of platelet life and death. Redox Biol 2025; 80:103489. [PMID: 39764976 PMCID: PMC11759559 DOI: 10.1016/j.redox.2025.103489] [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: 11/16/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Platelets, which are vital for blood clotting and immunity, need to maintain a delicately balanced relationship between generation and destruction. Recent studies have highlighted that reactive oxygen species (ROS), which act as second messengers in crucial signaling pathways, are crucial players in this dance. This review explores the intricate connection between ROS and platelets, highlighting their dual nature. Moderate ROS levels act as potent activators, promoting megakaryocyte (MK) differentiation, platelet production, and function. They enhance platelet binding to collagen, increase coagulation, and directly trigger cascades for thrombus formation. However, this intricate role harbors a double-edged sword. Excessive ROS unleash its destructive potential, triggering apoptosis and reducing the lifespan of platelets. High levels can damage stem cells and disrupt vital redox-dependent signaling, whereas uncontrolled activation promotes inappropriate clotting, leading to thrombosis. Maintaining a precise balance of ROS within the hematopoietic microenvironment is paramount for optimal platelet homeostasis. While significant progress has been made, unanswered questions remain concerning specific ROS signaling pathways and their impact on platelet disorders. Addressing these questions holds the key to unlocking the full potential of ROS-based therapies for treating platelet-related diseases such as thrombocytopenia and thrombosis. This review aims to contribute to this ongoing dialog and inspire further exploration of this exciting field, paving the way for novel therapeutic strategies that harness the benefits of ROS while mitigating their dangers.
Collapse
Affiliation(s)
- Rui Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jing Zeng
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoqin Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Miao Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qianqian Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qibing Mei
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Feihong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
14
|
da Costa AAS, Oliveira SR, Tavares TS, Meirelles DP, da Silva EV, da Silva ATF, León JE, Cardoso SV, de Aguiar MCF, da Silva TA, Caldeira PC. Contribution of HPV Status for Neutrophil Extracellular Traps Release in Oropharyngeal Cancer. J Oral Pathol Med 2025; 54:57-64. [PMID: 39581629 DOI: 10.1111/jop.13594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Oropharyngeal squamous cell carcinoma (OP-SCC) represents a public health problem and human papillomavirus (HPV) is one of the risk factors. Neutrophil extracellular traps (NET) are meshes of DNA strands and granule proteins. NET has been identified in diverse cancers, whether associated with viruses or not. However, there is no information on NET in OP-SCC. We aimed to evaluate the NET release by neutrophils in the OP-SCC microenvironment, stratified by HPV status. METHODS This cross-sectional study analyzed OP-SCC biopsy specimens diagnosed from 1997 to 2021. HPV status was determined by p16 immunohistochemistry and "in situ" hybridization. Neutrophils were detected by CD66b immunohistochemistry. Immunofluorescence was used to identify NET by co-localization of myeloperoxidase (MPO) and citrullinated histone H3 (H3Cit). Bivariate statistics, Kaplan-Meier survival analysis, and the log-rank test were performed. RESULTS HPV-positive and HPV-negative OP-SCC had similar CD66b + neutrophil infiltration (p > 0.05), but the release of NET was significantly increased in HPV-positive compared to HPV-negative OP-SCC samples (p < 0.05). Overall survival was not impacted by NET indexes (p > 0.05). CONCLUSION The presence of HPV may stimulate NET release in the OP-SCC microenvironment.
Collapse
Affiliation(s)
- Adriana Aparecida Silva da Costa
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Sicília Rezende Oliveira
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Thalita Soares Tavares
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela Pereira Meirelles
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Evânio Vilela da Silva
- Oral Pathology, Department of Stomatology, Public Oral Health, and Forensic Dentistry, School of Dentistry, Universidade de São Paulo (USP). Av. do Café - Subsetor Oeste, São Paulo, Brazil
| | - Anderson Tangerino Ferreira da Silva
- Oral Pathology, Department of Stomatology, Public Oral Health, and Forensic Dentistry, School of Dentistry, Universidade de São Paulo (USP). Av. do Café - Subsetor Oeste, São Paulo, Brazil
| | - Jorge Esquiche León
- Oral Pathology, Department of Stomatology, Public Oral Health, and Forensic Dentistry, School of Dentistry, Universidade de São Paulo (USP). Av. do Café - Subsetor Oeste, São Paulo, Brazil
| | - Sérgio Vitorino Cardoso
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Universidade Federal de Uberlândia (UFU). R. República Do Piratini, Uberlândia, Minas Gerais, Brazil
| | - Maria Cássia Ferreira de Aguiar
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Tarcília Aparecida da Silva
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Patrícia Carlos Caldeira
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais (UFMG). Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
15
|
Zhang S, Wang Z, Zhang Y, Dong X, Zhu Q, Yuan C, Lu G, Gong W, Bi Y, Wang Y. LASP1 inhibits the formation of NETs and alleviates acute pancreatitis by stabilizing F-actin polymerization in neutrophils. Biochem Biophys Res Commun 2025; 744:151134. [PMID: 39708397 DOI: 10.1016/j.bbrc.2024.151134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) play a significant role in the development of acute pancreatitis (AP). The actin-binding protein LASP1 regulates proteins associated with the cytoskeleton, yet its precise involvement in NETs and AP remains to be elucidated. METHODS To investigate the role of LASP1 in NETs and AP, several bioinformatics methods, such as weighted gene co-expression network analysis (WGCNA), differential analysis, and least absolute shrinkage and selection operator (LASSO) regression, were utilized to screen for feature genes based on the Gene Expression Omnibus (GEO) dataset. To further assess the impact of LASP1, both an in vitro model of 12-myristic-13-acetate phobolol (PMA)-induced NETs and a caerulein-induced AP model were employed. RESULTS Through WGCNA, AP-related module genes were screened, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were conducted to identify enriched pathways and functions. Six characteristic genes were identified through LASSO regression screening, with LASP1 being the most distinct. LASP1 reduces the generation of NETs induced by PMA in vitro. Mechanistically, LASP1 may increase F-actin protein levels by inhibiting the depolymerization of F-actin. Furthermore, our study utilizing a mouse AP model demonstrated that the LSAP1 recombinant protein effectively alleviated pancreatic necrosis in mice afflicted with AP. CONCLUSION LASP1 inhibits the formation of NETs and may alleviate AP by increasing the level of F-actin protein.
Collapse
Affiliation(s)
- Siqin Zhang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Yangzhou University, Kunshan, Suzhou, Jiangsu, China; Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhihao Wang
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuyan Zhang
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaowu Dong
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qingtian Zhu
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chenchen Yuan
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guotao Lu
- Department of Gastroenterology, Pancreatic Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weijuan Gong
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yawei Bi
- Department of Gastroenterology, Chinese PLA General Hospital First Medical Center, Beijing, China
| | - Yaodong Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Yangzhou University, Kunshan, Suzhou, Jiangsu, China.
| |
Collapse
|
16
|
Medhasi S, Sriwarom A, Permpalung N, Torvorapanit P, Plongla R, Chindamporn A, Worasilchai N. Ex vivo observation of Pythium insidiosum-antigen treated neutrophils on three Pythium insidiosum strains isolated from vascular pythiosis patients. Hum Vaccin Immunother 2024; 20:2304372. [PMID: 38314761 PMCID: PMC10854268 DOI: 10.1080/21645515.2024.2304372] [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: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
The mechanisms of Pythium insidiosum-antigen (PIA) immunotherapy activating a patient's immune system are unknown. We evaluated the interleukin-8 (IL-8) serum levels during P. insidiosum infection and after vaccination with PIA in vascular pythiosis cases. Furthermore, we studied the anti-P. insidiosum activity of neutrophils stimulated with various concentrations of PIA ex vivo in 3 strains of P. insidiosum isolated from vascular pythiosis patients. IL-8 serum levels were evaluated using the ELISA technique. We assessed the effect of PIA-stimulated neutrophils on the viability of zoospores using MTT assay, visualized neutrophil extracellular trap (NET) formation via microscopy, and measured the levels of double-stranded DNA (dsDNA) using PicoGreen dsDNA quantitation assay in 3 strains of P. insidiosum isolated from vascular pythiosis patients. Serum levels of IL-8 gradually lowered from the early to the end phases of vaccination with PIA among the surviving group of vascular pythiosis cases. Neutrophils stimulated with 0.01 µg/ml PIA reduced zoospore viability significantly compared to PIA-unstimulated neutrophils for strain 1 and strain 3 (p < .05). Neutrophils stimulated with 0.01, 0.1, 1, and 10 µg/ml PIA exhibited significantly lower zoospore viability than PIA-unstimulated neutrophils for strain 2 (p < .05). IL-8 can be used as a biomarker for monitoring vascular pythiosis cases treated with the PIA vaccine. Also, anti-P. insidiosum activity of PIA-stimulated neutrophils was probably due to the disruption of cellular activity in zoospores rather than the mechanisms based on the formation of NETs.
Collapse
Affiliation(s)
- Sadeep Medhasi
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand
| | - Apichaya Sriwarom
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nitipong Permpalung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pattama Torvorapanit
- Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Rongpong Plongla
- Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Ariya Chindamporn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Navaporn Worasilchai
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Tang J, Yue J, Tao Y, Zhao G, Yi X, Zhang M, Huang N, Cheng Y. Neutrophil Extracellular Traps Induce Brain Edema Around Intracerebral Hematoma via ERK-Mediated Regulation of MMP9 and AQP4. Transl Stroke Res 2024:10.1007/s12975-024-01318-w. [PMID: 39733198 DOI: 10.1007/s12975-024-01318-w] [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: 09/15/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024]
Abstract
Perihematomal edema (PHE) significantly aggravates secondary brain injury in patients with intracerebral hemorrhage (ICH), yet its detailed mechanisms remain elusive. Neutrophil extracellular traps (NETs) are known to exacerbate neurological deficits and worsen outcomes after stroke. This study explores the potential role of NETs in the pathogenesis of brain edema following ICH. The rat ICH model was created, immunofluorescence and Western blot were used to examine neutrophil accumulation, NET markers citrullinated histone H3 (CitH3) and myeloperoxidase (MPO), tight junction proteins (ZO-1 and Occludin), Aquaporin-4 (AQP4), matrix metalloproteinase-9 (MMP-9), and ERK phosphorylation (p-ERK) in brain tissues surrounding the hematoma. TUNEL staining and behavioral tests were employed to evaluate neuronal apoptosis and neurological dysfunction, while blood-brain barrier (BBB) permeability and brain edema were also measured by Evans blue and brain water content. Furthermore, the molecular mechanisms related to NETs-induced PHE were investigated using NETs, ERK, MMP-9 and AQP4 regulators, respectively. Ly6G+ neutrophils surrounding the hematoma developed NETs within 3 days post-ICH. NETs decreased tight junction proteins, destroyed BBB integrity, promoted brain edema, increased neuronal apoptosis, and exacerbated neurological deficits. Conversely, inhibition of NETs mitigated PHE, reduced neuronal apoptosis, and improved neurological functions. Mechanistically, NET-induced PHE was originated from impairment of BBB tight junction via ERK/MMP9 pathway, coupled with ERK-mediated AQP4 downregulation in perihematomal regions. These findings elucidated the effects of NETs on PHE, which offered promising insights for targeting NETs to relieve brain edema and secondary brain injury post-ICH.
Collapse
Affiliation(s)
- Jun Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Jianhe Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Xiaoyao Yi
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Maoxin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China.
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Rd, Yuzhong, Chongqing, 400010, China.
| |
Collapse
|
18
|
Zhang J, Xie X, Shen Q, Yuan C, Lu G, Xiao W, Gong W, Fu X, Feng X. Rhaponticin Alleviates Collagen-induced Arthritis by Inhibiting NLRP3/GSDMD-mediated Neutrophil Extracellular Traps. Inflammation 2024:10.1007/s10753-024-02228-7. [PMID: 39725843 DOI: 10.1007/s10753-024-02228-7] [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: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Neutrophil extracellular traps (NETs) play an important role in the inflammatory response and progressive joint destruction in rheumatoid arthritis (RA). Rhaponticin (Rha) is a stilbene glycoside compound with antioxidant and anti-inflammatory effects. This study aimed to investigate the therapeutic potential of Rha in RA, with a specific focus on its effects on NETs and on the underlying mechanisms of Rha. NETs formation induced by phorbol 12-myristate 13-acetate (PMA) and a collagen-induced arthritis (CIA) mouse model were implemented to evaluate the pharmacological effects of Rha in vitro and in vivo. The potential mechanism of Rha in improving RA was screened and verified using the SuperPred and DisGeNET databases. Disulfiram (a GSDMD inhibitor) and S100a8cre GSDMDfl/fl mice were used to confirm whether GSDMD is key to the role of Rha. The findings demonstrate that Rha significantly inhibited reactive oxygen species and NETs production in PMA-activated neutrophils. In vivo, Rha treatment significantly relieved joint symptoms in CIA mice and NETs production. Mechanistically, Rha reduced NETs production via inhibition of NLRP3/GSDMD activation. Neutrophil-specific GSDMD depletion eliminated the effects of Rha on NETs production in vitro. Disulfiram eliminated the effects of Rha on the inhibition of NETs production and alleviated joint inflammation in mice in vivo and in vitro. Overall, our results indicated that Rha exerts a protective effect against CIA by inhibiting NETs production through the NLRP3/GSDMD pathway. The results of this study provide new strategies for treating RA.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Xinyue Xie
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Qinhao Shen
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Chenchen Yuan
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Guotao Lu
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weiming Xiao
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weijuan Gong
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Xiaoyan Fu
- Department of Rehabilitation, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Yangzhou University, Kunshan, 215300, China.
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
| |
Collapse
|
19
|
Lagorgette L, Bogdanova DA, Belotserkovskaya EV, Garrido C, Demidov ON. PP2C phosphatases-terminators of suicidal thoughts. Cell Death Dis 2024; 15:919. [PMID: 39702569 DOI: 10.1038/s41419-024-07269-2] [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: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Cell death and related signaling pathways are essential during development and in various physiological and pathological conditions. Post-translational modifications such as ubiquitination and phosphorylation play an important role in these signaling pathways. The involvement of kinases - enzymes that catalyze protein phosphorylation - in cell death signaling has been extensively studied. On the other hand, not many studies have been devoted to analyzing the role in cell death of phosphatases, enzymes involved in the removal of phosphorylated residues added to proteins by kinases. Obviously, the two opposite reactions, phosphorylation and dephosphorylation, are equally important in the regulation of protein functions and subsequently in the execution of the cell death program. Here, we have summarized recent work on the involvement of serine-threonine PP2C phosphatases in cell death pathways, senescence and autophagy, focusing in particular on the most studied phosphatase PPM1D (PP2Cδ) as an example of the regulatory role of PP2Cs in cell death. The review should help to draw attention to the importance of PP2C family phosphatases in cell death checkpoints and to discover new targets for drug development.
Collapse
Affiliation(s)
- Lisa Lagorgette
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
| | - Daria A Bogdanova
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia
- Institute of Cytology RAS, St. Petersburg, Russia
| | | | - Carmen Garrido
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
- Center for Cancer Georges-François Leclerc, Dijon, France
| | - Oleg N Demidov
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France.
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia.
- Institute of Cytology RAS, St. Petersburg, Russia.
| |
Collapse
|
20
|
Espinosa G, Salinas-Varas C, Rojas-Barón L, Preußer C, Pogge von Strandmann E, Gärtner U, Conejeros I, Hermosilla C, Taubert A. Bovine PMN responses to extracellular vesicles released by Besnoitia besnoiti tachyzoites and B. besnoiti-infected host cells. Front Immunol 2024; 15:1509355. [PMID: 39749330 PMCID: PMC11693690 DOI: 10.3389/fimmu.2024.1509355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Bovine besnoitiosis is a re-emerging cattle disease caused by the apicomplexan parasite Besnoitia besnoiti, which severely affects individual animal welfare and profitability in cattle industry. We recently showed that B. besnoiti tachyzoite exposure to bovine polymorphonuclear neutrophils (PMN) effectively triggers neutrophil extracellular trap (NET) formation, leading to parasite immobilization hampering host cell infection. So far, the triggers of this defense mechanism remain unclear. Emerging evidence indicates that extracellular vesicles (EVs) modulate PMN effector functions, such as ROS production or NET formation. Therefore, we tested whether exposure of bovine PMN to EVs from different cellular sources affects classical PMN effector functions and cytokine/chemokine secretion. EVs were isolated from B. besnoiti-infected and non-infected host cells (bovine umbilical vein endothelial cells, BUVEC), from tachyzoite-exposed bovine PMN and from B. besnoiti tachyzoites. EV concentration and size was determined by Nano-Flow cytometry and EV nature was confirmed by both classical EV markers (CD9 and CD81) and transmission electron microscopy (TEM). Overall, PMN stimulation with both BUVEC- and tachyzoite-derived EVs significantly induced extracellular DNA release while EVs from PMN failed to affect NET formation. BUVEC and tachyzoite EV-driven NET formation was confirmed microscopically by the presence of DNA decorated with neutrophil elastase (NE) and histones in typical NET structures. Moreover, confocal microscopy revealed EVs to be internalized by bovine PMN. Referring to PMN activation, EVs from the different cellular sources all failed to affect glycolytic or oxidative responses of bovine PMN as detected by Seahorse®-based analytics and luminol-based chemoluminescence, thereby denying any role of NADPH oxidase (NOX) activity in EV-driven NET formation. Finally, exposure to B. besnoiti-infected BUVEC-derived EVs induced IL-1β and IL-6 release, but failed to drive CXCL8 release of bovine PMN. Hence, we overall demonstrated that EVs of selected cellular origin owned the capacity to trigger NOX-independent NET formation, were incorporated by PMN and selectively fostered IL-1β and IL-6 release.
Collapse
Affiliation(s)
- Gabriel Espinosa
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | | | - Lisbeth Rojas-Barón
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Preußer
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Elke Pogge von Strandmann
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Iván Conejeros
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
21
|
Bi J, Chen Y, Zhang J, Yan J, Ge A, Ye W, Liu C, Wen H, Ma C. Causal relationship between immune cells and periodontitis: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40918. [PMID: 39686447 DOI: 10.1097/md.0000000000040918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
This study employed Mendelian randomization (MR) analysis to explore potential causal relationships between 731 immune cell subtypes and periodontitis. Utilizing a 2-sample MR design, our study delved into the diverse landscape of immune cell interactions with periodontitis-associated factors. Multiple MR methods, including inverse variance weighting, weighted median, and MR-Egger tests, were employed to ensure reliability and mitigate potential pleiotropic effects. The study revealed significant causal effects (FDR < 0.15) between immune cells (B cells, maturation stages of T cells, Treg) and periodontitis. Notably, receptors like triggering receptor expressed on myeloid cells-1 (TREM-1) and triggering receptor expressed on myeloid cells-2 (TREM-2) exhibited intricate roles, warranting further investigation. In conclusion, this MR analysis elucidates complex causal relationships between immune cell subtypes and periodontitis. The findings provide a foundation for understanding systemic implications, offering insights for clinical practice and highlighting avenues for future research.
Collapse
Affiliation(s)
- Junlei Bi
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Yuxin Chen
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Jie Zhang
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Jiaqi Yan
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Aiyun Ge
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Wenhao Ye
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Changqing Liu
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Hebao Wen
- Physical Education Department, Bengbu Medical University, Bengbu City, Anhui Province, China
| | - Caiyun Ma
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, School of Life Science, Bengbu Medical University, Bengbu City, Anhui Province, China
| |
Collapse
|
22
|
Chowdhury CS, Kinsella RL, McNehlan ME, Naik SK, Lane DS, Talukdar P, Smirnov A, Dubey N, Rankin AN, McKee SR, Woodson R, Hii A, Chavez SM, Kreamalmeyer D, Beatty W, Mattila JT, Stallings CL. Type I IFN-mediated NET release promotes Mycobacterium tuberculosis replication and is associated with granuloma caseation. Cell Host Microbe 2024; 32:2092-2111.e7. [PMID: 39637864 PMCID: PMC11637906 DOI: 10.1016/j.chom.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Neutrophils are the most abundant cell type in the airways of tuberculosis patients. Mycobacterium tuberculosis (Mtb) infection induces the release of neutrophil extracellular traps (NETs); however, the molecular regulation and impact of NET release on Mtb pathogenesis are unknown. We find that during Mtb infection in neutrophils, PAD4 citrullinates histones to decondense chromatin that gets released as NETs in a manner that can maintain neutrophil viability and promote Mtb replication. Type I interferon promotes the formation of chromatin-containing vesicles that allow NET release without compromising plasma membrane integrity. Analysis of nonhuman primate granulomas supports a model where neutrophils are exposed to type I interferon from macrophages as they migrate into the granuloma, thereby enabling the release of NETs associated with necrosis and caseation. Our data reveal NET release as a promising target to inhibit Mtb pathogenesis.
Collapse
Affiliation(s)
- Chanchal Sur Chowdhury
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Kinsella
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael E McNehlan
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sumanta K Naik
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel S Lane
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Priyanka Talukdar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Asya Smirnov
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Neha Dubey
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ananda N Rankin
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel R McKee
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reilly Woodson
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Abigail Hii
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA; UCD School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Sthefany M Chavez
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Darren Kreamalmeyer
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Yipeng Z, Chao C, Ranran L, Tingting P, Hongping Q. Metabolism: a potential regulator of neutrophil fate. Front Immunol 2024; 15:1500676. [PMID: 39697327 PMCID: PMC11652355 DOI: 10.3389/fimmu.2024.1500676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are essential components of the innate immune system that defend against the invading pathogens, such as bacteria, viruses, and fungi, as well as having regulatory roles in various conditions, including tissue repair, cancer immunity, and inflammation modulation. The function of neutrophils is strongly related to their mode of cell death, as different types of cell death involve various cellular and molecular alterations. Apoptosis, a non-inflammatory and programmed type of cell death, is the most common in neutrophils, while other modes of cell death, including NETOsis, necrosis, necroptosis, autophagy, pyroptosis, and ferroptosis, have specific roles in neutrophil function regulation. Immunometabolism refers to energy and substance metabolism in immune cells, and profoundly influences immune cell fate and immune system function. Intercellular and intracellular signal transduction modulate neutrophil metabolism, which can, in turn, alter their activities by influencing various cell signaling pathways. In this review, we compile an extensive body of evidence demonstrating the role of neutrophil metabolism in their various forms of cell death. The review highlights the intricate metabolic characteristics of neutrophils and their interplay with various types of cell death.
Collapse
Affiliation(s)
| | | | | | - Pan Tingting
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| | - Qu Hongping
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Jorge-Rosas F, Díaz-Godínez C, García-Aguirre S, Martínez-Calvillo S, Carrero JC. Entamoeba histolytica-induced NETs are highly cytotoxic on hepatic and colonic cells due to serine proteases and myeloperoxidase activities. Front Immunol 2024; 15:1493946. [PMID: 39687618 PMCID: PMC11646992 DOI: 10.3389/fimmu.2024.1493946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
During intestinal and liver invasion by the protozoan parasite Entamoeba histolytica, extensive tissue destruction linked to large neutrophil infiltrates is observed. It has been proposed that microbicidal components of neutrophils are responsible for the damage, however, the mechanism by which they are released and act in the extracellular space remains unknown. In previous studies, we have shown that E. histolytica trophozoites induce NET formation, leading to the release of neutrophil granule content into extruded DNA. In this work, we evaluate the possible participation of NETs in the development of amoeba-associated pathology and analyze the contribution of anti-microbial components of the associated granules. E. histolytica-induced NETs were isolated and their effect on the viability and integrity of HCT 116 colonic and Hep G2 liver cultures were evaluated. The results showed that simple incubation of cell monolayers with purified NETs for 24 h resulted in cell detachment and death in a dose-dependent manner. The effect was thermolabile and correlated with the amount of DNA and protein present in NETs. Pretreatment of NETs with specific inhibitors of some microbicidal components suggested that serine proteases, are mostly responsible for the damage caused by NETs on HCT 116 cells, while the MPO activity was the most related to Hep G2 cells damage. Our study also points to a very important role of DNA as a scaffold for the activity of these proteins. We show evidence of the development of NETs in amoebic liver abscesses in hamsters as a preamble to evaluate their participation in tissue damage. In conclusion, these studies demonstrate that amoebic-induced NETs have potent cytotoxic effects on target cells and, therefore, may be responsible for the intense damage associated with tissue invasion by this parasite.
Collapse
Affiliation(s)
- Fabian Jorge-Rosas
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - César Díaz-Godínez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Samuel García-Aguirre
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Santiago Martínez-Calvillo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, EM, Mexico
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
25
|
Li J, Zhao H, Yang J, Wang M, Cao Z, Wang Y, Gu Z. The role and mechanism of extracellular traps in chronic rhinosinusitis. Biomed Pharmacother 2024; 181:117655. [PMID: 39486368 DOI: 10.1016/j.biopha.2024.117655] [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: 07/20/2024] [Revised: 09/24/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Chronic rhinosinusitis (CRS) is a common inflammatory disease of the nose that affects millions of individuals worldwide. Recent research has introduced the concept of an immunologic endotype based on the pathological characteristics of CRS and the types of inflammatory cell infiltration. This endotype concept is conducive to understanding CRS pathology and guiding further targeted therapy. Eosinophils and neutrophils infiltrate different proportions in different CRS endotypes and release extracellular traps (ETs) as a response to the extracellular immune response. The mechanisms of formation and biological roles of ETs are complex. ETs can trap extracellular microorganisms and limit the range of inflammation to some extent; however, excessive and long-term ETs may be related to disease severity. This review summarises and explores the mechanism of ETs and the advances in CRS research and proposes new insights into the interaction between ETs and programmed cell death (including autophagy, pyroptosis, and necroptosis) in CRS, providing new ideas for the targeted therapy of CRS.
Collapse
Affiliation(s)
- Jiani Li
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - He Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Meng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Zhiwei Cao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Yunxiu Wang
- Department of Clinical Trial Ward, Clinical Trial and Conversion Center, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Zhaowei Gu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
26
|
Wang N, Shi XL, Li D, Li BB, Liu P, Luo H. Neutrophil extracellular traps - an a-list-actor in a variety of diseases. Ann Hematol 2024; 103:5059-5069. [PMID: 39078437 DOI: 10.1007/s00277-024-05915-5] [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: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024]
Abstract
Neutrophil extracellular traps (NETs) represent a response mechanism in which activated neutrophils release DNA-based webs, adorned with histones and neutrophil proteases, to capture and eliminate invasive microorganisms. However, when these neutrophils become excessively activated, much more proteases associated with NETs are liberated into surrounding tissues or bloodstreams, thereby altering the cellular milieu and causing tissue damage. Recent research has revealed that NETs may play significant roles in the emergence and progression of various diseases, spanning from infections, inflammation to autoimmune disorders and cancers. In this review, we delve deeply into the intricate and complex mechanisms that underlie the formation of NETs and their profound interplay with various clinical pathologies. We aim to describe the application perspectives of NETs related proteins in specific disease diagnosis and treatment.
Collapse
Affiliation(s)
- Na Wang
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning, 116044, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Xiao-Lin Shi
- Department of Clinical Laboratory, Weihai Maternal and Child Health Hospital, Weihai, Shandong, 264200, PR China
| | - Dan Li
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Bin-Bin Li
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China.
| | - Hong Luo
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning, 116044, PR China.
| |
Collapse
|
27
|
Prendecki M, Gurung A, Pisacano N, Pusey CD. The role of neutrophils in ANCA-associated vasculitis. Immunol Lett 2024; 270:106933. [PMID: 39362307 DOI: 10.1016/j.imlet.2024.106933] [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: 07/24/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) is a group of rare systemic autoimmune diseases characterised by necrotising inflammation of small blood vessels and usually associated with circulating ANCA. The pathophysiology of AAV is complex, involving many aspects of the innate and adaptive immune system. Neutrophils are central to the pathogenesis of AAV as they are both the target of the autoantibody and effector cells mediating vascular injury. We describe mechanisms for ANCA induced activation of neutrophils, the pathogenic mechanisms by which this leads to endothelial cell injury, and how neutrophil crosstalk modulates other aspects of the immune system in AAV.
Collapse
Affiliation(s)
- Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom.
| | - Angila Gurung
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Noelle Pisacano
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
28
|
Kang W, Wang C, Wang M, Liu M, Hu W, Liang X, Yang J, Zhang Y. A key regulator of tumor-associated neutrophils: the CXCR2 chemokine receptor. J Mol Histol 2024; 55:1051-1061. [PMID: 39269537 DOI: 10.1007/s10735-024-10260-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: 03/13/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
In recent years, with the advance of research, the role of tumor-associated neutrophils (TANs) in tumors has become a research hotspot. As important effector cells in the innate immune system, neutrophils play a key role in the immune and inflammatory responses of the body. As the first line of defense against bacterial and fungal infections, neutrophils have the ability to kill invading pathogens. In the pathological state of malignant tumors, the phenotype of neutrophils is altered and has an important regulatory function in tumor development. The C-X-C motif chemokine receptor 2(CXCR2) is a key molecule that mediates the migration and aggregation signaling pathway of immune cells, especially neutrophils. This review focuses on the regulation of CXCR2 on TANs in the process of tumorigenesis and development, and emphasizes the application significance of CXCR2 inhibitors in blocking the migration of TANs to tumors.
Collapse
Affiliation(s)
- Wenyan Kang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Chengkun Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Minhui Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Meiqi Liu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Wei Hu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Xiaoqiu Liang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Juanli Yang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China.
| | - Yang Zhang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China.
| |
Collapse
|
29
|
Chakraborty C, Saha S, Bhattacharya M. Recent Advances in Immunological Landscape and Immunotherapeutic Agent of Nipah Virus Infection. Cell Biochem Biophys 2024; 82:3053-3069. [PMID: 39052192 DOI: 10.1007/s12013-024-01424-4] [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: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Over the last two decades, the Nipah virus (NiV) emerged as a highly lethal zoonotic pathogen to humans. Outbreaks occurred occasionally in South and Southeast Asia. Therefore, a safe and effective vaccine against the virus is needed to fight against the deadly virus. Understanding the immunological landscape during this lethal virus infection is necessary in this direction. However, we found scattered information on the immunological landscape of the virus's reservoir, as well as hosts such as humans and livestock. The review provides a recent understanding of the immunological landscape of the virus's reservoir, human hosts, monoclonal antibodies, and vaccines for NiV infection. To describe the immunological landscape, we divided our review article into some points. Firstly, we illustrated bats' immune response as a reservoir during the NiV infection. Secondly, we illustrated an overview of the molecular mechanisms underlying the immune response to the NiV infection, various immune cells, humans' innate immune response, adaptive immunity, and the landscape of cytokines and chemokines. We also discussed INF escape, NET evasion, the T cell landscape, and the B cell landscape during virus infection. Thirdly, we also demonstrated the potential monoclonal antibody therapeutics, and vaccines. Finally, neutralizing antibodies (nAbs) of NiV and potentially other therapeutic strategies were discussed. The review will help researchers for better understanding the immunological landscape, mAbs, and vaccines, enabling them to develop their next-generation versions.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India.
| | - Sagnik Saha
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756020, Odisha, India
| |
Collapse
|
30
|
Harithpriya K, Kaussikaa S, Kavyashree S, Geetha A, Ramkumar KM. Pathological insights into cell death pathways in diabetic wound healing. Pathol Res Pract 2024; 264:155715. [PMID: 39550997 DOI: 10.1016/j.prp.2024.155715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/27/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Diabetic foot ulcers (DFUs) are a microvascular complication that affects almost 21 % of the diabetic population. DFUs are characterized by lower limb abnormalities, chronic inflammation, and a heightened hypoxic environment. The challenge of healing these chronic wounds arises from impaired blood flow, neuropathy, and dysregulated cell death processes. The pathogenesis of DFUs involves intricate mechanisms of programmed cell death (PCD) in different cell types, which include keratinocytes, fibroblasts, and endothelial cells. The modes of cell death comprise apoptosis, autophagy, ferroptosis, pyroptosis, and NETosis, each defined by distinct biochemical hallmarks. These diverse mechanisms contribute to tissue injury by inducing neutrophil extracellular traps and generating cellular stressors like endoplasmic reticulum stress, oxidative stress, and inflammation. Through a comprehensive review of experimental studies identified from literature databases, this review synthesizes current knowledge on the critical signaling cascades implicated in programmed cell death within the context of diabetic foot ulcer pathology.
Collapse
Affiliation(s)
- Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, TN 603210, United States
| | - Srinivasan Kaussikaa
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, TN 603210, United States
| | - Srikanth Kavyashree
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, TN 603210, United States
| | - Avs Geetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, TN 603210, United States
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, TN 603210, United States.
| |
Collapse
|
31
|
Kim DY, Ryu JH, Kim JH, Lee EH, Baek JH, Woo KM. Targeting Age-Related Impaired Bone Healing: ZnO Nanoparticle-Infused Composite Fibers Modulate Excessive NETosis and Prolonged Inflammation in Aging. Int J Mol Sci 2024; 25:12851. [PMID: 39684562 DOI: 10.3390/ijms252312851] [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: 11/07/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Bone defects present significant challenges in clinical contexts, particularly among the elderly, and are often linked to altered innate immune responses; however, underlying mechanisms remain to be understood. This study investigates immune changes in early bone healing in aged mice, emphasizing the effects of zinc in modulating inflammatory processes. By exploring the role of zinc and NETosis in this process, we seek to develop novel therapeutic strategies that could improve bone repair in aging populations. Critical-sized calvarial bone defects were induced in young (8-week-old) and aged (18-month-old) mice, with RNA sequencing analysis. Zinc oxide nanoparticle-infused polycaprolactone (ZnPCL) scaffolds were then fabricated using electrospinning, and their effects on intracellular zinc levels, NETosis, M2 polarization, and bone formation were assessed through in vitro and in vivo experiments. In aged mice, bone healing was delayed, inflammation was prolonged, and NETosis was excessive. RNA sequencing identified alterations in zinc ion transport genes, alongside excessive NETosis. Aged mouse neutrophils exhibited low intracellular zinc levels. ZnPCL fibers effectively reduced NETosis and inflammation, promoted M2 macrophage polarization, and enhanced new bone formation, thereby improving bone healing in aged mice. This study demonstrates that ZnO nanoparticle-infused biomaterials, ZnPCL, effectively deliver zinc to neutrophils, reduce NETosis, promote M2 polarization, and enhance bone healing in aged mice.
Collapse
Affiliation(s)
- Do-Yeun Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Hyun Ryu
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Hyung Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Hye Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Hwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
- Department of Pharmacology & Dental Therapeutics, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Mi Woo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
- Department of Pharmacology & Dental Therapeutics, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
32
|
Muñoz-Caro T, Quiroz P, Abarca C, Gómez-Ceruti M, Alarcón P, Teuber S, Navarro M, Taubert A, Hermosilla C, Burgos RA. Fasciola hepatica Soluble Antigen ( FhAg)-Induced NETs Under Hypoxic Conditions Exert Cytotoxic Effects on Hepatic Cells In Vitro. Animals (Basel) 2024; 14:3456. [PMID: 39682421 DOI: 10.3390/ani14233456] [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: 09/13/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Fasciola hepatica is a parasitic trematode that causes fasciolosis in sheep, provoking a decrease in their reproductive capacity, weight gain, meat and milk production, and wool quality. In the pathogenesis of F. hepatica, the penetration and migration of parasitic stages through the liver provoke intense inflammatory immune responses and tissue damage. The aim of this study was to investigate the cytotoxic effects of Fascila hepatica-induced ovine NETs in exposed hepatocytes in vitro, and to analyze whether F. hepatica antigens (FhAg) trigger the release of ovine NETs under hypoxic conditions as well as the roles of matrix metalloproteinase-9 (MMP-9) and CD11b in this cellular process in vitro. Here, isolated ovine PMNs were co-cultured with FhAg under hypoxia (5% O2) and NETs were visualized via immunofluorescence analyses, confirming their classical characteristics. The quantification of NETs in response to FhAg in hypoxic conditions significantly enhanced the formation of anchored and cell-free NETs (p < 0.01), and NADPH oxidase (NOX) inhibitor diphenylene iodonium (DPI) significantly reduced their production (p < 0.05). Furthermore, the cytotoxic effect of NETs on hepatic cells was determined by using a live/dead-staining with Sytox Orange, thereby demonstrating that FhAg-induced NETs are cytotoxic for hepatic cells (p = 0.001). We additionally analyzed PMN supernatants to determine the enzymatic activity of MMP-9, observing that FhAg exposure enhances MMP-9 release in ovine PMNs (p < 0.05) but not in bovine PMNs. Interestingly, by using flow cytometric analysis, we determined that the exposure of PMNs to FhAg does not increase the CD11b surface expression of ovine PMNs. This could be an effect of the activation of other surface receptors or transcription factors involved in F. hepatica-induced NETosis. Consequently, we hypothesize that F. hepatica-induced NETs play a role in the pathogenesis of fasciolosis, contributing to liver tissue damage if released in an uncontrolled manner.
Collapse
Affiliation(s)
- Tamara Muñoz-Caro
- Escuela de Medicina Veterinaria, Facultad de Medicina Veterinaria y Recursos Naturales, Universidad Santo Tomás, Talca 3460000, Chile
| | - Pamela Quiroz
- Escuela de Medicina Veterinaria, Facultad de Medicina Veterinaria y Recursos Naturales, Universidad Santo Tomás, Talca 3460000, Chile
| | - Cristina Abarca
- Escuela de Medicina Veterinaria, Facultad de Medicina Veterinaria y Recursos Naturales, Universidad Santo Tomás, Talca 3460000, Chile
| | - Marcela Gómez-Ceruti
- Centro de Investigación de Ovinos Para El Secano OVISNOVA, Facultad de Medicina Veterinaria y Recursos Naturales, Universidad Santo Tomás, Talca 3460000, Chile
| | - Pablo Alarcón
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Stefanie Teuber
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Max Navarro
- Institute of Veterinary Clinical Science, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Anja Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Rafael A Burgos
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| |
Collapse
|
33
|
Datta R, Miskolci V, Gallego-López GM, Britt E, Gillette A, Kralovec A, Giese MA, Qian T, Votava J, Fan J, Huttenlocher A, Skala M. Single cell autofluorescence imaging reveals immediate metabolic shifts of neutrophils with activation across biological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605362. [PMID: 39211087 PMCID: PMC11360992 DOI: 10.1101/2024.07.26.605362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neutrophils, the most abundant leukocytes in human peripheral circulation, are crucial for the innate immune response. They are typically quiescent but rapidly activate in response to infection and inflammation, performing diverse functions such as oxidative burst, phagocytosis, and NETosis, which require significant metabolic adaptation. Deeper insights into such metabolic changes will help identify regulation of neutrophil functions in health and diseases. Due to their short lifespan and associated technical challenges, the metabolic processes of neutrophils are not completely understood. This study uses optical metabolic imaging (OMI), which entails optical redox ratio and fluorescence lifetime imaging microscopy of intrinsic metabolic coenzymes NAD(P)H and FAD to assess the metabolic state of single neutrophils. Primary human neutrophils were imaged in vitro under a variety of activation conditions and metabolic pathway inhibitors, while metabolic and functional changes were confirmed with mass spectrometry, oxidative burst, and NETosis measurements. Our findings show that neutrophils undergo rapid metabolic remodeling to a reduced redox state indicated by changes in NAD(P)H lifetime and optical redox ratio, with a shift to an oxidized redox state during activation. Additionally, single cell OMI analysis reveals a heterogeneous metabolic response across neutrophils and human donors to live pathogen infection ( Pseudomonas aeruginosa and Toxoplasma gondii ). Finally, consistent OMI changes with activation were confirmed between in vitro human and in vivo zebrafish larvae neutrophils. This study demonstrates the potential of OMI as a versatile tool for studying neutrophil metabolism and underscores its use across different biological systems, offering insights into neutrophil metabolic activity and function at a single cell level.
Collapse
|
34
|
Cui X, Li T, Yang J, Li X, Xuan P, Wang H. Predictive Value of dsDNA and Nucleosomes as Neutrophil Extracellular Traps-Related Biomarkers for COVID-19 in Older Patients. J Inflamm Res 2024; 17:8831-8838. [PMID: 39564546 PMCID: PMC11575441 DOI: 10.2147/jir.s414688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Background Previous studies have demonstrated that neutrophil extracellular traps (NETs) are crucial in infectious diseases. This study aims to evaluate the clinical value of NET-related biomarkers in identifying the risk of COVID-19 and diagnosing the disease. Methods This study involved 32 patients who tested positive for COVID-19 via polymerase chain reaction (PCR) between April and August 2023. During the same period, 30 healthy volunteers were enrolled as a control group. The principal biomarkers related to NETs are citrullinated histone H3 (CitH3), double-stranded DNA (dsDNA), myeloperoxidase-DNA complex (MPO-DNA), and Nucleosome. Elevated levels in two or more of these biomarkers indicate raised NET concentrations. Multivariable logistic regression analysis was employed to assess whether NET-related biomarkers were the independent risk factor of COVID-19. The diagnostic value of NET-related biomarkers in COVID-19 was further evaluated using receiver operating characteristic (ROC) curve analysis. Statistical procedures were executed in SPSS software (version 24.0, USA). Results Compared with the control group, patients infected with COVID-19 had higher levels of dsDNA and nucleosomes (P < 0.001). Correlation analysis revealed a positive correlation between dsDNA levels and neutrophil count (r = 0.309, P = 0.015) as well as between nucleosome levels and neutrophil count (r = 0.446, P < 0.001). Further analysis showed that dsDNA and nucleosomes were independent risk factors for COVID-19 infection. ROC curve analysis showed that dsDNA area under the curve (AUC) = 0.777, 95% confidence interval (CI), 0.661-0.893, P < 0.001, and nucleosomes (AUC = 0.884, 95% CI, 0.778-0.991, P < 0.001) had well diagnostic value in the diagnosing COVID-19 infection. Conclusion NET-related biomarkers, dsDNA and nucleosomes, were independent risk factors of COVID-19 infection and potentially useful biomarkers in diagnosing COVID-19 infection in older patients.
Collapse
Affiliation(s)
- Xudong Cui
- Respiratory and Critical Care Medicine Department, Inner Mongolia Baogang Hospital, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Tiewei Li
- Department of Clinical Laboratory, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Key Laboratory of Children's Infection and Immunity, Zhengzhou, People's Republic of China
| | - Jingping Yang
- Respiratory and Critical Care Medicine Department, Inner Mongolia Baogang Hospital, The Third Affiliated Hospital of Inner Mongolia Medical University, Baotou, People's Republic of China
| | - Xiaojuan Li
- Department of Clinical Laboratory, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Key Laboratory of Children's Infection and Immunity, Zhengzhou, People's Republic of China
| | - Pengfei Xuan
- Respiratory and Critical Care Medicine Department, Inner Mongolia Baogang Hospital, The Third Affiliated Hospital of Inner Mongolia Medical University, Baotou, People's Republic of China
| | - Hongyan Wang
- Respiratory and Critical Care Medicine Department, Inner Mongolia Baogang Hospital, The Third Affiliated Hospital of Inner Mongolia Medical University, Baotou, People's Republic of China
| |
Collapse
|
35
|
Di Vito L, Di Giusto F, Mazzotta S, Scalone G, Bruscoli F, Silenzi S, Selimi A, Angelini M, Galieni P, Grossi P. Management of vulnerable patient phenotypes and acute coronary syndrome mechanisms. Int J Cardiol 2024; 415:132365. [PMID: 39029561 DOI: 10.1016/j.ijcard.2024.132365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Atherosclerosis is a chronic vascular disease. Its prevalence increases with aging. However, atherosclerosis may also affect young subjects without significant exposure to the classical risk factors. Recent evidence indicates clonal hematopoiesis of indeterminate potential (CHIP) as a novel cardiovascular risk factor that should be suspected in young patients. CHIP represents a link between impaired bone marrow and atherosclerosis. Atherosclerosis may present with an acute symptomatic manifestation or subclinical events that favor plaque growth. The outcome of a plaque relies on a balance of innate and environmental factors. These factors can influence the processes that initiate and propagate acute plaque destabilization leading to intraluminal thrombus formation or subclinical vessel healing. Thirty years ago, the first autopsy study revealed that coronary plaques can undergo rupture even in subjects without a known cardiovascular history. Nowadays, cardiac magnetic resonance studies demonstrate that this phenomenon is not rare. Myocardial infarction is mainly due to plaque rupture and plaque erosion that have different pathophysiological mechanisms. Plaque erosion carries a better prognosis as compared to plaque rupture. Thus, a tailored conservative treatment has been proposed and some studies demonstrated it to be safe. On the contrary, plaque rupture is typically associated with inflammation and anti-inflammatory treatments have been proposed in response to persistently elevate biomarkers of systemic inflammation. In conclusion, atherosclerosis may present in different forms or phenotypes. Vulnerable patient phenotypes, identified by using intravascular imaging techniques, biomarkers, or even genetic analyses, are characterized by distinctive pathophysiological mechanisms. These different phenotypes merit tailored management.
Collapse
Affiliation(s)
- Luca Di Vito
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy.
| | | | - Serena Mazzotta
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Giancarla Scalone
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Filippo Bruscoli
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Simona Silenzi
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Adelina Selimi
- University Hospital "Umberto I-Lancisi-Salesi", Ancona, Italy
| | - Mario Angelini
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Piero Galieni
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | | |
Collapse
|
36
|
Yu X, Chen Z, Bao W, Jiang Y, Ruan F, Wu D, Le K. The neutrophil extracellular traps in neurological diseases: an update. Clin Exp Immunol 2024; 218:264-274. [PMID: 38975702 PMCID: PMC11557138 DOI: 10.1093/cei/uxae057] [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: 01/16/2024] [Revised: 05/08/2024] [Accepted: 07/06/2024] [Indexed: 07/09/2024] Open
Abstract
Neutrophil extracellular traps (NETs) released by neutrophils are web-like DNA structures adhered to granulin proteins with bactericidal activity and can be an important mechanism for preventing pathogen dissemination or eliminating microorganisms. However, they also play important roles in diseases of other systems, such as the central nervous system. We tracked the latest advances and performed a review based on published original and review articles related to NETs and neurological diseases. Generally, neutrophils barely penetrate the blood-brain barrier into the brain parenchyma, but when pathological changes such as infection, trauma, or neurodegeneration occur, neutrophils rapidly infiltrate the central nervous system to exert their defensive effects. However, neutrophils may adversely affect the host when they uncontrollably release NETs upon persistent neuroinflammation. This review focused on recent advances in understanding the mechanisms and effects of NETs release in neurological diseases, and we also discuss the role of molecules that regulate NETs release in anticipation of clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong S.A.R., China
| |
Collapse
|
37
|
Gao P, Zhou J, Sun L, Liu D. Neutrophil Extracellular Traps in Oral Diseases. Oral Dis 2024. [PMID: 39530338 DOI: 10.1111/odi.15197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE To summarize the current knowledge of the neutrophil extracellular traps (NETs) and its critical role in various oral diseases. METHODS We reviewed the recent research on NETs through PubMed and Web of Science. An analysis of recent research results was summarized from three aspects: NETs induction and formation, functions of NETs, and NETs in oral diseases. RESULTS The relationship between neutrophils and NETs is critical to the body's defense against microbial invasion. NETs can effectively combat pathogens with an anti-inflammatory effect and meanwhile it can contribute to inflammation. Moreover, it can synergize with other immune cells to respond to stimuli, such as pathogens, host-derived mediators, and drugs. It was revealed that NETs play different roles to influence various oral diseases like periodontitis, endodontic infection, oral mucosal diseases, maxillofacial tumors, and many other oral diseases. CONCLUSION The balance between the protective and potentially harmful effects of NETs is a key factor in determining the outcome of infections and inflammatory responses. The role of NETs in oral diseases needs to be further studied to enable better understanding of its role in the different oral diseases.
Collapse
Affiliation(s)
- Pengfei Gao
- Department of Periodontology, Suzhou Stomatological Hospital, Suzhou, Jiangsu, China
| | - Jun Zhou
- Department of Conservative Dentistry, Division of Biomaterials and Engineering, Showa University School of Dentistry, Tokyo, Japan
| | - Lu Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Dayong Liu
- Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
38
|
Lin C, Herlihy SE, Li M, Deng H, Kim R, Bernabei L, Rosenwasser M, Gabrilovich DI, Vogl DT, Nefedova Y. Neutrophil extracellular traps promote tumor chemoresistance to anthracyclines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622533. [PMID: 39605505 PMCID: PMC11601256 DOI: 10.1101/2024.11.07.622533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The microenvironment plays an important role in promoting tumor cell chemoresistance, but the mechanisms responsible for this effect are not clear. Here, using models of multiple myeloma (MM) and solid cancers, we demonstrate a novel mechanism mediated by neutrophils, a major cell population in the bone marrow (BM), that protects cancer cells from chemotherapeutics. We show that in response to tumor-derived soluble factors, BM neutrophils release their DNA in the form of neutrophil extracellular traps (NETs). Cell-free DNA derived from NETs is then taken up by tumor cells via endocytosis and localizes to the cytoplasm. We found that both NETs and cell-free DNA taken up by tumor cells can bind anthracyclines, leading to tumor cell resistance to this class of chemotherapeutic agents. Targeting cell-free DNA with Pulmozyme or blocking NET formation with a PAD4 inhibitor abrogates the chemoprotective effect of neutrophils and restores sensitivity of tumor cells to anthracyclines.
Collapse
|
39
|
Xia M, Han Y, Sun L, Li D, Zhu C, Li D. The role of neutrophils in osteosarcoma: insights from laboratory to clinic. Front Immunol 2024; 15:1490712. [PMID: 39582869 PMCID: PMC11582048 DOI: 10.3389/fimmu.2024.1490712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
Osteosarcoma, a highly aggressive malignant bone tumor, is significantly influenced by the intricate interactions within its tumor microenvironment (TME), particularly involving neutrophils. This review delineates the multifaceted roles of neutrophils, including tumor-associated neutrophils (TANs) and neutrophil extracellular traps (NETs), in osteosarcoma's pathogenesis. TANs exhibit both pro- and anti-tumor phenotypes, modulating tumor growth and immune evasion, while NETs facilitate tumor cell adhesion, migration, and immunosuppression. Clinically, neutrophil-related markers such as the neutrophil-to-lymphocyte ratio (NLR) predict patient outcomes, highlighting the potential for neutrophil-targeted therapies. Unraveling these complex interactions is crucial for developing novel treatment strategies that harness the TME to improve osteosarcoma management.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongsong Li
- Department of Orthopedics, The First Hospital of Jilin University,
Changchun, Jilin, China
| |
Collapse
|
40
|
Inoue M, Takayama K, Hashimoto R, Enomoto M, Date N, Ohsumi A, Mizowaki T. Hyponatremia unleashes neutrophil extracellular traps elevating life-threatening pulmonary embolism risk. Proc Natl Acad Sci U S A 2024; 121:e2404947121. [PMID: 39475645 PMCID: PMC11551416 DOI: 10.1073/pnas.2404947121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/27/2024] [Indexed: 11/13/2024] Open
Abstract
Neutrophil extracellular traps (NETs), essential for controlling infections, can induce various pathologies when dysregulated. Known triggers for infection-independent NETs release exist, yet a comprehensive understanding of the conditions prompting such responses is lacking. In this study, we identify hyponatremia as an independent inducer of NETs release, a common clinical condition that disrupts sodium/calcium exchange within neutrophils. This disruption leads to an excess of intracellular calcium, subsequent elevation of reactive oxygen species (ROS), and the citrullination of histone H3, culminating in the activation of NETs-release pathways. Notably, under hyponatremic conditions, this mechanism is exacerbated during infectious states, leading to the deposition of NETs in the lungs and increasing the risk of life-threatening pulmonary embolism. Our findings underscore the critical role of sodium and calcium homeostasis in neutrophil functionality and provide insights into the pathogenesis of hyponatremia-associated diseases, highlighting potential therapeutic interventions targeting NETs dynamics.
Collapse
Affiliation(s)
- Minoru Inoue
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Radiation and Proton Therapy Center, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Rina Hashimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Masahiro Enomoto
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Naoki Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| |
Collapse
|
41
|
Bun M, Kawano M, Yamamoto G, Sakata M, Shimura K, Toda A, Nakamura K, Kinose Y, Kodama M, Hashimoto K, Kobayashi E, Sawada K, Kimura T. G-CSF induces neutrophil extracellular traps formation and promotes ovarian cancer peritoneal dissemination. J Leukoc Biol 2024; 116:1157-1168. [PMID: 39082070 DOI: 10.1093/jleuko/qiae166] [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: 01/12/2024] [Revised: 06/16/2024] [Accepted: 07/30/2024] [Indexed: 11/05/2024] Open
Abstract
Epithelial ovarian cancer is characterized by aggressive peritoneal dissemination. Neutrophils are mobilized to peritoneal cavity in some patients with ovarian cancer dissemination; however, its pathological significance remains unknown. This study aimed to investigate the role of neutrophil extracellular traps (NETs) in ovarian cancer dissemination. We conducted a retrospective analysis of clinical data and samples from 340 patients with ovarian cancer who underwent primary surgery between 2007 and 2016 at the Osaka University Hospital. In vitro, NETs formation was induced by stimulating human peripheral neutrophils. The human ovarian cancer cell line, OVCAR8, was cocultured with NETs. For an ovarian cancer dissemination mouse model, we performed an intraperitoneal injection of OVCAR8 cells into nude mice. The association between NETs and peritoneal dissemination was explored, and model mice were treated with the PAD4 inhibitor GSK484 to assess antitumor efficacy. Neutrophilia (neutrophil count >7000/mm3) correlated with shorter survival, advanced peritoneal dissemination, elevated granulocyte colony-stimulating factor (G-CSF) levels, increased neutrophil count in ascites, and augmented NETs foci in peritoneal dissemination sites. In vitro assays revealed that G-CSF stimulated neutrophils to form NETs, promoting cancer cell adhesion. In vivo investigations revealed that G-CSF-producing tumor-bearing mice had accelerated peritoneal dissemination and poor prognosis. NETs formation was pathologically observed at the peritoneal dissemination sites. Inhibition of NETs formation by GSK484 significantly delayed peritoneal dissemination in vivo. In conclusion, G-CSF was associated with intra-abdominal NETs formation and increased peritoneal dissemination. NETs represent potential therapeutic targets for ovarian cancer, particularly in patients with neutrophilia.
Collapse
Affiliation(s)
- Michiko Bun
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mahiru Kawano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Gaku Yamamoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mina Sakata
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kotaro Shimura
- Department of Obstetrics and Gynecology, Osaka Rosai Hospital, 1179-3, Nakasone, Kita-ku, Sakai, Osaka 591-8025, Japan
| | - Aska Toda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koji Nakamura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuto Kinose
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Michiko Kodama
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kae Hashimoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eiji Kobayashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Kenjiro Sawada
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
42
|
Bhattacharya M, Spencer BL, Kwiecinski JM, Podkowik M, Putzel G, Pironti A, Shopsin B, Doran KS, Horswill AR. Collagen binding adhesin restricts Staphylococcus aureus skin infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621145. [PMID: 39554114 PMCID: PMC11565922 DOI: 10.1101/2024.11.01.621145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Staphylococcus aureus causes approximately 80% of skin and soft tissue infections (SSTIs). Collagen is the most abundant human extracellular matrix protein with critical roles in wound healing, and S. aureus encodes a collagen binding adhesin (Cna). The role of this protein during skin infections is unknown. Here we report that inability to bind collagen results in worsened pathology of intradermal Δcna S. aureus infection. WT/Cna+ S. aureus showed reduced infection severity, aggregate formation, and significantly improved clearance of bacteria. Cna binds to the collagen-like domain of serum C1q protein to reduce its opsonophagocytic functions. We demonstrate that infection of C1qKO mice with WT bacteria show results similar to the Δcna group. Conversely, inability to bind collagen resulted in an amplified inflammatory response caused in part by macrophage and neutrophil small molecule mediators released at the infection site (MMP-9, MMP-12, LTB4), resulting in increased immune cell infiltration and death.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
| | - Jakub M. Kwiecinski
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Gregory Putzel
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora CO, USA
- Department of Veterans Affairs, Eastern Colorado Healthcare System, Denver, CO, USA
- Lead author
| |
Collapse
|
43
|
Seo Y, Kim SI, Song SH, Kim JG, Gu JY, Jeon HW, Lee M, Kim HK. Elevation of circulating neutrophil extracellular traps in endometrial cancer: Poor prognostic value of cell-free double-stranded DNA. Transl Oncol 2024; 49:102072. [PMID: 39128260 PMCID: PMC11366898 DOI: 10.1016/j.tranon.2024.102072] [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: 02/08/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024] Open
Abstract
OBJECTIVE Neutrophils produce neutrophil extracellular traps (NETs) by releasing nuclear contents into the extracellular environment. NETs are associated with systemic inflammation and cancer development and progression. We aimed to investigate whether NET markers are associated with the prognosis of endometrial cancer. METHODS Circulating levels of three NET markers (histone-DNA complex, cell-free double-stranded DNA (dsDNA), and neutrophil elastase) were measured in 98 patients with endometrial cancer who underwent surgery as primary treatment between January 2015 and June 2018 and 45 healthy women. Area under the receiver operating characteristic curve (AUC) analyses were conducted to investigate the diagnostic and prognostic utility of the markers for endometrial cancer. RESULTS Patients with endometrial cancer showed significantly higher levels of the three NET markers than those in healthy controls. In discriminating endometrial cancer patients from healthy controls, the three NET markers showed AUC values in the following order: cell-free dsDNA (0.832; 95 % CI, 0.760-0.889), histone-DNA complex (0.740; 95 % CI, 0.660-0.809), and neutrophil elastase (0.689; 95 % CI, 0.607-0.764), comparable to those of CA-125 (0.741; 95 % CI, 0.659-0.813). Multivariate analysis adjusting for FIGO stage, histology, and lymphovascular space invasion, and lymph node involvement revealed that cell-free dsDNA level (cutoff: 95.2 ng/mL) was an independent prognostic marker for poor progression-free (adjusted HR, 2.75; 95 % CI, 1.096.92; P = 0.032) and overall survival (adjusted HR, 11.51; 95 % CI, 2.0664.22; P = 0.005) for patients with endometrial cancer. CONCLUSION High levels of circulating NET markers were observed in patients with endometrial cancer. Cell-free dsDNA levels may play a role as prognostic markers for endometrial cancer.
Collapse
Affiliation(s)
- Yeonju Seo
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jisoo G Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Ja-Yoon Gu
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hye Won Jeon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea; Department of Obstetrics and Gynecology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea; Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea.
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
44
|
Hallak J, Caldini EG, Teixeira TA, Correa MCM, Duarte-Neto AN, Zambrano F, Taubert A, Hermosilla C, Drevet JR, Dolhnikoff M, Sanchez R, Saldiva PHN. Transmission electron microscopy reveals the presence of SARS-CoV-2 in human spermatozoa associated with an ETosis-like response. Andrology 2024; 12:1799-1807. [PMID: 38469742 DOI: 10.1111/andr.13612] [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: 09/04/2023] [Revised: 01/05/2024] [Accepted: 01/23/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Severe acute syndrome coronavirus 2 can invade a variety of tissues, including the testis. Even though this virus is scarcely found in human semen polymerase chain reaction tests, autopsy studies confirm the viral presence in all testicular cell types, including spermatozoa and spermatids. OBJECTIVE To investigate whether the severe acute syndrome coronavirus 2 is present inside the spermatozoa of negative polymerase chain reaction-infected men up to 3 months after hospital discharge. MATERIALS AND METHODS This cross-sectional study included 13 confirmed moderate-to-severe COVID-19 patients enrolled 30-90 days after the diagnosis. Semen samples were obtained and examined with real-time polymerase chain reaction for RNA detection and by transmission electron microscopy. RESULTS In moderate-to-severe clinical scenarios, we identified the severe acute syndrome coronavirus 2 inside spermatozoa in nine of 13 patients up to 90 days after discharge from the hospital. Moreover, some DNA-based extracellular traps were reported in all studied specimens. DISCUSSION AND CONCLUSION Although severe acute syndrome coronavirus 2 was not present in the infected men's semen, it was intracellularly present in the spermatozoa till 3 months after hospital discharge. The Electron microscopy (EM) findings also suggest that spermatozoa produce nuclear DNA-based extracellular traps, probably in a cell-free DNA-dependent manner, similar to those previously described in the systemic inflammatory response to COVID-19. In moderate-to-severe cases, the blood-testes barrier grants little defence against different pathogenic viruses, including the severe acute syndrome coronavirus 2. The virus could also use the epididymis as a post-testicular route to bind and fuse to the mature spermatozoon and possibly accomplish the reverse transcription of the single-stranded viral RNA into proviral DNA. These mechanisms can elicit extracellular cell-free DNA formation. The potential implications of our findings for assisted conception must be addressed, and the evolutionary history of DNA-based extracellular traps as preserved ammunition in animals' innate defence might improve our understanding of the severe acute syndrome coronavirus 2 pathophysiology in the testis and spermatozoa.
Collapse
Affiliation(s)
- Jorge Hallak
- Departamento de Cirurgia, Disciplina de Urologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Androscience, Science & Innovation Center in Andrology and High-Complex Clinical and Research Andrology Laboratory., Androscience Institute, Sao Paulo, Brasil
| | - Elia G Caldini
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thiago A Teixeira
- Androscience, Science & Innovation Center in Andrology and High-Complex Clinical and Research Andrology Laboratory., Androscience Institute, Sao Paulo, Brasil
- Departamento de Cirurgia, Divisão de Urologia, Hospital Universitário da Universidade Federal do Amapá, Amapá, Brazil
| | | | - Amaro N Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fabiola Zambrano
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
- Center of Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Joël R Drevet
- GReD Institute, CNRS-INSERM-Université Clermont Auvergne, Faculty of Medicine, Clermont-Ferrand, France
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raul Sanchez
- Center of Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Paulo H N Saldiva
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Midtbø HMD, Borchel A, Morton HC, Paley R, Monaghan S, Haugland GT, Øvergård AC. Cell death induced by Lepeophtheirus salmonis labial gland protein 3 in salmonid fish leukocytes: A mechanism for disabling host immune responses. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109992. [PMID: 39481500 DOI: 10.1016/j.fsi.2024.109992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The salmon louse (Lepeophtheirus salmonis) is an ectoparasite feeding on mucus, skin, and blood of salmonids. On parasitised fish erosions and, at later lice stages, ulcerations appear at the louse feeding site. In susceptible species like Atlantic salmon (Salmo salar) with a limited rejection of lice, only a mild inflammatory response with minor influx of immune cells is seen at these lesions, as the salmon louse secrete proteins that can dampen immune responses. In a previous study, Lepeophtheirus salmonis labial gland protein 3 (LsLGP3) was suggested to dampen cellular responses, and the present study aimed at increasing our understanding of its mode of action. LsLGP3 was found to be secreted on to the host skin, and both in vivo and in vitro experiments were performed to elucidate its function. Histological analysis of the louse attachment site revealed an epidermal and dermal influx of mainly macrophages and granulocytes after 5 days post infestation. The immune cell influx was deeper in the dermis throughout the louse infestation, and LsLGP3 may be involved in dampening this response. Enriched populations of Atlantic salmon B-cells, T-cells, granulocytes, and monocytes were exposed to recombinant LsLGP3 (recLGP3) in vitro, resulting in a significant decrease in cell viability compared to non-exposed controls. An apoptotic cell morphology with "beads-on-a-string" like protrusions was seen in all leukocyte cell fractions after recLGP3 exposure, but not in erythrocytes or keratocytes. A decreased viability was also detected in pink salmon leucocytes, which was not in leucocytes from non-salmonid species. These functional insights suggest that LsLGP3 specifically induces apoptosis of salmonid leukocytes and is likely a key protein secreted by the lice that disables the Atlantic salmon ability to mount an adequate immune response towards the salmon louse. In vivo LsLGP3 knock down studies indicated that the effect is localised primarily at the lice feeding site, without affecting immune cells that are not situated adjacent to the lice-inflicted lesion. The findings from this study could significantly aid in the development of new immune based anti-salmon louse prophylactic measures and treatments.
Collapse
Affiliation(s)
| | - Andreas Borchel
- Department of Biological Sciences, University of Bergen, P.O. Box 7803, NO-5020, Bergen, Norway
| | - H Craig Morton
- Institute of Marine Research, P.O. Box 1870 Nordnes, NO-5817, Bergen, Norway
| | - Richard Paley
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), The Nothe, Barrack Road, Weymouth, DT4 8UB, United Kingdom
| | - Sean Monaghan
- Institute of Aquaculture, University of Stirling, Stirling, United Kingdom
| | - Gyri Teien Haugland
- Department of Biological Sciences, University of Bergen, P.O. Box 7803, NO-5020, Bergen, Norway
| | - Aina-Cathrine Øvergård
- Department of Biological Sciences, University of Bergen, P.O. Box 7803, NO-5020, Bergen, Norway
| |
Collapse
|
46
|
Wang K, Wang X, Song L. Unraveling the complex role of neutrophils in lymphoma: From pathogenesis to therapeutic approaches (Review). Mol Clin Oncol 2024; 21:85. [PMID: 39347476 PMCID: PMC11428085 DOI: 10.3892/mco.2024.2783] [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: 05/22/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Lymphoma, a malignancy of the lymphatic system, which is critical for maintaining the body's immune defenses, has become a focal point in recent research due to its intricate interplay with neutrophils-white blood cells essential for combating infections and inflammation. Unlike prior perceptions associating neutrophils only with tumor support, contemporary studies underscore their intricate and multifaceted involvement in the immune response to lymphoma. Recognizing the nuanced participation of neutrophils in lymphoma is crucial for developing innovative treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Engineering, School of Life Sciences and Biotechnology, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiao Wang
- Reproduction Medicine Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong 524002, P.R. China
| | - Li Song
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
47
|
Xie R, Sher KHJ, Tang SYC, Yam IYL, Lee CH, Wu Q, Yap DYH. Dysregulation of neutrophil extracellular traps (NETs)-related genes in the pathogenesis of diabetic kidney disease - Results from bioinformatics analysis and translational studies. Clin Immunol 2024; 268:110379. [PMID: 39396625 DOI: 10.1016/j.clim.2024.110379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/24/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The role of Neutrophil extracellular traps (NETs) in the immunopathogenesis of Diabetic Kidney Disease (DKD) remains elusive. We used a machine learning approach to identify differentially expressed genes (DEGs) associated with NETs in human DKD kidney biopsy datasets and validated the results using single-nucleus RNA sequencing datasets. The expressions of these candidate genes and related cytokines were verified in blood obtained from DKD patients. Three NETs-associated genes (ITGAM, ITGB2 and TLR7) were identified, which all showed significant upregulation in both glomerular and tubulointerstitial compartments in human DKD kidneys. DKD patients showed significantly higher number of activated neutrophils with increased ITGAM and ITGB2 expression, higher serum IL-6 but lower IL-10, compared to healthy controls (p all <0.01). This study suggests that dysregulation of NETs-associated genes ITGAM and ITGB2 are related to the pathogenesis of DKD, and may serve as novel diagnostic markers and therapeutic targets in DKD.
Collapse
Affiliation(s)
- Ruiyan Xie
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Hong Kong 999077, HKSAR, China
| | - Ka Ho Jason Sher
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Hong Kong 999077, HKSAR, China
| | - Sin Yu Cindy Tang
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Hong Kong 999077, HKSAR, China
| | - Irene Ya Lin Yam
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Hong Kong 999077, HKSAR, China
| | - C H Lee
- Division of Endocrinology & Metabolism, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen 518028, HKSAR, China
| | - Qiongli Wu
- Shenzhen Experimental Education School, Shenzhen, China
| | - Desmond Yat Hin Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, The University of Hong Kong, Hong Kong 999077, HKSAR, China.
| |
Collapse
|
48
|
Li S, Xu G, Guo Z, Liu Y, Ouyang Z, Li Y, Huang Y, Sun Q, Giri BR, Fu Q. Deficiency of hasB accelerated the clearance of Streptococcus equi subsp. Zooepidemicus through gasdermin d-dependent neutrophil extracellular traps. Int Immunopharmacol 2024; 140:112829. [PMID: 39083933 DOI: 10.1016/j.intimp.2024.112829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Streptococcus equi subsp. zooepidemicus (S. zooepidemicus, SEZ) is an essential zoonotic bacterial pathogen that can cause various inflammation, such as meningitis, endocarditis, and pneumonia. UDP-glucose dehydrogenase (hasB) is indispensable in synthesizing SEZ virulence factor hyaluronan capsules. Our study investigated the infection of hasB on mice response to SEZ by employing a constructed capsule-deficient mutant strain designated as the ΔhasB strain. This deficiency was associated with a reduced SEZ bacterial load in the mice's blood and peritoneal lavage fluid (PLF) post-infection. Besides, the ΔhasB SEZ strain exhibited a higher propensity for neutrophil infiltration and release of cell-free DNA (cfDNA) in vivo compared to the wild-type (WT) SEZ strain. In vitro experiments further revealed that ΔhasB SEZ more effectively induced the formation of neutrophil extracellular traps (NETs) containing histone 3 (H3), neutrophil elastase (NE), and DNA, than its WT counterpart. Moreover, the release of NETs was determined to be gasdermin D (GSDMD)-dependent during the infection process. Taken together, these findings underscore that the deficiency of the hasB gene in SEZ leads to enhanced GSDMD-dependent NET release from neutrophils, thereby reducing SEZ's capacity to resist NETs-mediated eradication during infection. Our finding paves the way for the development of innovative therapeutic strategies against SEZ.
Collapse
Affiliation(s)
- Shun Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Guobin Xu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhiliang Ouyang
- Houjie Town Agricultural Technology Service Center, Dongguan, Guangdong, China
| | - Yajuan Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Qinqin Sun
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Bikash R Giri
- Department of Zoology, K.K.S. Women's College, Balasore, Odisha, India
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China.
| |
Collapse
|
49
|
Shvedov M, Sherstyukova E, Kandrashina S, Inozemtsev V, Sergunova V. Atomic Force Microscopy and Scanning Ion-Conductance Microscopy for Investigation of Biomechanical Characteristics of Neutrophils. Cells 2024; 13:1757. [PMID: 39513864 PMCID: PMC11545488 DOI: 10.3390/cells13211757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Scanning probe microscopy (SPM) is a versatile tool for studying a wide range of materials. It is well suited for investigating living matter, for example, in single-cell neutrophil studies. SPM has been extensively utilized to analyze cell physical properties, providing detailed insights into their structural and functional characteristics at the nanoscale. Its long-standing application in this field highlights its essential role in cell biology and immunology research, significantly contributing to understanding cellular mechanics and interactions. In this review, we discuss the application of SPM techniques, specifically atomic force microscopy (AFM) and scanning ion-conductance microscopy (SICM), to study the fundamental functions of neutrophils. In addition, recent advances in the application of SPM in single-cell immunology are discussed. The application of these techniques allows for obtaining data on the morphology, topography, and mechanical and electrochemical properties of neutrophils with high accuracy.
Collapse
Affiliation(s)
- Mikhail Shvedov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Research Institute of General Reanimatology, 107031 Moscow, Russia; (E.S.); (S.K.); (V.I.)
| | - Ekaterina Sherstyukova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Research Institute of General Reanimatology, 107031 Moscow, Russia; (E.S.); (S.K.); (V.I.)
| | - Snezhanna Kandrashina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Research Institute of General Reanimatology, 107031 Moscow, Russia; (E.S.); (S.K.); (V.I.)
| | - Vladimir Inozemtsev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Research Institute of General Reanimatology, 107031 Moscow, Russia; (E.S.); (S.K.); (V.I.)
- Koltzov Institute of Development Biology of Russia Academy of Science, 119334 Moscow, Russia
| | - Viktoria Sergunova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, V.A. Negovsky Research Institute of General Reanimatology, 107031 Moscow, Russia; (E.S.); (S.K.); (V.I.)
| |
Collapse
|
50
|
Azzouz D, Palaniyar N. How Do ROS Induce NETosis? Oxidative DNA Damage, DNA Repair, and Chromatin Decondensation. Biomolecules 2024; 14:1307. [PMID: 39456240 PMCID: PMC11505619 DOI: 10.3390/biom14101307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are intricate, DNA-based, web-like structures adorned with cytotoxic proteins. They play a crucial role in antimicrobial defense but are also implicated in autoimmune diseases and tissue injury. The process of NET formation, known as NETosis, is a regulated cell death mechanism that involves the release of these structures and is unique to neutrophils. NETosis is heavily dependent on the production of reactive oxygen species (ROS), which can be generated either through NADPH oxidase (NOX) or mitochondrial pathways, leading to NOX-dependent or NOX-independent NETosis, respectively. Recent research has revealed an intricate interplay between ROS production, DNA repair, and NET formation in different contexts. UV radiation can trigger a combined process of NETosis and apoptosis, known as apoNETosis, driven by mitochondrial ROS and DNA repair. Similarly, in calcium ionophore-induced NETosis, both ROS and DNA repair are key components, but only play a partial role. In the case of bacterial infections, the early stages of DNA repair are pivotal. Interestingly, in serum-free conditions, spontaneous NETosis occurs through NOX-derived ROS, with early-stage DNA repair inhibition halting the process, while late-stage inhibition increases it. The intricate balance between DNA repair processes and ROS production appears to be a critical factor in regulating NET formation, with different pathways being activated depending on the nature of the stimulus. These findings not only deepen our understanding of the mechanisms behind NETosis but also suggest potential therapeutic targets for conditions where NETs contribute to disease pathology.
Collapse
Affiliation(s)
- Dhia Azzouz
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|