1
|
Park EJ, Lee YS, Jun EM, Lee BW, Park SM, Lee HJ. Immune-Enhancing Effects of Two Potential Probiotic Strains Latilactobacillus curvatus WiKim0169 and Pediococcus acidilactici WiKim0170 in a Cyclophosphamide-Induced Immunosuppression Rat Model. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10585-4. [PMID: 40423876 DOI: 10.1007/s12602-025-10585-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2025] [Indexed: 05/28/2025]
Abstract
Emerging evidence suggests that probiotics are beneficial for immunity and play a crucial role in regulating gut microbiota. However, the immune-enhancing effects of specific bacterial species remain uncertain. This study investigated the effects of two potential probiotic strains, Latilactobacillus curvatus WiKim0169 (Wilac L004, W4) and Pediococcus acidilactici WiKim0170 (Wilac L002, W2) isolated from fermented cabbage, on immune enhancement and gut microbiota changes in a cyclophosphamide-induced immunosuppression rat model. The results revealed that W4 and W2 improved natural killer cell activity, serum nitrite, and immunoglobulin levels. They also increased cytokine levels and activated the nuclear factor-κB pathway, substantiating the underlying mechanism of the immune-enhancing effects. Additionally, both strains altered gut microbiota composition by increasing bacteria that are being studied for their potential association with immune function. Taken together, both strains improved immune-related biomarkers and the gut microbiota. These findings suggest W4 and W2 as promising probiotics with immune-enhancing properties.
Collapse
Affiliation(s)
- Eun-Jung Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - You-Suk Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - Eun-Min Jun
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - Byung Wook Lee
- Microbiome R&D Center, Pharmsville Co., Ltd, Seoul, 07793, Republic of Korea
| | - Sang Min Park
- Microbiome R&D Center, Pharmsville Co., Ltd, Seoul, 07793, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea.
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.
- Gachon Biomedical Convergence Institute, Gachon University Gil Medical Center, Incheon, 21565, Republic of Korea.
| |
Collapse
|
2
|
Liu Y, Sheng S, Wu L, Wang H, Xue H, Wang R. Flavonoid-rich extract of Paederia scandens (Lour.) Merrill improves hyperuricemia by regulating uric acid metabolism and gut microbiota. Food Chem 2025; 471:142857. [PMID: 39823906 DOI: 10.1016/j.foodchem.2025.142857] [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: 05/01/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
Paederia scandens (Lour.) Merrill flavonoid-rich extract (PSMF) has shown excellent xanthine oxidase (XOD) inhibitory activity in our previous study. However, the efficacy of PSMF in mitigating hyperuricemia (HUA) remains to be elucidated. In this study, we investigated the effects and mechanisms of PSMF on alleviating in HUA mice. The results showed that PSMF intervention reduced serum levels of uric acid (UA), creatinine (CRE), and blood urea nitrogen (BUN), and inhibited the activities of XOD and adenosine deaminase (ADA). In addition, PSMF treatment not only attenuated the inflammatory response and renal damage but also regulated the expression of UA synthesis genes and UA excretion genes. Finally, PSMF ameliorated gut microbiota dysbiosis in HUA mice by enriching the abundance of short-chain fatty acid (SCFA)-producing bacteria. In summary, PSMF appears to be a promising natural source for the prevention and treatment of HUA.
Collapse
Affiliation(s)
- Yuyi Liu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Shanling Sheng
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Linye Wu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Huixian Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hui Xue
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Ruimin Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
3
|
Ye X, Ren D, Chen Q, Shen J, Wang B, Wu S, Zhang H. Resolution of inflammation during rheumatoid arthritis. Front Cell Dev Biol 2025; 13:1556359. [PMID: 40206402 PMCID: PMC11979130 DOI: 10.3389/fcell.2025.1556359] [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: 01/06/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes synovial joint inflammation as well as bone destruction and erosion, typically characterized by joint pain, swelling, and stiffness, with complications and persistent pain after remission posing a significant health burden for RA patients. The etiology of RA has not yet been fully elucidated, but a large number of studies have shown that the initiation of inflammation in RA is closely related to T-cell activation, the production of a variety of pro-inflammatory cytokines, macrophage M1/M2 imbalance, homeostatic imbalance of the intestinal flora, fibroblast-like synoviocytes (FLSs) and synovial tissue macrophages (STMs) in the synovial lumen of joints that exhibit an aggressive phenotype. While the resolution of RA is less discussed, therefore, we provided a systematic review of the relevant remission mechanisms including blocking T cell activation, regulating macrophage polarization status, modulating the signaling pathway of FLSs, modulating the subpopulation of STMs, and inhibiting the relevant inflammatory factors, as well as the probable causes of persistent arthritis pain after the remission of RA and its pain management methods. Achieving resolution in RA is crucial for improving the quality of life and long-term prognosis of patients. Thus, understanding these mechanisms provide novel potential for further drug development and treatment of RA.
Collapse
Affiliation(s)
- Xiaoou Ye
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Dan Ren
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Qingyuan Chen
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Jiquan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Bo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| |
Collapse
|
4
|
Wang X, Pan L, Niu D, Zhou J, Shen M, Zeng Z, Gong W, Yang E, Tang Y, Cheng G, Sun C. Jingfang Granules alleviates the lipid peroxidation induced ferroptosis in rheumatoid arthritis rats by regulating gut microbiota and metabolism of short chain fatty acids. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119160. [PMID: 39608616 DOI: 10.1016/j.jep.2024.119160] [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: 09/30/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, bone and cartilage damage, musculoskeletal pain, swelling, and stiffness. Inflammation is one of the key factors that induce RA. Jingfang Granule (JFG) is a traditional Chinese medicine (TCM) with significant anti-inflammatory effects. Clinical studies have confirmed that JFG can be used to treat RA, but the mechanism is still vague. PURPOSE This study was designed to evaluate the protective function and the mechanism of JFG on rats with RA. STUDY DESIGN AND METHODS Complete Freud's Adjuvant (CFA) was used to establish a rat RA model, and JFG or Diclofenac Sodium (Dic) was orally administered. Foot swelling and hematoxylin eosin (H&E) staining were used to test the therapeutic effect of JFG on RA treatment, while ELISA kits were used to detect serum cytokines. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), and reactive oxygen species (ROS) were used to evaluate oxidative stress levels. The integration of label-free proteomics, fecal short chain fatty acid (SCFA) targeted metabolomics, peripheral blood SCFA, medium and long chain fatty acid targeted metabolomics, and 16S rDNA sequencing of gut microbiota were used to screen the mechanism. Western blot technology was used to validate the results of multiple omics studies. Serum D-Lactic acid, lipopolysaccharide specific IgA antibody (LPS IgA), diamine oxidase (DAO), and colon Claudin 5 and ZO-1 were used to evaluate the intestinal barrier. RESULTS The results confirmed that JFG effectively protected rats from RA injury, which was confirmed by improved foot swelling and synovial pathology. At the same time, JFG reduced the levels of TNF-α, IL-1β, and IL-6 in serum by inhibiting the NLRP3 inflammasome signaling pathway and TLR4/NF-κB signaling pathway in synovial tissue. Multiple omics studies indicated that JFG increased the abundance of gut microbiota and regulated the number of gut bacteria, thereby increased the levels of Acetic acid, Propionic acid, and Butyric acid in the gut and serum of RA rats, which activated AMPK to regulate fatty acid metabolism and fatty acid biosynthesis, thereby inhibited lipid oxidative stress induced ferroptosis to improve tissue damage caused by RA. Meanwhile, JFG improved the intestinal barrier by upregulating the expresses of Claudin 5 and ZO-1, which was confirmed by low concentrations of D-Lactic acid, LPS-SIgA and DAO in serum. CONCLUSIONS This study confirmed that JFG improved the disturbance of fatty acid metabolism by modulating gut microbiota and the production of fecal SCFAs to activate AMPK, and then inhibited ferroptosis caused by lipid oxidative stress in synovium tissue and prevented AR injury. This study proposes for the first time to investigate the mechanism of JFG treatment for RA from the perspective of the "Gut-joint" axis, and provides a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Xiuwen Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Wenqiao Gong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Enhua Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yunfeng Tang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guoliang Cheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
5
|
Wu Y, Cao Z, Liu W, Cahoon JG, Wang K, Wang P, Hu L, Chen Y, Moser M, Vella AT, Ley K, Wen L, Fan Z. Nanoscopy reveals integrin clustering reliant on kindlin-3 but not talin-1. Cell Commun Signal 2025; 23:12. [PMID: 39773732 PMCID: PMC11707915 DOI: 10.1186/s12964-024-02024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Neutrophils are the most abundant leukocytes in human blood, and their recruitment is essential for innate immunity and inflammatory responses. The initial and critical step of neutrophil recruitment is their adhesion to vascular endothelium, which depends on G protein-coupled receptor (GPCR) triggered integrin inside-out signaling that induces β2 integrin activation and clustering on neutrophils. Kindlin-3 and talin-1 are essential regulators for the inside-out signaling induced β2 integrin activation. However, their contribution in the inside-out signaling induced β2 integrin clustering is unclear because conventional assays on integrin clustering are usually performed on adhered cells, where integrin-ligand binding concomitantly induces integrin outside-in signaling. METHODS We used flow cytometry and quantitative super-resolution stochastic optical reconstruction microscopy (STORM) to quantify β2 integrin activation and clustering, respectively, in kindlin-3 and talin-1 knockout leukocytes. We also tested whether wildtype or Pleckstrin homology (PH) domain deleted kindlin-3 can rescue the kindlin-3 knockout phenotypes. RESULTS GPCR-triggered inside-out signaling alone can induce β2 integrin clustering. As expected, both kindlin-3 and talin-1 knockout decreases integrin activation. Interestingly, only kindlin-3 but not talin-1 contributes to integrin clustering in the scenario of inside-out-signaling, wherein a critical role of the PH domain of kindlin-3 was highlighted. CONCLUSIONS Since talin was known to facilitate integrin clustering in outside-in-signaling-involved cells, our finding provides a paradigm shift by suggesting that the molecular mechanisms of integrin clustering upon inside-out signaling and outside-in signaling are different. Our data also contradict the conventional assumption that integrin activation and clustering are tightly inter-connected by showing separated regulation of the two during inside-out signaling. Our study provides a new mechanism that shows kindlin-3 regulates β2 integrin clustering and suggests that integrin clustering should be assessed independently, aside from integrin activation, when studying leukocyte adhesion in inflammatory diseases.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Ziming Cao
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Wei Liu
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Jason G Cahoon
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Kepeng Wang
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Penghua Wang
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, Department of Pathology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Markus Moser
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Lai Wen
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, 89557, USA.
| | - Zhichao Fan
- Department of Immunology, University of Connecticut School of Medicine, Connecticut, Farmington, 06030, USA.
| |
Collapse
|
6
|
Hanata N, Kaplan MJ. The role of neutrophil extracellular traps in inflammatory rheumatic diseases. Curr Opin Rheumatol 2025; 37:64-71. [PMID: 39258603 PMCID: PMC11602361 DOI: 10.1097/bor.0000000000001054] [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] [Indexed: 09/12/2024]
Abstract
PURPOSE OF REVIEW Dysregulation in neutrophil extracellular trap (NET) formation and degradation has been reported in several inflammatory rheumatic diseases. This review summarizes the recent advances in the understanding the role of NETs in the context of inflammatory rheumatic diseases. RECENT FINDINGS NET formation is enhanced in peripheral blood of patients with large vessel vasculitis and polymyalgia rheumatica. NETs are detected in affected organs in autoimmune conditions, and they might play pathological roles in tissues. Several understudied medications and supplements suppress NET formation and ameliorate animal models of inflammatory rheumatic diseases. NETs and anti-NET antibodies have potential utility as disease biomarkers. SUMMARY Growing evidence has suggested the contribution of NET dysregulation to the pathogenesis of several inflammatory rheumatic diseases. Further research is warranted in regard to clinical impact of modulating aberrant NET formation and clearance in inflammatory rheumatic diseases.
Collapse
Affiliation(s)
- Norio Hanata
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Chasov V, Gilyazova E, Ganeeva I, Zmievskaya E, Davletshin D, Valiullina A, Bulatov E. Gut Microbiota Modulation: A Novel Strategy for Rheumatoid Arthritis Therapy. Biomolecules 2024; 14:1653. [PMID: 39766360 PMCID: PMC11674688 DOI: 10.3390/biom14121653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to joint inflammation, progressive tissue damage and significant disability, severely impacting patients' quality of life. While the exact mechanisms underlying RA remain elusive, growing evidence suggests a strong link between intestinal microbiota dysbiosis and the disease's development and progression. Differences in microbial composition between healthy individuals and RA patients point to the role of gut microbiota in modulating immune responses and promoting inflammation. Therapies targeting microbiota restoration have demonstrated promise in improving treatment efficacy, enhancing patient outcomes and slowing disease progression. However, the complex interplay between gut microbiota and autoimmune pathways in RA requires further investigation to establish causative relationships and mechanisms. Here, we review the current understanding of the gut microbiota's role in RA pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
8
|
Himcinschi ME, Uscatescu V, Gherghe G, Stoian I, Vlad A, Popa DC, Coriu D, Anghel A. The Role of Neutrophil Extracellular Traps in the Outcome of Malignant Epitheliomas: Significance of CA215 Involvement. Diagnostics (Basel) 2024; 14:328. [PMID: 38337844 PMCID: PMC10855654 DOI: 10.3390/diagnostics14030328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Neutrophil extracellular traps (NETs) were originally discovered as a part of the innate immune response of the host to bacteria. They form a web-like structure that can immobilize microorganisms or exhibit direct antimicrobial properties, such as releasing reactive oxygen species (ROS). NETs are established when neutrophils undergo a sort of cellular death following exposure to ROS, chemokines, cytokines, or other soluble factors. This process results in the release of the neutrophil's DNA in a web-like form, which is decorated with citrullinated histones (H3/H4-cit), neutrophil elastase (NE), and myeloperoxidase (MPO). Emerging studies have put into perspective that NETs play an important role in oncology as they were shown to influence tumor growth, malignant initiation, and proliferation, mediate the transition from endothelial to mesenchymal tissue, stimulate angiogenesis or metastasis, and can even help cancer cells evade the immune response. The role of NETs in cancer therapy resides in their ability to form and act as a mechanical barrier that will provide the primary tumor with a reduced response to irradiation or pharmaceutical penetration. Subsequently, cancer cells are shown to internalize NETs and use them as a strong antioxidant when pharmaceutical treatment is administered. In this review, we explored the role of NETs as part of the tumor microenvironment (TME), in the context of malignant epitheliomas, which are capable of an autonomous production of CA215, a subvariant of IgG, and part of the carcinoembryonic antigen (CEA) superfamily. Studies have shown that CA215 has a functional Fc subdivision able to activate the Fc-gamma-RS receptor on the surface of neutrophils. This activation may afterward stimulate the production of NETs, thus indicating CA215 as a potential factor in cancer therapy surveillance.
Collapse
Affiliation(s)
- Mihai Emanuel Himcinschi
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.E.H.); (A.A.)
| | - Valentina Uscatescu
- Department of Hematology, Fundeni Clinical Institute, 022328 Bucharest, Romania (D.C.)
| | - Georgiana Gherghe
- Department of Hematology, Fundeni Clinical Institute, 022328 Bucharest, Romania (D.C.)
| | - Irina Stoian
- Department of Functional Sciences I/Biochemistry, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Adelina Vlad
- Department of Functional Sciences I/Physiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Delia Codruța Popa
- Department of Hematology, Fundeni Clinical Institute, 022328 Bucharest, Romania (D.C.)
| | - Daniel Coriu
- Department of Hematology, Fundeni Clinical Institute, 022328 Bucharest, Romania (D.C.)
| | - Andrei Anghel
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.E.H.); (A.A.)
| |
Collapse
|