1
|
Liao XZ, Xie RX, Zheng SY, Fan CL, Zuo MY, Chen SX, Zhu JQ, Li J. Bioinformatics and molecular docking reveal Cryptotanshinone as the active anti-inflammation component of Qu-Shi-Xie-Zhuo decoction by inhibiting S100A8/A9-NLRP3-IL-1β signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156257. [PMID: 39631292 DOI: 10.1016/j.phymed.2024.156257] [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: 08/23/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Gout is a common type of arthritis marked by monosodium urate (MSU) crystal deposition in joints, triggering an inflammatory response. Qu-Shi-Xie-Zhuo (QSXZ), a traditional Chinese medicine (TCM) formula, has been clinically used for the treatment of gouty arthritis (GA). PURPOSE The study sought to examine the impact of QSXZ on GA and to delve into the pharmacological mechanisms that underlie its effects. METHODS The chemical constituents of QSXZ were analyzed through UPLC-MS. MSU-induced acute gouty arthritis (AGA) and subcutaneous (SC) air pouch models in mice were employed to evaluate the anti-inflammatory properties of QSXZ and its primary active compound, Cryptotanshinone (CTS). To investigate the therapeutic mechanisms of QSXZ, we used MS-based network pharmacology, transcriptomic analysis, molecular docking and multiscale bioassays. RESULTS Treatment of QSXZ revealed a significant reduction of inflammatory cell infiltration and the expression of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin -1β (IL-1β). Based on UPLC/MS/MS results, 49 components were considered the active ingredients of QSXZ. Network pharmacology analysis indicated that QSXZ regulates multiple inflammation-related pathways. Subsequent transcriptomic analysis showed that QSXZ regulates gene expression of S100A8 and S100A9. Our investigation observed an increased expression of S100A8 and S100A9 in monocytes derived from gout patients. Molecular docking and molecular dynamics simulation analysis revealed the binding pattern and interaction between QSXZ active compound CTS and S100A8/A9, and subsequent surface plasmon resonance (SPR) and cell thermal shift assay (CETSA) experiments verified the direct interaction between them. To investigate the mechanisms of action, we conducted RT-PCR, Western blotting, immunohistochemistry, flow cytometry, and measured the inflammatory response. Our findings highlight the pathogenic role of S100A8/A9 mediated TLR4-NLRP3 axis in gout and review outstanding therapeutic effects of QSXZ and its primary active compound CTS on MSU-induced experimental models. CONCLUSIONS In summary, this study substantiates the therapeutic potential of QSXZ and its primary active compound CTS, as promising alternative treatments for GA. Our findings provide valuable insight into the critical pharmacological mechanism of QSXZ in regulating inflammation, highlighting its potential therapeutic effects in GA management.
Collapse
Affiliation(s)
- Xiao-Zhong Liao
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Rui-Xia Xie
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Song-Yuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Cui-Ling Fan
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Meng-Yue Zuo
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Shi-Xian Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun-Qing Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
2
|
Sun C, Xie Y, Zhu C, Guo L, Xu B, Qin H, Li X. Prognostic value of serum Mrp 8/14 in sepsis-induced acute respiratory distress syndrome patients: a retrospective cohort study. PeerJ 2024; 12:e18718. [PMID: 39686985 PMCID: PMC11648679 DOI: 10.7717/peerj.18718] [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: 08/08/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Background Mrp 8/14 is abundantly secreted by activated neutrophils during infection and inflammation. However, its prognostic value in acute respiratory distress dyndrome (ARDS) induced by sepsis is poorly understood. Our aim was to investigate the relationship between serum Mrp 8/14 and the prognosis in sepsis-induced ARDS patients admitted to the intensive care unit (ICU). Methods Serum Mrp 8/14 concentrations were analyzed in 118 ARDS patients induced by sepsis included in the analytical study. Patients were enrolled upon admission to the ICU of Nanjing Hospital affiliated to Nanjing Medical University. The baseline information and clinical outcomes were obtained. Patients were divided into survivor group and non-survivor group according to whether they died during ICU hospitalization. Results The serum Mrp 8/14 levels were significantly increased in the non-survivor group compared to the survivor group (P < 0.05). Logistic regression analysis showed that serum Mrp 8/14, albumin and APACHE II were the independent factors for predicting the prognosis of sepsis-induced ARDS during ICU hospitalization after adjustment. Additionally, the area under the receiver operating characteristic curve for Mrp 8/14 combined with albumin was associated with ICU mortality and was higher than that of Mrp 8/14, albumin, APACHE II and Mrp 8/14 combined with APACHE II (all P < 0.05). A nomogram was constructed to predict ICU mortality and the c-indexes of predictive accuracy was 0.830 in the cohort (P < 0.05). Conclusions The serum Mrp 8/14 upon ICU admission in septic patients may be useful for predicting mortality in sepsis-induced ARDS patients during ICU hospitalization.
Collapse
Affiliation(s)
- Caizhi Sun
- Department of Emergency Medicine, Lianyungang Clinical College, Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongpeng Xie
- Department of Emergency Medicine, Lianyungang Clinical College, Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Chenchen Zhu
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Guo
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bowen Xu
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haidong Qin
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaomin Li
- Department of Emergency Medicine, Lianyungang Clinical College, Nanjing Medical University, Lianyungang, Jiangsu, China
| |
Collapse
|
3
|
Tian Q, Li Z, Yan Z, Jiang S, Zhao X, Wang L, Li M. Inflammatory role of S100A8/A9 in the central nervous system non-neoplastic diseases. Brain Res Bull 2024; 218:111100. [PMID: 39396712 DOI: 10.1016/j.brainresbull.2024.111100] [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: 07/17/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
S100A8 (MRP8) and S100A9 (MRP14) are critical mediators of the inflammatory response; they are usually present as heterodimers because of the instability of homodimers. Studies have demonstrated that S100A8/A9 expression is significantly upregulated in several central nervous system (CNS) diseases. S100A8/A9 is actively released by neutrophils and monocytes; it plays a key role in regulating the inflammatory response by stimulating leukocyte recruitment and inducing cytokine secretion during inflammation. Additionally, S100A8/A9 can be used as a diagnostic biomarker for several CNS diseases and as a predictor of therapeutic response to inflammation-related diseases. In this work, we reviewed our current understanding of S100A8/A9 overexpression in inflammation and its importance in the development and progression of CNS inflammatory diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and stroke, and the functional roles and therapeutic applications of S100A8/A9 in these diseases. Finally, we discussed the current barriers and future research directions of S100A8/A9 in CNS diseases.
Collapse
Affiliation(s)
- Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Zhijie Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Ziang Yan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Shengming Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Xincan Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Lei Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, China; Department of Neurosurgery, Yichang Central People's Hospital, Yichang, Hubei, China.
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
4
|
Li R, Colombo M, Wang G, Rodriguez-Romera A, Benlabiod C, Jooss NJ, O’Sullivan J, Brierley CK, Clark SA, Pérez Sáez JM, Aragón Fernández P, Schoof EM, Porse B, Meng Y, Khan AO, Wen S, Dong P, Zhou W, Sousos N, Murphy L, Clarke M, Olijnik AA, C. Wong Z, Karali CS, Sirinukunwattana K, Ryou H, Norfo R, Cheng Q, Carrelha J, Ren Z, Thongjuea S, Rathinam VA, Krishnan A, Royston D, Rabinovich GA, Mead AJ, Psaila B. A proinflammatory stem cell niche drives myelofibrosis through a targetable galectin-1 axis. Sci Transl Med 2024; 16:eadj7552. [PMID: 39383242 PMCID: PMC7616771 DOI: 10.1126/scitranslmed.adj7552] [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: 07/15/2023] [Revised: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
Myeloproliferative neoplasms are stem cell-driven cancers associated with a large burden of morbidity and mortality. Most patients present with early-stage disease, but a substantial proportion progress to myelofibrosis or secondary leukemia, advanced cancers with a poor prognosis and high symptom burden. Currently, it remains difficult to predict progression, and therapies that reliably prevent or reverse fibrosis are lacking. A major bottleneck to the discovery of disease-modifying therapies has been an incomplete understanding of the interplay between perturbed cellular and molecular states. Several cell types have individually been implicated, but a comprehensive analysis of myelofibrotic bone marrow is lacking. We therefore mapped the cross-talk between bone marrow cell types in myelofibrotic bone marrow. We found that inflammation and fibrosis are orchestrated by a "quartet" of immune and stromal cell lineages, with basophils and mast cells creating a TNF signaling hub, communicating with megakaryocytes, mesenchymal stromal cells, and proinflammatory fibroblasts. We identified the β-galactoside-binding protein galectin-1 as a biomarker of progression to myelofibrosis and poor survival in multiple patient cohorts and as a promising therapeutic target, with reduced myeloproliferation and fibrosis in vitro and in vivo and improved survival after galectin-1 inhibition. In human bone marrow organoids, TNF increased galectin-1 expression, suggesting a feedback loop wherein the proinflammatory myeloproliferative neoplasm clone creates a self-reinforcing niche, fueling progression to advanced disease. This study provides a resource for studying hematopoietic cell-niche interactions, with relevance for cancer-associated inflammation and disorders of tissue fibrosis.
Collapse
Affiliation(s)
- Rong Li
- CAMS Oxford Institute; University of Oxford; Oxford, United Kingdom (UK)
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Michela Colombo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Human Technopole; Milan, Italy
| | - Guanlin Wang
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- MRC WIMM Centre for Computational Biology, University of Oxford; Oxford, United Kingdom
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
- Qizhi Institute, Shanghai, China
| | - Antonio Rodriguez-Romera
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Camelia Benlabiod
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Natalie J. Jooss
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Jennifer O’Sullivan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Charlotte K. Brierley
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Sally-Ann Clark
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Juan M. Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | - Erwin M. Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark; Denmark
| | - Bo Porse
- The Finsen Laboratory, Copenhagen University Hospital; Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Department of Clinical Medicine, University of Copenhagen; Copenhagen, Denmark
| | - Yiran Meng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Abdullah O. Khan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Sean Wen
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Pengwei Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Nikolaos Sousos
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Lauren Murphy
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Matthew Clarke
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Aude-Anais Olijnik
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Zoë C. Wong
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Christina Simoglou Karali
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Korsuk Sirinukunwattana
- Oxford Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford; Oxford, UK
| | - Hosuk Ryou
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford; Oxford, UK
| | - Ruggiero Norfo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Qian Cheng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford; Oxford, UK
| | - Zemin Ren
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Supat Thongjuea
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine; Farmington, ConnecticutUSA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, California, USA
| | - Daniel Royston
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adam J Mead
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Bethan Psaila
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
De Luca G, Goette NP, Lev PR, Baroni Pietto MC, Marin Oyarzún CP, Castro Ríos MA, Moiraghi B, Sackmann F, Kamiya LJ, Verri V, Caula V, Fernandez V, Vicente A, Pose Cabarcos J, Caruso V, Camacho MF, Larripa IB, Khoury M, Marta RF, Glembotsky AC, Heller PG. Elevated levels of damage-associated molecular patterns HMGB1 and S100A8/A9 coupled with toll-like receptor-triggered monocyte activation are associated with inflammation in patients with myelofibrosis. Front Immunol 2024; 15:1365015. [PMID: 39391311 PMCID: PMC11465240 DOI: 10.3389/fimmu.2024.1365015] [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: 01/03/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Inflammation plays a pivotal role in the pathogenesis of primary and post-essential thrombocythemia or post-polycythemia vera myelofibrosis (MF) in close cooperation with the underlying molecular drivers. This inflammatory state is induced by a dynamic spectrum of inflammatory cytokines, although recent evidence points to the participation of additional soluble inflammatory mediators. Damage-associated molecular patterns (DAMPs) represent endogenous signals released upon cell death or damage which trigger a potent innate immune response. We assessed the contribution of two prototypical DAMPs, HMGB1 and S100A8/A9, to MF inflammation. Circulating HMGB1 and S100A8/A9 were elevated in MF patients in parallel to the degree of systemic inflammation and levels increased progressively during advanced disease stages. Patients with elevated DAMPs had higher frequency of adverse clinical features, such as anemia, and inferior survival, suggesting their contribution to disease progression. Monocytes, which are key players in MF inflammation, were identified as a source of S100A8/A9 but not HMGB1 release, while both DAMPs correlated with cell death parameters, such as serum LDH and cell-free DNA, indicating that passive release is an additional mechanism leading to increased DAMPs. HMGB1 and S100A8/A9 promote inflammation through binding to Toll-like receptor (TLR) 4, whereas the former also binds TLR2. Monocytes from MF patients were shown to be hyperactivated at baseline, as reflected by higher CD11b and tissue factor exposure and increased expression levels of proinflammatory cytokines IL-1β and IL-6. Patient monocytes showed preserved TLR4 and TLR2 expression and were able to mount normal or even exacerbated functional responses and cytokine upregulation following stimulation of TLR4 and TLR2. Elevated levels of endogenous TLR ligands HMGB1 and S100A8/A9 coupled to the finding of preserved or hyperreactive TLR-triggered responses indicate that DAMPs may promote monocyte activation and cytokine production in MF, fueling inflammation. Plasma IL-1β and IL-6 were elevated in MF and correlated with DAMPs levels, raising the possibility that DAMPs could contribute to cytokine generation in vivo. In conclusion, this study highlights that, in cooperation with classic proinflammatory cytokines, DAMPs represent additional inflammatory mediators that may participate in the generation of MF inflammatory state, potentially providing novel biomarkers of disease progression and new therapeutic targets.
Collapse
Affiliation(s)
- Geraldine De Luca
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Nora P. Goette
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Paola R. Lev
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria C. Baroni Pietto
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cecilia P. Marin Oyarzún
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | | | | - Laureano J. Kamiya
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Veronica Verri
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Caula
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanina Fernandez
- Departamento de Hematología, Hospital Posadas, Buenos Aires, Argentina
| | - Angeles Vicente
- Departamento de Hematología, Hospital Alemán, Buenos Aires, Argentina
| | - Julio Pose Cabarcos
- Departamento de Hematología, Sanatorio Otamendi Miroli, Buenos Aires, Argentina
| | - Vanesa Caruso
- Departamento de Hematología, Hospital Piñero, Buenos Aires, Argentina
| | - Maria F. Camacho
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene B. Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marina Khoury
- Departamento de Docencia e Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosana F. Marta
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana C. Glembotsky
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula G. Heller
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
6
|
Wang JC, Shi G, Chen C, Wong C, Gotlieb V, Joseph G, Nair KV, Boyapati L, Ladan E, Symanowski JT, Sun L. TLR2 Derangements Likely Play a Significant Role in the Inflammatory Response and Thrombosis in Patients with Ph(-) Classical Myeloproliferative Neoplasm. Mediators Inflamm 2024; 2024:1827127. [PMID: 39157201 PMCID: PMC11329310 DOI: 10.1155/2024/1827127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/17/2024] [Accepted: 06/17/2024] [Indexed: 08/20/2024] Open
Abstract
We investigated the role of toll-like receptors (TLRs) in inflammatory pathways in Philadelphia chromosome-negative myeloproliferative neoplasms (Ph(-)MPNs). TLR2 expression was increased in ET, PV, and MPN (grouped as (PV + (ET) + MF)), whereas TLR4 was elevated only in MPN. TLR3, 7, and 9 were not elevated. Cultured monocyte-derived dendritic cells and plasma assays in TLR2-elevated patients were found to secrete more cytokines than those from TLR2-normal patients. These facts suggest that TLR2 is the major inflammatory pathways in MPN. We also measured S100A9 and reactive oxygen species (ROS), revealing increased S100A9 in PV, MF, and MPN, while ROS were only increased in MF. These data suggests that MPNs initially involve TLR2, with minor contributions from TLR4, and with S100A9, leading to ROS formation, JAK2 mutation, and progression to MF or leukemia. Furthermore, patients with JAK2 mutations or leukocytosis exhibited higher TLR2 expression. In leukocyte-platelet interactions, cells from MPN patients displayed a stronger response to a TLR2 agonist than TLR4 agonist. A TLR2 inhibitor (but not a TLR4 inhibitor) attenuated this response. Thrombosis incidence was higher in TLR2-elevated patients (29%) than in TLR2-normal patients (19%). These findings suggest that TLR2 likely contributes to thrombosis in MPN.
Collapse
Affiliation(s)
- Jen Chin Wang
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Guanfang Shi
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Chi Chen
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Ching Wong
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Vladimir Gotlieb
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Gardith Joseph
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Kiron V Nair
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Lakshmi Boyapati
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Enayati Ladan
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - James T. Symanowski
- Department of Biostatistics and Data SciencesLevine Cancer Institute, Charlotte, NC, USA
| | - Lishi Sun
- Division of Hematology/OncologyBrookdale University Hospital Medical Center, Brooklyn, NY, USA
| |
Collapse
|
7
|
Wang Y, Shi Y, Shao Y, Lu X, Zhang H, Miao C. S100A8/A9 hi neutrophils induce mitochondrial dysfunction and PANoptosis in endothelial cells via mitochondrial complex I deficiency during sepsis. Cell Death Dis 2024; 15:462. [PMID: 38942784 PMCID: PMC11213914 DOI: 10.1038/s41419-024-06849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 06/30/2024]
Abstract
S100a8/a9, largely released by polymorphonuclear neutrophils (PMNs), belongs to the S100 family of calcium-binding proteins and plays a role in a variety of inflammatory diseases. Although S100a8/a9 has been reported to trigger endothelial cell apoptosis, the mechanisms of S100a8/a9-induced endothelial dysfunction during sepsis require in-depth research. We demonstrate that high expression levels of S100a8/a9 suppress Ndufa3 expression in mitochondrial complex I via downregulation of Nrf1 expression. Mitochondrial complex I deficiency contributes to NAD+-dependent Sirt1 suppression, which induces mitochondrial disorders, including excessive fission and blocked mitophagy, and mtDNA released from damaged mitochondria ultimately activates ZBP1-mediated PANoptosis in endothelial cells. Moreover, based on comprehensive scRNA-seq and bulk RNA-seq analyses, S100A8/A9hi neutrophils are closely associated with the circulating endothelial cell count (a useful marker of endothelial damage), and S100A8 is an independent risk factor for poor prognosis in sepsis patients.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xihua Lu
- Department of Anesthesiology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Zeng W, Gao Y, Wang Q, Chi J, Zhu Z, Diao Q, Li X, Wang Z, Qu M, Shi Y. Preliminary clinical analysis and pathway study of S100A8 as a biomarker for the diagnosis of acute deep vein thrombosis. Sci Rep 2024; 14:13298. [PMID: 38858401 PMCID: PMC11164926 DOI: 10.1038/s41598-024-61728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
Herein, we aimed to identify blood biomarkers that compensate for the poor specificity of D-dimer in the diagnosis of deep vein thrombosis (DVT). S100A8 was identified by conducting protein microarray analysis of blood samples from patients with and without DVT. We used ELISA to detect S100A8, VCAM-1, and ICAM-1 expression levels in human blood and evaluated their correlations. Additionally, we employed human recombinant protein S100A8 to induce human umbilical vein endothelial cells and examined the role of the TLR4/MAPK/VCAM-1 and ICAM-1 signaling axes in the pathogenic mechanism of S100A8. Simultaneously, we constructed a rat model of thrombosis induced by inferior vena cava stenosis and detected levels of S100A8, VCAM-1, and ICAM-1 in the blood of DVT rats using ELISA. The associations of thrombus tissue, neutrophils, and CD68-positive cells with S100A8 and p38MAPK, TLR4, and VCAM-1 expression levels in vein walls were explored. The results revealed that blood S100A8 was significantly upregulated during the acute phase of DVT and activated p38MAPK expression by combining with TLR4 to enhance the expression and secretion of VCAM-1 and ICAM-1, thereby affecting the occurrence and development of DVT. Therefore, S100A8 could be a potential biomarker for early diagnosis and screening of DVT.
Collapse
Affiliation(s)
- Wenjie Zeng
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Yangyang Gao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qitao Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Junyu Chi
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ziyan Zhu
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qingfei Diao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Xin Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhen Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ming Qu
- Vascular Gland Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Yongquan Shi
- Department of Clinical Laboratory Center, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| |
Collapse
|
9
|
Sun C, Xie Y, Zhu C, Guo L, Wei J, Xu B, Song Y, Qin H, Li X. Serum Mrp 8/14 as a Potential Biomarker for Predicting the Occurrence of Acute Respiratory Distress Syndrome Induced by Sepsis: A Retrospective Controlled Study. J Inflamm Res 2024; 17:2939-2949. [PMID: 38764498 PMCID: PMC11100500 DOI: 10.2147/jir.s457547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Background To date, there are no studies regarding the Mrp 8/14 in predicting the occurrence of acute respiratory distress syndrome (ARDS) induced by sepsis. Thus, the objective of this study was to investigate the expression of Myeloid-related proteins 8 and 14 (Mrp 8/14) and its role in ARDS induced by sepsis. Methods A total of 168 septic patients were enrolled in the observational study. The baseline information and clinical outcomes were obtained retrospectively. Serum Mrp 8/14 level was determined by enzyme linked immunosorbent assay (ELISA). The patients were categorized into sepsis and ARDS group based on whether they developed ARDS during the intensive care unit (ICU) hospitalization. Results There was significant difference in the level of Mrp 8/14 between the sepsis group and ARDS groups (P < 0.05). Mrp 8/14 correlated positively with procalcitonin (PCT), interleukin-6 (IL-6), acute physiology and chronic health evaluation II (APACHE II) score, sequential organ failure assessment (SOFA) score on day 1, mechanical ventilation time, length of ICU stay and hospitalization expenses in ICU (all P < 0.05). Logistic regression analysis showed Mrp 8/14 was the independent factor for forecasting the occurrence of sepsis- induced ARDS (P < 0.05). The areas under receiver operating characteristic curves for Mrp 8/14 were higher than that of PCT, APACHE II score and SOFA score on day 1 (P < 0.05). Conclusion The serum Mrp 8/14 level at admission may be a potential marker for predicting the occurrence of ARDS induced by sepsis. Early detection of serum Mrp 8/14 could help clinicians to identify and evaluate the severity of ARDS induced by sepsis.
Collapse
Affiliation(s)
- Caizhi Sun
- Department of Emergency Medicine, Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang City, Lianyungang, Jiangsu, 222000, People’s Republic of China
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Yongpeng Xie
- Department of Emergency Medicine, Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang City, Lianyungang, Jiangsu, 222000, People’s Republic of China
| | - Chenchen Zhu
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Lei Guo
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Jingjing Wei
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Bowen Xu
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Yang Song
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Haidong Qin
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing City, Jiangsu Province, 210006, People’s Republic of China
| | - Xiaomin Li
- Department of Emergency Medicine, Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang City, Lianyungang, Jiangsu, 222000, People’s Republic of China
| |
Collapse
|
10
|
Zhang P, You N, Ding Y, Zhu W, Wang N, Xie Y, Huang W, Ren Q, Qin T, Fu R, Zhang L, Xiao Z, Cheng T, Ma X. Gadd45g insufficiency drives the pathogenesis of myeloproliferative neoplasms. Nat Commun 2024; 15:2989. [PMID: 38582902 PMCID: PMC10998908 DOI: 10.1038/s41467-024-47297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
Despite the identification of driver mutations leading to the initiation of myeloproliferative neoplasms (MPNs), the molecular pathogenesis of MPNs remains incompletely understood. Here, we demonstrate that growth arrest and DNA damage inducible gamma (GADD45g) is expressed at significantly lower levels in patients with MPNs, and JAK2V617F mutation and histone deacetylation contribute to its reduced expression. Downregulation of GADD45g plays a tumor-promoting role in human MPN cells. Gadd45g insufficiency in the murine hematopoietic system alone leads to significantly enhanced growth and self-renewal capacity of myeloid-biased hematopoietic stem cells, and the development of phenotypes resembling MPNs. Mechanistically, the pathogenic role of GADD45g insufficiency is mediated through a cascade of activations of RAC2, PAK1 and PI3K-AKT signaling pathways. These data characterize GADD45g deficiency as a novel pathogenic factor in MPNs.
Collapse
Affiliation(s)
- Peiwen Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Na You
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yiyi Ding
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wenqi Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Nan Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yueqiao Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wanling Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tiejun Qin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.
| | - Xiaotong Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| |
Collapse
|
11
|
Wang T, Zhao C, Zhang J, Li S, Zhang Y, Gong Y, Zhou Y, Yan L, Zhang S, Zhang Z, Hu H, Liu A, Bai X, Zou Z. Whitening of brown adipose tissue inhibits osteogenic differentiation via secretion of S100A8/A9. iScience 2024; 27:108857. [PMID: 38303710 PMCID: PMC10830855 DOI: 10.1016/j.isci.2024.108857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
The mechanism by which brown adipose tissue (BAT) regulates bone metabolism is unclear. Here, we reveal that BAT secretes S100A8/A9, a previously unidentified BAT adipokine (batokine), to impair bone formation. Brown adipocytes-specific knockout of Rheb (RhebBAD KO), the upstream activator of mTOR, causes BAT malfunction to inhibit osteogenesis. Rheb depletion induces NF-κB dependent S100A8/A9 secretion from brown adipocytes, but not from macrophages. In wild-type mice, age-related Rheb downregulation in BAT is associated with enhanced S100A8/A9 secretion. Either batokines from RhebBAD KO mice, or recombinant S100A8/A9, inhibits osteoblast differentiation of mesenchymal stem cells in vitro by targeting toll-like receptor 4 on their surfaces. Conversely, S100A8/A9 neutralization not only rescues the osteogenesis repressed in the RhebBAD KO mice, but also alleviates age-related osteoporosis in wild-type mice. Collectively, our data revealed an unexpected BAT-bone crosstalk driven by Rheb-S100A8/A9, uncovering S100A8/A9 as a promising target for the treatment, and potentially, prevention of osteoporosis.
Collapse
Affiliation(s)
- Ting Wang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chaoran Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiahuan Zhang
- Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Youming Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Gong
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yingyue Zhou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lei Yan
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopadics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongling Hu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Anling Liu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhipeng Zou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Manan MR, Kipkorir V, Nawaz I, Waithaka MW, Srichawla BS, Găman AM, Diaconu CC, Găman MA. Acute myocardial infarction in myeloproliferative neoplasms. World J Cardiol 2023; 15:571-581. [PMID: 38058401 PMCID: PMC10696206 DOI: 10.4330/wjc.v15.i11.571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of hematologic malignancies characterized by an abnormal proliferation of cells of the myeloid lineage. Affected individuals are at increased risk for cardiovascular and thrombotic events. Myocardial infarction (MI) may be one of the earliest clinical manifestations of MPNs or may be a thrombotic complication that develops during the natural course of the disease. In the present review, we examine the epidemiology, pathogenesis, clinical presentation, and management of MI in MPNs based on the available literature. Moreover, we review potential biomarkers that could mediate the MI-MPNs crosstalk, from classical biochemical tests, e.g., lactate dehydrogenase, creatine kinase and troponins, to pro-inflammatory cytokines, oxidative stress markers, and clonal hematopoiesis.
Collapse
Affiliation(s)
| | - Vincent Kipkorir
- Department of Human Anatomy and Physiology, University of Nairobi, Nairobi 00100, Kenya
| | - Iqra Nawaz
- Quaid-e-Azam Medical College, Bahawalpur 63100, Pakistan
| | | | - Bahadar Singh Srichawla
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, Craiova 200143, Romania
- Clinic of Hematology, Filantropia City Hospital, Craiova 200143, Romania
| | - Camelia Cristina Diaconu
- Department of Internal Medicine, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Internal Medicine Clinic, Clinical Emergency Hospital of Bucharest, Bucharest 105402, Romania
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest 030304, Romania.
| |
Collapse
|
13
|
Luo Z, Wei Z, Zhang G, Chen H, Li L, Kang X. Achilles' Heel-The Significance of Maintaining Microenvironmental Homeostasis in the Nucleus Pulposus for Intervertebral Discs. Int J Mol Sci 2023; 24:16592. [PMID: 38068915 PMCID: PMC10706299 DOI: 10.3390/ijms242316592] [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: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
The dysregulation of intracellular and extracellular environments as well as the aberrant expression of ion channels on the cell membrane are intricately linked to a diverse array of degenerative disorders, including intervertebral disc degeneration. This condition is a significant contributor to low back pain, which poses a substantial burden on both personal quality of life and societal economics. Changes in the number and function of ion channels can disrupt the water and ion balance both inside and outside cells, thereby impacting the physiological functions of tissues and organs. Therefore, maintaining ion homeostasis and stable expression of ion channels within the cellular microenvironment may prove beneficial in the treatment of disc degeneration. Aquaporin (AQP), calcium ion channels, and acid-sensitive ion channels (ASIC) play crucial roles in regulating water, calcium ions, and hydrogen ions levels. These channels have significant effects on physiological and pathological processes such as cellular aging, inflammatory response, stromal decomposition, endoplasmic reticulum stress, and accumulation of cell metabolites. Additionally, Piezo 1, transient receptor potential vanilloid type 4 (TRPV4), tension response enhancer binding protein (TonEBP), potassium ions, zinc ions, and tungsten all play a role in the process of intervertebral disc degeneration. This review endeavors to elucidate alterations in the microenvironment of the nucleus pulposus during intervertebral disc degeneration (IVDD), with a view to offer novel insights and approaches for exploring therapeutic interventions against disc degeneration.
Collapse
Affiliation(s)
- Zhangbin Luo
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Ziyan Wei
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Haiwei Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|
14
|
Yu J, Zhao B, Pi Q, Zhou G, Cheng Z, Qu C, Wang X, Kong L, Luo S, Du D, Guo Y. Deficiency of S100A8/A9 attenuates pulmonary microvascular leakage in septic mice. Respir Res 2023; 24:288. [PMID: 37978525 PMCID: PMC10655323 DOI: 10.1186/s12931-023-02594-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND We have reported a positive correlation between S100 calcium-binding protein (S100) A8/S100A9 and sepsis-induced lung damage before. However, limited knowledge exists concerning the biological role of S100A8/A9 in pulmonary vascular endothelial barrier dysfunction, as well as the diagnostic value of S100A8/A9 in sepsis. METHODS Sepsis was induced in C57BL/6J mice and S100A9-knockout (KO) mice through the cecal ligation and puncture (CLP). Pulmonary vascular leakage was determined by measuring extravasated Evans blue (EB). Reverse transcription polymerase chain reaction and the histological score were used to evaluate inflammation and lung injury, respectively. Recombinant S100A8/A9 (rhS100A8/A9) was used to identify the effects of S100A8/A9 on endothelial barrier dysfunction in human umbilical vein endothelial cells (HUVECs). Additionally, the diagnostic value of S100A8/A9 in sepsis was assessed using receiver operating characteristic. RESULTS S100A8/A9 expression was up-regulated in the lungs of CLP-operated mice. S100A9 KO significantly reversed CLP-induced hypothermia and hypotension, resulting in an improved survival rate. S100A9 KO also decreased the inflammatory response, EB leakage, and histological scores in the lungs of CLP-operated mice. Occludin and VE-cadherin expressions were decreased in the lungs of CLP-operated mice; However, S100A9 KO attenuated this decrease. Moreover, CLP-induced signal transducer and activator of transcription 3 (STAT3) and p38/extracellular signal-regulated kinase (ERK) signalling activation and apoptosis were mitigated by S100A9 KO in lungs. In addition, rhS100A8/A9 administration significantly decreased occludin and VE-cadherin expressions, increased the phosphorylated (p)-ERK/ERK, p-p38/p38, and B-cell leukaemia/lymphoma 2 protein (Bcl-2)-associated X protein/Bcl-2 ratios in HUVECs. CONCLUSION The present study demonstrated S100A8/A9 aggravated sepsis-induced pulmonary inflammation, vascular permeability, and lung injury. This was achieved, at least partially, by activating the P38/STAT3/ERK signalling pathways. Moreover, S100A8/A9 showed the potential as a biomarker for sepsis diagnosis.
Collapse
Affiliation(s)
- Jiang Yu
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400010, China
| | - Qiangzhong Pi
- Department of Respiratory Medicine, Southwest Hospital, Army Military Medical University, Chongqing, P.R. China
| | - Guoxiang Zhou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhe Cheng
- Department of Cardiology, Chongqing University three Gorges Hospital, Chongqing, 404199, China
| | - Can Qu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400010, China
| | - Suxin Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dingyuan Du
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China.
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400010, China.
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
15
|
De Luca G, Lev PR, Camacho MF, Goette NP, Sackmann F, Castro Ríos MA, Moiraghi B, Cortes Guerrieri V, Bendek G, Carricondo E, Enrico A, Vallejo V, Varela A, Khoury M, Gutierrez M, Larripa IB, Marta RF, Glembotsky AC, Heller PG. High cell-free DNA is associated with disease progression, inflammasome activation and elevated levels of inflammasome-related cytokine IL-18 in patients with myelofibrosis. Front Immunol 2023; 14:1161832. [PMID: 38035089 PMCID: PMC10687201 DOI: 10.3389/fimmu.2023.1161832] [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: 02/08/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Myelofibrosis (MF) is a clonal hematopoietic stem cell disorder classified among chronic myeloproliferative neoplasms, characterized by exacerbated myeloid and megakaryocytic proliferation and bone marrow fibrosis. It is induced by driver (JAK2/CALR/MPL) and high molecular risk mutations coupled to a sustained inflammatory state that contributes to disease pathogenesis. Patient outcome is determined by stratification into risk groups and refinement of current prognostic systems may help individualize treatment decisions. Circulating cell-free (cf)DNA comprises short fragments of double-stranded DNA, which promotes inflammation by stimulating several pathways, including inflammasome activation, which is responsible for IL-1β and IL-18 maturation and release. In this work, we assessed the contribution of cfDNA as a marker of disease progression and mediator of inflammation in MF. cfDNA was increased in MF patients and higher levels were associated with adverse clinical outcome, a high-risk molecular profile, advanced disease stages and inferior overall survival, indicating its potential value as a prognostic marker. Cell-free DNA levels correlated with tumor burden parameters and markers of systemic inflammation. To mimic the effects of cfDNA, monocytes were stimulated with poly(dA:dT), a synthetic double-stranded DNA. Following stimulation, patient monocytes released higher amounts of inflammasome-processed cytokine, IL-18 to the culture supernatant, reflecting enhanced inflammasome function. Despite overexpression of cytosolic DNA inflammasome sensor AIM2, IL-18 release from MF monocytes was shown to rely mainly on the NLRP3 inflammasome, as it was prevented by NLRP3-specific inhibitor MCC950. Circulating IL-18 levels were increased in MF plasma, reflecting in vivo inflammasome activation, and highlighting the previously unrecognized involvement of this cytokine in MF cytokine network. Monocyte counts were higher in patients and showed a trend towards correlation with IL-18 levels, suggesting monocytes represent a source of circulating IL-18. The close correlation shown between IL-18 and cfDNA levels, together with the finding of enhanced DNA-triggered IL-18 release from monocytes, suggest that cfDNA promotes inflammation, at least in part, through inflammasome activation. This work highlights cfDNA, the inflammasome and IL-18 as additional players in the complex inflammatory circuit that fosters MF progression, potentially providing new therapeutic targets.
Collapse
Affiliation(s)
- Geraldine De Luca
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paola R. Lev
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria F. Camacho
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Nora P. Goette
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | | | | | - Beatriz Moiraghi
- Departamento de Hematología, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Veronica Cortes Guerrieri
- División Hematología Clínica, IDIM Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Georgina Bendek
- Departamento de Hematología, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Emiliano Carricondo
- Departamento de Hematología, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Alicia Enrico
- Departamento de Hematología, Hospital Italiano de La Plata, Buenos Aires, Argentina
| | - Veronica Vallejo
- Departamento de Hematología, Instituto Cardiovascular de Buenos Aires, Buenos Aires, Argentina
| | - Ana Varela
- Departamento de Hematología, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Marina Khoury
- Departamento de Docencia e Investigación, IDIM Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marina Gutierrez
- Unidad Genómica, Laboratorio Stamboulian, Buenos Aires, Argentina
| | - Irene B. Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Rosana F. Marta
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana C. Glembotsky
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula G. Heller
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Kelliher S, Gamba S, Weiss L, Shen Z, Marchetti M, Schieppati F, Scaife C, Madden S, Bennett K, Fortune A, Maung S, Fay M, Ní Áinle F, Maguire P, Falanga A, Kevane B, Krishnan A. Platelet proteo-transcriptomic profiling validates mediators of thrombosis and proteostasis in patients with myeloproliferative neoplasms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563619. [PMID: 37961700 PMCID: PMC10634751 DOI: 10.1101/2023.10.23.563619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Patients with chronic Myeloproliferative Neoplasms (MPN) including polycythemia vera (PV) and essential thrombocythemia (ET) exhibit unique clinical features, such as a tendency toward thrombosis and hemorrhage, and risk of disease progression to secondary bone marrow fibrosis and/or acute leukemia. Although an increase in blood cell lineage counts (quantitative features) contribute to these morbid sequelae, the significant qualitative abnormalities of myeloid cells that contribute to vascular risk are not well understood. Here, we address this critical knowledge gap via a comprehensive and untargeted profiling of the platelet proteome in a large (n= 140) cohort of patients (from two independent sites) with an established diagnosis of PV and ET (and complement prior work on the MPN platelet transcriptome from a third site). We discover distinct MPN platelet protein expression and confirm key molecular impairments associated with proteostasis and thrombosis mechanisms of potential relevance to MPN pathology. Specifically, we validate expression of high-priority candidate markers from the platelet transcriptome at the platelet proteome (e.g., calreticulin (CALR), Fc gamma receptor (FcγRIIA) and galectin-1 (LGALS1) pointing to their likely significance in the proinflammatory, prothrombotic and profibrotic phenotypes in patients with MPN. Together, our proteo-transcriptomic study identifies the peripherally-derived platelet molecular profile as a potential window into MPN pathophysiology and demonstrates the value of integrative multi-omic approaches in gaining a better understanding of the complex molecular dynamics of disease.
Collapse
Affiliation(s)
- Sarah Kelliher
- School of Medicine, University College Dublin, Dublin, Ireland
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Sara Gamba
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Luisa Weiss
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Zhu Shen
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina Marchetti
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Schieppati
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Caitriona Scaife
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kathleen Bennett
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Anne Fortune
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Su Maung
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michael Fay
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland
| | - Patricia Maguire
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, University College Dublin, Dublin, Ireland
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
- University of Milano-Bicocca, Department of Medicine and Surgery, Monza, Italy
| | - Barry Kevane
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Anandi Krishnan
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Rutgers University, Piscataway, NJ
- Stanford Cancer Institute, Stanford, CA, USA
| |
Collapse
|
17
|
Silva de Melo BM, Veras FP, Zwicky P, Lima D, Ingelfinger F, Martins TV, da Silva Prado D, Schärli S, Publio G, Hiroki CH, Melo PH, Saraiva A, Norbiato T, Lima L, Ryffel B, Vogl T, Roth J, Waisman A, Nakaya HI, da Silva Souza C, Cunha FQ, Cunha TM, Becher B, Alves-Filho JC. S100A9 Drives the Chronification of Psoriasiform Inflammation by Inducing IL-23/Type 3 Immunity. J Invest Dermatol 2023; 143:1678-1688.e8. [PMID: 36921684 DOI: 10.1016/j.jid.2023.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023]
Abstract
Psoriasis is a chronic inflammatory skin disorder driven by the IL-23/type 3 immune response. However, molecular mechanisms sustaining the chronicity of inflammation and psoriatic lesions remain elusive. Combining systematic analyses of several transcriptomic datasets, we delineated gene signatures across human psoriatic skin, identifying S100A9 as one of the most up-regulated genes, which was confirmed in lesioned skin from patients with psoriasis and preclinical psoriasiform skin inflammation models. Genetic ablation or pharmacologic inhibition of S100A9 alleviated Aldara-induced skin inflammation. By single-cell mapping of human psoriatic skin and bone marrow chimeric mice experiments, we identified keratinocytes as the major source of S100A9. Mechanistically, S100A9 induced IL-23 production by dendritic cells, driving the IL-23/type 3 immunity in psoriasiform skin inflammation. In addition, the cutaneous IL-23/IL-17 axis induced epidermal S100A9 expression in human and experimental psoriasis. Thus, we showed an autoregulatory circuit between keratinocyte-derived S100A9 and IL-23/type 3 immunity during psoriasiform inflammation, identifying a crucial function of S100A9 in the chronification of psoriasis.
Collapse
Affiliation(s)
- Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flávio Protásio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Diógenes Lima
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Douglas da Silva Prado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Stefanie Schärli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gabriel Publio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos Hiroji Hiroki
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Paulo Henrique Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - André Saraiva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thainá Norbiato
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Leonardo Lima
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Department of Cell Biology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Cacilda da Silva Souza
- Department of Internal Medicine, Dermatology Division, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
18
|
Rahmat-Zaie R, Amini J, Haddadi M, Beyer C, Sanadgol N, Zendedel A. TNF-α/STAT1/CXCL10 mutual inflammatory axis that contributes to the pathogenesis of experimental models of multiple sclerosis: A promising signaling pathway for targeted therapies. Cytokine 2023; 168:156235. [PMID: 37267677 DOI: 10.1016/j.cyto.2023.156235] [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: 12/24/2022] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Identifying mutual neuroinflammatory axis in different experimental models of multiple sclerosis (MS) is essential to evaluate the de- and re-myelination processes and improve therapeutic interventions' reproducibility. METHODS The expression profile data set of EAE (GSE47900) and cuprizone (GSE100663) models were downloaded from the Gene Expression Omnibus database. The R package and GEO2R software processed these raw chip data. Gene Ontology (GO) functional analysis, KEGG pathway analysis, and protein-protein interaction network analysis were performed to investigate interactions between common differentially expressed genes (DEGs) in all models. Finally, the ELISA method assessed the protein level of highlighted mutual cytokines in serum. RESULTS Our data introduced 59 upregulated [CXCL10, CCL12, and GBP6 as most important] and 17 downregulated [Serpinb1a, Prr18, and Ugt8a as most important] mutual genes. The signal transducer and activator of transcription 1 (STAT1) and CXCL10 were the most crucial hub proteins among mutual upregulated genes. These mutual genes were found to be mainly involved in the TNF-α, TLRs, and complement cascade signaling, and animal models shared 26 mutual genes with MS individuals. Finally, significant upregulation of serum level of TNF-α/IL-1β/CXCL10 cytokines was confirmed in all models in a relatively similar pattern. CONCLUSION For the first time, our study revealed the common neuroinflammatory pathway in animal models of MS and introduced candidate hub genes for better evaluating the preclinical efficacy of pharmacological interventions and designing prospective targeted therapies.
Collapse
Affiliation(s)
- Roya Rahmat-Zaie
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Javad Amini
- Department of Medical Biotechnology and Molecular Science, North Khorasan University of Medical Science, Bojnurd, Iran
| | - Mohammad Haddadi
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran; Institute of Neuroanatomy, RWTH University Hospital Aachen, 52074 Aachen, Germany.
| | - Adib Zendedel
- Institute of Anatomy, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| |
Collapse
|
19
|
Wang YH, Chen YJ, Lai YH, Wang MC, Chen YY, Wu YY, Yang YR, Tsou HY, Li CP, Hsu CC, Huang CE, Chen CC. Mutation-Driven S100A8 Overexpression Confers Aberrant Phenotypes in Type 1 CALR-Mutated MPN. Int J Mol Sci 2023; 24:8747. [PMID: 37240094 PMCID: PMC10217897 DOI: 10.3390/ijms24108747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Numerous pathogenic CALR exon 9 mutations have been identified in myeloproliferative neoplasms (MPN), with type 1 (52bp deletion; CALRDEL) and type 2 (5bp insertion; CALRINS) being the most prevalent. Despite the universal pathobiology of MPN driven by various CALR mutants, it is unclear why different CALR mutations result in diverse clinical phenotypes. Through RNA sequencing followed by validation at the protein and mRNA levels, we found that S100A8 was specifically enriched in CALRDEL but not in CALRINS MPN-model cells. The expression of S100a8 could be regulated by STAT3 based on luciferase reporter assay complemented with inhibitor treatment. Pyrosequencing demonstrated relative hypomethylation in two CpG sites within the potential pSTAT3-targeting S100a8 promoter region in CALRDEL cells as compared to CALRINS cells, suggesting that distinct epigenetic alteration could factor into the divergent S100A8 levels in these cells. The functional analysis confirmed that S100A8 non-redundantly contributed to accelerated cellular proliferation and reduced apoptosis in CALRDEL cells. Clinical validation showed significantly enhanced S100A8 expression in CALRDEL-mutated MPN patients compared to CALRINS-mutated cases, and thrombocytosis was less prominent in those with S100A8 upregulation. This study provides indispensable insights into how different CALR mutations discrepantly drive the expression of specific genes that contributes to unique phenotypes in MPN.
Collapse
Affiliation(s)
- Ying-Hsuan Wang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Ying-Ju Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yi-Hua Lai
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Ming-Chung Wang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yi-Yang Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yu-Ying Wu
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yao-Ren Yang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Hsing-Yi Tsou
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Chian-Pei Li
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Chia-Chen Hsu
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Cih-En Huang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chih-Cheng Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
20
|
Zhao B, Yu J, Luo Y, Xie M, Qu C, Shi Q, Wang X, Zhao X, Kong L, Zhao Y, Guo Y. Deficiency of S100 calcium binding protein A9 attenuates vascular dysfunction in aged mice. Redox Biol 2023; 63:102721. [PMID: 37163872 DOI: 10.1016/j.redox.2023.102721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND S100 calcium-binding protein A9 (S100A9) is a danger-associated molecular pattern molecule that mediates the inflammatory response. Inflammation is essential in aging-related cardiovascular diseases. However, less is known regarding the role of S100A9 in vascular aging. METHODS S100A9 null mice were used to investigate the role of S100A9 in aging-related pathologies. Artery rings were used to measure the functional characteristics of vascular with a pressurized myograph. Telomere length, Sirtuin activity, oxidative stress, and endothelial nitric oxide synthetase (eNOS) activity were used to elevate vascular senescence. Intraperitoneal glucose tolerance (IPGTT) and insulin sensitivity test (IST) were employed to investigate the effects of S100A9 on insulin resistance. Inflammation response was reflected by the concentration of inflammatory cytokines. The Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE) inhibitors were used to identify the downstream molecular mechanisms of S100A9 in aging-induced senescence in endothelial cells. RESULTS S100A9 expression in vascular increased with aging in mice and humans. Deficiency of S100A9 alleviated vascular senescence in aged mice, as evidenced by increased telomere length, Sirtuin activity, and eNOS activity. Meanwhile, S100A9 knockout improved endothelium-dependent vasodilatation and endothelial continuity in aged mice. Moreover, the increased insulin resistance, oxidative stress, and inflammation were mitigated by S100A9 deletion in aged mice. In vitro, S100A9 induced senescence in endothelial cells, and that effect was blunted by TLR4 but not RAGE inhibitors. CONCLUSION The present study suggested that S100A9 may contribute to aging-related pathologies and endothelial dysfunction via the TLR4 pathway. Therefore, targeting S100A9/TLR4 signaling pathway may represent a crucial therapeutic strategy to prevent age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Boying Zhao
- Vascular Surgery Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China
| | - Jiang Yu
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuan Luo
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China
| | - Can Qu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qiong Shi
- The Department of Laboratory Medicine, M.O.E. Key Laboratory of Laboratory Medical Diagnostics, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xingji Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400010, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, 400010, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, 400010, China
| | - Yu Zhao
- Vascular Surgery Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
21
|
Chen Y, Ouyang Y, Li Z, Wang X, Ma J. S100A8 and S100A9 in Cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188891. [PMID: 37001615 DOI: 10.1016/j.bbcan.2023.188891] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/31/2023]
Abstract
S100A8 and S100A9 are Ca2+ binding proteins that belong to the S100 family. Primarily expressed in neutrophils and monocytes, S100A8 and S100A9 play critical roles in modulating various inflammatory responses and inflammation-associated diseases. Forming a common heterodimer structure S100A8/A9, S100A8 and S100A9 are widely reported to participate in multiple signaling pathways in tumor cells. Meanwhile, S100A8/A9, S100A8, and S100A9, mainly as promoters, contribute to tumor development, growth and metastasis by interfering with tumor metabolism and the microenvironment. In recent years, the potential of S100A8/A9, S100A9, and S100A8 as tumor diagnostic or prognostic biomarkers has also been demonstrated. In addition, an increasing number of potential therapies targeting S100A8/A9 and related signaling pathways have emerged. In this review, we will first expound on the characteristics of S100A8/A9, S100A9, and S100A8 in-depth, focus on their interactions with tumor cells and microenvironments, and then discuss their clinical applications as biomarkers and therapeutic targets. We also highlight current limitations and look into the future of S100A8/A9 targeted anti-cancer therapy.
Collapse
|
22
|
Bai B, Xu Y, Chen H. Pathogenic roles of neutrophil-derived alarmins (S100A8/A9) in heart failure: From molecular mechanisms to therapeutic insights. Br J Pharmacol 2023; 180:573-588. [PMID: 36464854 DOI: 10.1111/bph.15998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
An excessive neutrophil count is recognized as a valuable predictor of inflammation and is associated with a higher risk of adverse cardiac events in patients with heart failure. Our understanding of the effectors used by neutrophils to inflict proinflammatory actions needs to be advanced. Recently, emerging evidence has demonstrated a causative role of neutrophil-derived alarmins (i.e. S100A8/A9) in aggravating cardiac injuries by induction of inflammation. In parallel with the neutrophil count, high circulating levels of S100A8/A9 proteins powerfully predict mortality in patients with heart failure. As such, a deeper understanding of the biological functions of neutrophil-derived S100A8/A9 proteins would offer novel therapeutic insights. Here, the basic biology of S100A8/A9 proteins and their pleiotropic roles in cardiovascular diseases are discussed, focusing on heart failure. We also consider the evidence that therapeutic targeting of S100A8/A9 proteins by the humanized vaccine, antibodies or inhibitors is able to town down inflammatory injuries.
Collapse
Affiliation(s)
- Bo Bai
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.,Department of Cardiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Yun Xu
- Department of Cardiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Haibo Chen
- Department of Cardiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| |
Collapse
|
23
|
Mascarenhas J, Gleitz HFE, Chifotides HT, Harrison CN, Verstovsek S, Vannucchi AM, Rampal RK, Kiladjian JJ, Vainchenker W, Hoffman R, Schneider RK, List AF. Biological drivers of clinical phenotype in myelofibrosis. Leukemia 2023; 37:255-264. [PMID: 36434065 PMCID: PMC9898039 DOI: 10.1038/s41375-022-01767-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Abstract
Myelofibrosis (MF) is a myeloproliferative disorder that exhibits considerable biological and clinical heterogeneity. At the two ends of the disease spectrum are the myelodepletive or cytopenic phenotype and the myeloproliferative phenotype. The cytopenic phenotype has a high prevalence in primary MF (PMF) and is characterized by low blood counts. The myeloproliferative phenotype is typically associated with secondary MF (SMF), mild anemia, minimal need for transfusion support, and normal to mild thrombocytopenia. Differences in somatic driver mutations and allelic burden, as well as the acquisition of non-driver mutations further influences these phenotypic differences, prognosis, and response to therapies such as JAK2 inhibitors. The outcome of patients with the cytopenic phenotype are comparatively worse and frequently pose a challenge to treat given the inherent exacerbation of cytopenias. Recent data indicate that an innate immune deregulated state that hinges on the myddosome-IRAK-NFκB axis favors the cytopenic myelofibrosis phenotype and offers opportunity for novel treatment approaches. We will review the biological and clinical features of the MF disease spectrum and associated treatment considerations.
Collapse
Affiliation(s)
- John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hélène F E Gleitz
- Department of Developmental Biology, Erasmus Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Helen T Chifotides
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Srdan Verstovsek
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Raajit K Rampal
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebekka K Schneider
- Department of Developmental Biology, Erasmus Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Institute of Cell and Tumor Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | | |
Collapse
|
24
|
Chen J, Huang Y, Wang B, Lu Y, Jian J, Tang J, Cai J. Characterization of S100A12 from nile tilapia (Oreochromis niloticus) and its roles on inflammatory responses. FISH & SHELLFISH IMMUNOLOGY 2022; 131:401-407. [PMID: 36243273 DOI: 10.1016/j.fsi.2022.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/21/2022] [Accepted: 10/06/2022] [Indexed: 06/16/2023]
Abstract
S100A12 is a member of S100 proteins family that induces pro-inflammatory response via ligating with the receptor for advanced glycation end products (RAGE) and subsequent activation of intracellular signal transduction pathways. But information about fish S100A12 remain largely unclear. In this study, the S100A12 homolog (On-S100A12) was identified from Nile tilapia (Oreochromis niloticus). On-S100A12 was mainly expressed in liver and intestine. After Streptococcus agalactiae infection in vivo, S100A12 significantly increased in brain, intestine, liver and head kidney, suggesting S100A12 might played roles in immune response. The further in vitro experiments found that recombinant protein of S100A12 (rOn-S100A12) upregulated the expression of IL1-β, TLR2, TNF-α and inhibited the expression of IL-10, indicating On-S100A12 promoted inflammatory response and activation of M1 macrophages. The present data lay a foundation to further explore the roles of fish S100 during immune defense and will also be beneficial for better understanding of fish immune-regulatory network.
Collapse
Affiliation(s)
- Junxi Chen
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bei Wang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jufen Tang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Jia Cai
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088, PR China; Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), 524002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangxi Key Lab for Marine Natural Products and Combinational Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Centre, Guangxi Academy of Sciences, Nanning, 530007, PR China.
| |
Collapse
|
25
|
Immanuel T, Li J, Green TN, Bogdanova A, Kalev-Zylinska ML. Deregulated calcium signaling in blood cancer: Underlying mechanisms and therapeutic potential. Front Oncol 2022; 12:1010506. [PMID: 36330491 PMCID: PMC9623116 DOI: 10.3389/fonc.2022.1010506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Intracellular calcium signaling regulates diverse physiological and pathological processes. In solid tumors, changes to calcium channels and effectors via mutations or changes in expression affect all cancer hallmarks. Such changes often disrupt transport of calcium ions (Ca2+) in the endoplasmic reticulum (ER) or mitochondria, impacting apoptosis. Evidence rapidly accumulates that this is similar in blood cancer. Principles of intracellular Ca2+ signaling are outlined in the introduction. We describe different Ca2+-toolkit components and summarize the unique relationship between extracellular Ca2+ in the endosteal niche and hematopoietic stem cells. The foundational data on Ca2+ homeostasis in red blood cells is discussed, with the demonstration of changes in red blood cell disorders. This leads to the role of Ca2+ in neoplastic erythropoiesis. Then we expand onto the neoplastic impact of deregulated plasma membrane Ca2+ channels, ER Ca2+ channels, Ca2+ pumps and exchangers, as well as Ca2+ sensor and effector proteins across all types of hematologic neoplasms. This includes an overview of genetic variants in the Ca2+-toolkit encoding genes in lymphoid and myeloid cancers as recorded in publically available cancer databases. The data we compiled demonstrate that multiple Ca2+ homeostatic mechanisms and Ca2+ responsive pathways are altered in hematologic cancers. Some of these alterations may have genetic basis but this requires further investigation. Most changes in the Ca2+-toolkit do not appear to define/associate with specific disease entities but may influence disease grade, prognosis, treatment response, and certain complications. Further elucidation of the underlying mechanisms may lead to novel treatments, with the aim to tailor drugs to different patterns of deregulation. To our knowledge this is the first review of its type in the published literature. We hope that the evidence we compiled increases awareness of the calcium signaling deregulation in hematologic neoplasms and triggers more clinical studies to help advance this field.
Collapse
Affiliation(s)
- Tracey Immanuel
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jixia Li
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan City, China
| | - Taryn N. Green
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Haematology Laboratory, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
26
|
Wang J, Wang X, Zhang C, Zhou X. Microplastics induce immune suppression via S100A8 downregulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113905. [PMID: 35868177 DOI: 10.1016/j.ecoenv.2022.113905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/05/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
Microplastic (MP) pollution has been largely reported in the daily consumption of water and food, however, the toxicities of MPs to human beings remain largely uncovered. We found that MPs in drinking water significantly impaired mouse immune function by reducing spleen weight, CD8+ T cell amount and raising CD4+ to CD8+ T cell ratio. We performed proteomics and phosphoproteomics by LC-MS/MS and found MPs significantly induced 130 and 57 proteins upregulated in proteome and phosphoproteome, and 191 and 37 proteins downregulated in proteome and phosphoproteome, separately. Bioinformatic analysis show that asthma, mineral absorption, and the IL-17 signaling pathway were significantly enriched and may be involved in MP-induced spleen damage and immune suppression. We verified the top 3 differentially expressed proteins and phosphoproteins by western blot, and we further showed that S100A8 was significantly downregulated by MPs via histochemistry staining. Our results revealed that MPs can induce spleen damage and immune suppression by reducing S100A8 expression, suggesting an underestimated influence and mechanism of MPs on the mammalian immune system.
Collapse
Affiliation(s)
- Juan Wang
- Henan Institute for Food and Drug Control, Zhengzhou, Henan 450000, China
| | - Xiaojuan Wang
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Conghui Zhang
- Department of clinical laboratory medicine, Xiangya Medical School of Central South University, Changsha, Hunan 410013, China; Department of clinical laboratory medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xiao Zhou
- Department of clinical laboratory medicine, Xiangya Medical School of Central South University, Changsha, Hunan 410013, China; Department of clinical laboratory medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
27
|
Venugopal S, Mascarenhas J. The odyssey of pacritinib in myelofibrosis. Blood Adv 2022; 6:4905-4913. [PMID: 35622972 PMCID: PMC9631669 DOI: 10.1182/bloodadvances.2022007524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022] Open
Abstract
Myelofibrosis (MF) can present with symptomatic splenomegaly and/or cytopenias including thrombocytopenia. Disease-related thrombocytopenia is a poor prognostic factor with a median overall survival of less than 2 years. Currently approved JAK1/2 inhibitors have not been evaluated in patients with platelets ≤ 50 × 109/L and in fact could potentiate thrombocytopenia because of their combined JAK1/2 inhibitory activity. Pacritinib (PAC), a selective JAK2, fms-like tyrosine kinase 3, interleukin-1 receptor-associated kinase 1 multikinase inhibitor was developed to meet this unmet need. PAC was evaluated in 2 randomized phase 3 trials in the frontline setting (PERSIST-1, PAC 400 mg daily vs best available therapy) and second-line setting in patients with MF with platelets ≤ 100 × 109/L (PERSIST-2, PAC 400 mg daily or 200 mg twice daily vs best available therapy). PERSIST-1 met its primary end point; however, the development of PAC hit a brief pause because of a US Food and Drug Administration-mandated clinical hold for excess of bleeding and cardiac events in the PAC 400 mg daily arm in the PERSIST-1 study. Although the PERSIST-2 study was terminated abruptly because of this clinical hold, it met its splenic response end point and demonstrated a trend toward symptom improvement. Subsequent, diligent review of the PERSIST-1 and PERSIST-2 studies did not confirm an excess of severe bleeding or cardiac events on the PAC arm. Additionally, the dose finding PAC203 study endorsed the safety and efficacy of 200 mg twice daily, leading to the approval of PAC for the treatment of patients with MF with platelets ≤ 50 × 109/L.
Collapse
Affiliation(s)
- Sangeetha Venugopal
- Department of Leukemia, MD Anderson Cancer Center, University of Texas, Houston, TX; and
| | - John Mascarenhas
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
28
|
Regulation of S100As Expression by Inflammatory Cytokines in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:ijms23136952. [PMID: 35805957 PMCID: PMC9267105 DOI: 10.3390/ijms23136952] [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: 05/16/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The calcium-binding proteins S100A4, S100A8, and S100A9 are upregulated in chronic lymphocytic leukemia (CLL), while the S100A9 promotes NF-κB activity during disease progression. The S100-protein family has been involved in several malignancies as mediators of inflammation and proliferation. The hypothesis of our study is that S100A proteins are mediators in signaling pathways associated with inflammation-induced proliferation, such as NF-κB, PI3K/AKT, and JAK/STAT. The mononuclear cells (MNCs) of CLL were treated with proinflammatory IL-6, anti-inflammatory IL-10 cytokines, inhibitors of JAK1/2, NF-κB, and PI3K signaling pathways, to evaluate S100A4, S100A8, S100A9, and S100A12 expression as well as NF-κB activation by qRT-PCR, immunocytochemistry, and immunoblotting. The quantity of S100A4, S100A8, and S100A9 positive cells (p < 0.05) and their protein expression (p < 0.01) were significantly decreased in MNCs of CLL patients compared to healthy controls. The S100A levels were generally increased in CD19+ cells compared to MNCs of CLL. The S100A4 gene expression was significantly stimulated (p < 0.05) by the inhibition of the PI3K/AKT signaling pathway in MNCs. IL-6 stimulated S100A4 and S100A8 protein expression, prevented by the NF-κB and JAK1/2 inhibitors. In contrast, IL-10 reduced S100A8, S100A9, and S100A12 protein expressions in MNCs of CLL. Moreover, IL-10 inhibited activation of NF-κB signaling (4-fold, p < 0.05). In conclusion, inflammation stimulated the S100A protein expression mediated via the proliferation-related signaling and balanced by the cytokines in CLL.
Collapse
|
29
|
Wang JC, Sun L. PD-1/PD-L1, MDSC Pathways, and Checkpoint Inhibitor Therapy in Ph(-) Myeloproliferative Neoplasm: A Review. Int J Mol Sci 2022; 23:5837. [PMID: 35628647 PMCID: PMC9143160 DOI: 10.3390/ijms23105837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
There has been significant progress in immune checkpoint inhibitor (CPI) therapy in many solid tumor types. However, only a single failed study has been published in treating Ph(-) myeloproliferative neoplasm (MPN). To make progress in CPI studies on this disease, herein, we review and summarize the mechanisms of activation of the PD-L1 promoter, which are as follows: (a) the extrinsic mechanism, which is activated by interferon gamma (IFN γ) by tumor infiltration lymphocytes (TIL) and NK cells; (b) the intrinsic mechanism of EGFR or PTEN loss resulting in the activation of the MAPK and AKT pathways and then stat 1 and 3 activation; and (c) 9p24 amplicon amplification, resulting in PD-L1 and Jak2 activation. We also review the literature and postulate that many of the failures of CPI therapy in MPN are likely due to excessive MDSC activities. We list all of the anti-MDSC agents, especially those with ruxolitinib, IMID compounds, and BTK inhibitors, which may be combined with CPI therapy in the future as part of clinical trials applying CPI therapy to Ph(-) MPN.
Collapse
Affiliation(s)
- Jen-Chin Wang
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA;
| | | |
Collapse
|
30
|
Sun F, Zhang H, Huang T, Shi J, Wei T, Wang Y. S100A9 blockade improves the functional recovery after spinal cord injury via mediating neutrophil infiltration. Exp Ther Med 2022; 23:291. [PMID: 35317450 PMCID: PMC8908460 DOI: 10.3892/etm.2022.11220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/27/2022] [Indexed: 11/18/2022] Open
Abstract
Spinal cord injury (SCI) refers to damage to the spinal cord resulting from trauma, disease or degeneration. Controlling the inflammatory process and restoring neural homeostasis is hypothesized to prevent injury aggravation. S100 calcium-binding protein A9 (S100A9) is a pro-inflammatory alarm protein, which is expressed in and released by activated neutrophils. However, whether S100A9 could serve as an effective target for the treatment of SCI has not been reported to date. In the present study, a T10 spinal cord contusion injury model was established in Sprague-Dawley rats. S100A9 expression level was determined in the serum and injured spinal cord tissue via ELISA, reverse transcription-quantitative PCR (RT-qPCR) and western blotting. The S100A9-specific blocker, ABR-238901 (ABR), was administered during the inflammatory phase of SCI, as a form of treatment. Subsequently, the morphological structure, neuronal viability and inflammatory levels of injured spinal cord were observed by histopathology, immunohistochemistry and RT-qPCR. In the obtained results, S100A9 was found to be highly expressed in the injured spinal cord and serum in the first 3 days after SCI. However, at 28 days after surgery, ABR treatment significantly improved motor function, reduced the cavity formation and neutrophil infiltration in the lesion, which was verified via H&E staining and immunohistochemistry for myeloperoxidase. Furthermore, ABR treatment was found to effectively improve the survival and viability of neurons, as shown via Nissl staining and immunofluorescence of the synaptic plasticity markers, microtubule associated protein 2 and neurofilament 200. Moreover, S100A9 blockade effectively upregulated the mRNA expression level of the anti-inflammatory genes, IL-4 and IL-10 and downregulated the mRNA expression level of the pro-inflammatory factors, IL-1β, IL-6 and TNF-α. In addition, S100A9 blockade notably alleviated the apoptosis level of the injured nerve cells. Therefore, the findings of the present study revealed that S100A9 may be a useful target for the treatment of SCI.
Collapse
Affiliation(s)
- Feng Sun
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Haiwei Zhang
- Imaging, General Hospital of Heilongjiang General Administration of Agriculture and Reclamation, Harbin, Heilongjiang 150000, P.R. China
| | - Tianwen Huang
- Department of Orthopedics, General Hospital of Heilongjiang General Administration of Agriculture and Reclamation, Harbin, Heilongjiang 150000, P.R. China
| | - Jianhui Shi
- Department of Orthopaedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150000, P.R. China
| | - Tianli Wei
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Yansong Wang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
31
|
Baumeister J, Maié T, Chatain N, Gan L, Weinbergerova B, de Toledo MAS, Eschweiler J, Maurer A, Mayer J, Kubesova B, Racil Z, Schuppert A, Costa I, Koschmieder S, Brümmendorf TH, Gezer D. Early and late stage MPN patients show distinct gene expression profiles in CD34 + cells. Ann Hematol 2021; 100:2943-2956. [PMID: 34390367 PMCID: PMC8592960 DOI: 10.1007/s00277-021-04615-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022]
Abstract
Myeloproliferative neoplasms (MPN), comprising essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), are hematological disorders of the myeloid lineage characterized by hyperproliferation of mature blood cells. The prediction of the clinical course and progression remains difficult and new therapeutic modalities are required. We conducted a CD34+ gene expression study to identify signatures and potential biomarkers in the different MPN subtypes with the aim to improve treatment and prevent the transformation from the rather benign chronic state to a more malignant aggressive state. We report here on a systematic gene expression analysis (GEA) of CD34+ peripheral blood or bone marrow cells derived from 30 patients with MPN including all subtypes (ET (n = 6), PV (n = 11), PMF (n = 9), secondary MF (SMF; post-ET-/post-PV-MF; n = 4)) and six healthy donors. GEA revealed a variety of differentially regulated genes in the different MPN subtypes vs. controls, with a higher number in PMF/SMF (200/272 genes) than in ET/PV (132/121). PROGENγ analysis revealed significant induction of TNFα/NF-κB signaling (particularly in SMF) and reduction of estrogen signaling (PMF and SMF). Consistently, inflammatory GO terms were enriched in PMF/SMF, whereas RNA splicing–associated biological processes were downregulated in PMF. Differentially regulated genes that might be utilized as diagnostic/prognostic markers were identified, such as AREG, CYBB, DNTT, TIMD4, VCAM1, and S100 family members (S100A4/8/9/10/12). Additionally, 98 genes (including CLEC1B, CMTM5, CXCL8, DACH1, and RADX) were deregulated solely in SMF and may be used to predict progression from early to late stage MPN.
Collapse
Affiliation(s)
- Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Tiago Maié
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany.,Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Lin Gan
- IZKF Genomics Core Facility, RWTH Aachen University Medical School, Aachen, Germany
| | - Barbora Weinbergerova
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Marcelo A S de Toledo
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Jörg Eschweiler
- Department of Orthopedic Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Angela Maurer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Blanka Kubesova
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Zdenek Racil
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Andreas Schuppert
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany.,Joint Research Center for Computational Biomedicine, RWTH Aachen, Aachen, Germany
| | - Ivan Costa
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany.,Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany. .,Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
32
|
Soyfer EM, Fleischman AG. Inflammation in Myeloid Malignancies: From Bench to Bedside. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:160-167. [PMID: 35663100 PMCID: PMC9138438 DOI: 10.36401/jipo-21-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/21/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Myeloid malignancies, stemming from a somatically mutated hematopoietic clone, can cause a wide variety of clinical consequences, including pancytopenia in myelodysplastic syndrome, overproduction of three myeloid lineages in myeloproliferative neoplasm, and the rapid growth of immature hematopoietic cells in acute myeloid leukemia (AML). It is becoming clear that inflammation is a hallmark feature of clonal myeloid conditions, ranging from clonal hematopoiesis of indeterminate potential to AML. Fundamental findings from laboratory research on inflammation in myeloid malignancies has potential implications for diagnosis, prognostication, and treatment in these diseases. In this review, we highlighted some pertinent basic science findings regarding the role of inflammation in myeloid malignancies and speculated how these findings could impact the clinical care of patients.
Collapse
Affiliation(s)
- Eli M Soyfer
- School of Medicine, University of California, Irvine, CA, USA
| | - Angela G Fleischman
- Division of Hematology/Oncology, UC Irvine Health, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, USA
| |
Collapse
|
33
|
Wang T, Du G, Wang D. The S100 protein family in lung cancer. Clin Chim Acta 2021; 520:67-70. [PMID: 34089725 DOI: 10.1016/j.cca.2021.05.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/31/2022]
Abstract
The S100 protein family is involved in the pathogenesis of several malignancies including lung cancer. Recent studies have shown that one member, S100A2, was over-expressed in advanced stage non-small cell lung cancer (NSCLC). Another, S100A6, demonstrated variable expression in different lung cancer subtypes. Research using NSCLC cell lines reported that SIX3 inhibited cell metastasis and proliferation via S100P down-regulation. This review represents an update on S100 proteins in lung cancer from 2017 to 2021 and includes the aforementioned as well as S100A4, S100A7, and S100B. Inconsistencies in mechanisms of action for S100A8/S100A9 are highlighted and a comprehensive evaluation of the most recent evidence for the S100 proteins in lung cancer is presented.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory Medicine, Xi'an People's Hospital (Xi'an No.4 Hospital), Xi'an 710004, China
| | - Ge Du
- Department of Rehabilitation Center for Elderly, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Beijing 100144, China
| | - Dong Wang
- Department of Radiology, Xi'an People's Hospital (Xi'an No.4 Hospital), Xi'an 710004, China.
| |
Collapse
|
34
|
Xiang H, Guo F, Tao X, Zhou Q, Xia S, Deng D, Li L, Shang D. Pancreatic ductal deletion of S100A9 alleviates acute pancreatitis by targeting VNN1-mediated ROS release to inhibit NLRP3 activation. Theranostics 2021; 11:4467-4482. [PMID: 33754072 PMCID: PMC7977474 DOI: 10.7150/thno.54245] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/03/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have proven that the overall pathophysiology of pancreatitis involves not only the pancreatic acinar cells but also duct cells, however, pancreatic duct contribution in acinar cells homeostasis is poorly known and the molecular mechanisms leading to acinar insult and acute pancreatitis (AP) are unclear. Our previous work also showed that S100A9 protein level was notably increased in AP rat pancreas through iTRAQ-based quantitative proteomic analysis. Therefore, we investigated the actions of injured duct cells on acinar cells and the S100A9-related effects and mechanisms underlying AP pathology in the present paper. Methods: In this study, we constructed S100A9 knockout (s100a9-/-) mice and an in vitro coculture system for pancreatic duct cells and acinar cells. Moreover, a variety of small molecular inhibitors of S100A9 were screened from ChemDiv through molecular docking and virtual screening methods. Results: We found that the upregulation of S100A9 induces cell injury and inflammatory response via NLRP3 activation by targeting VNN1-mediated ROS release; and loss of S100A9 decreases AP injury in vitro and in vivo. Moreover, molecular docking and mutant plasmid experiments proved that S100A9 has a direct interaction with VNN1 through the salt bridges formation of Lys57 and Glu92 residues in S100A9 protein. We further found that compounds C42H60N4O6 and C28H29F3N4O5S can significantly improve AP injury in vitro and in vivo through inhibiting S100A9-VNN1 interaction. Conclusions: Our study showed the important regulatory effect of S100A9 on pancreatic duct injury during AP and revealed that inhibition of the S100A9-VNN1 interaction may be a key therapeutic target for this disease.
Collapse
|
35
|
Zhao B, Lu R, Chen J, Xie M, Zhao X, Kong L. S100A9 blockade prevents lipopolysaccharide-induced lung injury via suppressing the NLRP3 pathway. Respir Res 2021; 22:45. [PMID: 33549095 PMCID: PMC7866705 DOI: 10.1186/s12931-021-01641-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Background S100 calcium binding protein A9 (S100A9) is a pro-inflammatory alarmin associated with several inflammation-related diseases. However, the role of S100A9 in lung injury in sepsis has not been fully investigated. Therefore, the present study aimed to determine the role of S100A9 in a lipopolysaccharide (LPS)-induced lung injury murine model and its underlying molecular mechanisms. Methods LPS was utilized to induce sepsis and lung injury in C57BL/6 or NOD-like receptor family pyrin domain containing 3 (NLRP3)−/− mice. To investigate the effects of S100A9 blockade, mice were treated with a specific inhibitor of S100A9. Subsequently, lung injury and inflammation were evaluated by histology and enzyme‑linked immunosorbent assay (ELISA), respectively. Furthermore, western blot analysis and RT-qPCR were carried out to investigate the molecular mechanisms underlying the effects of S100A9. Results S100A9 was upregulated in the lung tissues of LPS-treated mice. However, inhibition of S100A9 alleviated LPS-induced lung injury. Additionally, S100A9 blockade also attenuated the inflammatory responses and apoptosis in the lungs of LPS-challenged mice. Furthermore, the increased expression of NLRP3 was also suppressed by S100A9 blockade, while S100A9 blockade had no effect on NLRP3−/− mice. In vitro, S100A9 downregulation mitigated LPS-induced inflammation. Interestingly, these effects were blunted by NLRP3 overexpression. Conclusion The results of the current study suggested that inhibition of S100A9 could protect against LPS-induced lung injury via inhibiting the NLRP3 pathway. Therefore, S100A9 blockade could be considered as a novel therapeutic strategy for lung injury in sepsis.
Collapse
Affiliation(s)
- Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.,Vascular Surgery Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Renfu Lu
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Jianjun Chen
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Xingji Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.
| |
Collapse
|
36
|
May O, Yatime L, Merle NS, Delguste F, Howsam M, Daugan MV, Paul-Constant C, Billamboz M, Ghinet A, Lancel S, Dimitrov JD, Boulanger E, Roumenina LT, Frimat M. The receptor for advanced glycation end products is a sensor for cell-free heme. FEBS J 2020; 288:3448-3464. [PMID: 33314778 DOI: 10.1111/febs.15667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/06/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023]
Abstract
Heme's interaction with Toll-like receptor 4 (TLR4) does not fully explain the proinflammatory properties of this hemoglobin-derived molecule during intravascular hemolysis. The receptor for advanced glycation end products (RAGE) shares many features with TLR4 such as common ligands and proinflammatory, prothrombotic, and pro-oxidative signaling pathways, prompting us to study its involvement as a heme sensor. Stable RAGE-heme complexes with micromolar affinity were detected as heme-mediated RAGE oligomerization. The heme-binding site was located in the V domain of RAGE. This interaction was Fe3+ -dependent and competitive with carboxymethyllysine, another RAGE ligand. We confirmed a strong basal gene expression of RAGE in mouse lungs. After intraperitoneal heme injection, pulmonary TNF-α, IL1β, and tissue factor gene expression levels increased in WT mice but were significantly lower in their RAGE-/- littermates. This may be related to the lower activation of ERK1/2 and Akt observed in the lungs of heme-treated, RAGE-/- mice. Overall, heme binds to RAGE with micromolar affinity and could promote proinflammatory and prothrombotic signaling in vivo, suggesting that this interaction could be implicated in heme-overload conditions.
Collapse
Affiliation(s)
- Olivia May
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France.,CHU Lille, Nephrology Department, Univ. Lille, France.,UMR_S 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Laure Yatime
- LPHI, UMR 5235, CNRS, INSERM, University of Montpellier, France
| | - Nicolas S Merle
- UMR_S 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | - Florian Delguste
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France
| | - Mike Howsam
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France
| | - Marie V Daugan
- UMR_S 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France
| | | | - Muriel Billamboz
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France.,Yncréa Hauts-de-France, Ecole des Hautes Etudes d'Ingénieur, Health & Environment Department, Team Sustainable Chemistry, Laboratoire de Chimie Durable et Santé, UCLille, France
| | - Alina Ghinet
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France.,Yncréa Hauts-de-France, Ecole des Hautes Etudes d'Ingénieur, Health & Environment Department, Team Sustainable Chemistry, Laboratoire de Chimie Durable et Santé, UCLille, France.,Faculty of Chemistry, 'Alexandru Ioan Cuza' University of Iasi, Romania
| | - Steve Lancel
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France
| | - Jordan D Dimitrov
- UMR_S 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, France
| | - Eric Boulanger
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France
| | - Lubka T Roumenina
- UMR_S 1138, Centre de Recherche des Cordeliers, INSERM, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, France
| | - Marie Frimat
- Inserm, Institut Pasteur de Lille, U1167 - RID-AGE, Univ. Lille, France.,CHU Lille, Nephrology Department, Univ. Lille, France
| |
Collapse
|
37
|
Synergic Crosstalk between Inflammation, Oxidative Stress, and Genomic Alterations in BCR-ABL-Negative Myeloproliferative Neoplasm. Antioxidants (Basel) 2020; 9:antiox9111037. [PMID: 33114087 PMCID: PMC7690801 DOI: 10.3390/antiox9111037] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/06/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) have recently been revealed to be related to chronic inflammation, oxidative stress, and the accumulation of reactive oxygen species. It has been proposed that MPNs represent a human inflammation model for tumor advancement, in which long-lasting inflammation serves as the driving element from early tumor stage (over polycythemia vera) to the later myelofibrotic cancer stage. It has been theorized that the starting event for acquired stem cell alteration may occur after a chronic inflammation stimulus with consequent myelopoietic drive, producing a genetic stem cell insult. When this occurs, the clone itself constantly produces inflammatory components in the bone marrow; these elements further cause clonal expansion. In BCR-ABL1-negative MPNs, the driver mutations include JAK 2, MPL, and CALR. Transcriptomic studies of hematopoietic stem cells from subjects with driver mutations have demonstrated the upregulation of inflammation-related genes capable of provoking the development of an inflammatory state. The possibility of acting on the inflammatory state as a therapeutic approach in MPNs appears promising, in which an intervention operating on the pathways that control the synthesis of cytokines and oxidative stress could be effective in reducing the possibility of leukemic progression and onset of complications.
Collapse
|
38
|
Lee JS, Lee NR, Kashif A, Yang SJ, Nam AR, Song IC, Gong SJ, Hong MH, Kim G, Seok PR, Lee MS, Sung KH, Kim IS. S100A8 and S100A9 Promote Apoptosis of Chronic Eosinophilic Leukemia Cells. Front Immunol 2020; 11:1258. [PMID: 32903598 PMCID: PMC7438788 DOI: 10.3389/fimmu.2020.01258] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
S100A8 and S100A9 function as essential factors in inflammation and also exert antitumor or tumorigenic activity depending on the type of cancer. Chronic eosinophilic leukemia (CEL) is a rare hematological malignancy having elevated levels of eosinophils and characterized by the presence of the FIP1L1-PDGFRA fusion gene. In this study, we examined the pro-apoptotic mechanisms of S100A8 and S100A9 in FIP1L1-PDGFRα+ eosinophilic cells and hypereosinophilic patient cells. S100A8 and S100A9 induce apoptosis of the FIP1L1-PDGFRα+ EoL-1 cells via TLR4. The surface TLR4 expression increased after exposure to S100A8 and S100A9 although total TLR4 expression decreased. S100A8 and S100A9 suppressed the FIP1L1-PDGFRα-mediated signaling pathway by downregulating FIP1L1-PDGFRα mRNA and protein expression and triggered cell apoptosis by regulating caspase 9/3 pathway and Bcl family proteins. S100A8 and S100A9 also induced apoptosis of imatinib-resistant EoL-1 cells (EoL-1-IR). S100A8 and S100A9 blocked tumor progression of xenografted EoL-1 and EoL-1-IR cells in NOD-SCID mice and evoked apoptosis of eosinophils derived from hypereosinophilic syndrome as well as chronic eosinophilic leukemia. These findings may contribute to a progressive understanding of S100A8 and S100A9 in the pathogenic and therapeutic mechanism of hematological malignancy.
Collapse
Affiliation(s)
- Ji-Sook Lee
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan, South Korea
| | - Na Rae Lee
- Department of Biomedical Laboratory Science, Eulji University School of Medicine, Daejeon, South Korea
| | - Ayesha Kashif
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, South Korea
| | - A Reum Nam
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, South Korea.,Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, Daejeon, South Korea
| | - Soo-Jung Gong
- Department of Internal Medicine, Eulji Medical Center, Eulji University School of Medicine, Daejeon, South Korea
| | - Min Hwa Hong
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| | - Geunyeong Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| | - Pu Reum Seok
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Kee-Hyung Sung
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| | - In Sik Kim
- Department of Biomedical Laboratory Science, Eulji University School of Medicine, Daejeon, South Korea.,Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, South Korea
| |
Collapse
|
39
|
Silva CR, Melo BMS, Silva JR, Lopes AH, Pereira JA, Cecilio NT, Berlink J, Souza GG, Lucas G, Vogl T, Cunha FQ, Alves-Filho JC, Cunha TM. S100A9 plays a pivotal role in a mouse model of herpetic neuralgia via TLR4/TNF pathway. Brain Behav Immun 2020; 88:353-362. [PMID: 32243898 DOI: 10.1016/j.bbi.2020.03.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/18/2020] [Accepted: 03/29/2020] [Indexed: 02/01/2023] Open
Abstract
Herpetic neuralgia is a painful condition following herpes zoster disease, which results from Varicella-zoster virus reactivation in the dorsal or trigeminal sensory ganglia. Nevertheless, the pathophysiological mechanisms involved in herpetic neuralgia are not well understood. Recently, we identified, that neuroimmune-glia interactions in the sensory ganglion is a critical mechanism for the development of herpetic neuralgia. Here, we investigate the contribution of S100A9, a well-known pro-inflammatory molecule produced by myeloid cells, for the development of herpetic neuralgia using a murine model of HSV-1 infection. We found that cutaneous HSV-1 infection results in an increase of S100A9 expression in the Dorsal Root Ganglia (DRGs). Infiltrating neutrophils into the DRGs were the main source of S100A9 post HSV-1 infection. Functionally, genetic or pharmacological inhibition of S100A9 impairs the development of HSV-1 infection-induced mechanical pain hypersensitivity. Finally, we found that the pronociceptive role of S100A9 in herpetic neuralgia depends on the TLR4/TNF pathway. These results unraveled previously unknown mechanisms involved in the pathophysiology of herpetic neuralgia and indicate that S100A9 might be an important target for novel therapies aiming acute herpetic neuralgia.
Collapse
Affiliation(s)
- Cássia R Silva
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Graduated Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlândia, 38408-100 Uberlândia MG, Brazil
| | - Bruno M S Melo
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jaqueline R Silva
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Alexandre H Lopes
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Janaina A Pereira
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Nerry T Cecilio
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jonilson Berlink
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Bahia, Brazil
| | - Giovani G Souza
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Guilherme Lucas
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thomas Vogl
- Institute of Immunology, University of Münster, D-48149 Münster, Germany
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José C Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
40
|
Carvalho A, Lu J, Francis JD, Moore RE, Haley KP, Doster RS, Townsend SD, Johnson JG, Damo SM, Gaddy JA. S100A12 in Digestive Diseases and Health: A Scoping Review. Gastroenterol Res Pract 2020; 2020:2868373. [PMID: 32184815 PMCID: PMC7061133 DOI: 10.1155/2020/2868373] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/05/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Calgranulin proteins are an important class of molecules involved in innate immunity. These members of the S100 class of the EF-hand family of calcium-binding proteins have numerous cellular and antimicrobial functions. One protein in particular, S100A12 (also called EN-RAGE or calgranulin C), is highly abundant in neutrophils during acute inflammation and has been implicated in immune regulation. Structure-function analyses reveal that S100A12 has the capacity to bind calcium, zinc, and copper, processes that contribute to nutritional immunity against invading microbial pathogens. S100A12 is a ligand for the receptor for advanced glycation end products (RAGE), toll-like receptor 4 (TLR4), and CD36, which promote cellular and immunological pathways to alter inflammation. We conducted a scoping review of the existing literature to define what is known about the association of S100A12 with digestive disease and health. Results suggest that S100A12 is implicated in gastroenteritis, necrotizing enterocolitis, gastritis, gastric cancer, Crohn's disease, irritable bowel syndrome, inflammatory bowel disease, and digestive tract cancers. Together, these results reveal S100A12 is an important molecule broadly associated with the pathogenesis of digestive diseases.
Collapse
Affiliation(s)
- Alexandre Carvalho
- Internal Medicine Program, St. Joseph Mercy Hospital, Ann Arbor, Michigan, USA
| | - Jacky Lu
- Department of Pathology, Microbiology, And Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jamisha D. Francis
- Department of Pathology, Microbiology, And Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Rebecca E. Moore
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Kathryn P. Haley
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, USA
| | - Ryan S. Doster
- Department of Pathology, Microbiology, And Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Jeremiah G. Johnson
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Steven M. Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, USA
- Departments of Biochemistry and Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Department of Structural Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology, And Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, Tennessee, USA
| |
Collapse
|
41
|
Diklić M, Mitrović-Ajtić O, Subotički T, Djikić D, Kovačić M, Bjelica S, Beleslin-Čokić B, Tošić M, Leković D, Gotić M, Santibanez JF, Čokić VP. IL6 inhibition of inflammatory S100A8/9 proteins is NF-κB mediated in essential thrombocythemia. Cell Biochem Funct 2019; 38:362-372. [PMID: 31885098 DOI: 10.1002/cbf.3482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Accepted: 12/15/2019] [Indexed: 11/07/2022]
Abstract
This study has been performed to determine the mechanism of activation of the myeloid related S100A proteins by inflammatory cytokines in myeloproliferative neoplasm (MPN). Besides microarray analysis of MPN-derived CD34+ cells, we analysed the pro-inflammatory IL6 and anti-inflammatory IL10 dependence of NF-κB, PI3K-AKT, and JAK-STAT signalling during induction of S100A proteins in mononuclear cells of MPN, by immunoblotting and flow cytometry. We observed the reduced gene expression linked to NF-κB and inflammation signalling in MPN-derived CD34+ cells. Both IL6 and IL10 reduced S100A8 and 100A9 protein levels mediated via NF-κB and PI3K signalling, respectively, in mononuclear cells of essential thrombocythemia (ET). We also determined the increased percentage of S100A8 and S100A9 positive granulocytes in ET and primary myelofibrosis, upgraded by the JAK2V617F mutant allele burden. S100A8/9 heterodimer induced JAK1/2-dependent mitotic arrest of the ET-derived granulocytes. SIGNIFICANCE OF THE STUDY: We demonstrated that inflammation reduced the myeloid related S100A8/9 proteins by negative feedback mechanism in ET. S100A8/9 can be a diagnostic marker of inflammation in MPN, supported by the concomitant NF-κB and JAK1/2 signalling inhibition in regulation of myeloproliferation and therapy of MPN.
Collapse
Affiliation(s)
- Miloš Diklić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Olivera Mitrović-Ajtić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Tijana Subotički
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Dragoslava Djikić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Marijana Kovačić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Bojana Beleslin-Čokić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Genetic Laboratory, Clinical Center of Serbia, Belgrade, Serbia
| | - Milica Tošić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Danijela Leković
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Mirjana Gotić
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Vladan P Čokić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| |
Collapse
|
42
|
Zhu J, Wang Z, Chen F. Association of Key Genes and Pathways with Atopic Dermatitis by Bioinformatics Analysis. Med Sci Monit 2019; 25:4353-4361. [PMID: 31184315 PMCID: PMC6582687 DOI: 10.12659/msm.916525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Atopic dermatitis is a chronic inflammatory disease of the skin. It has a high prevalence worldwide and affected persons are prone to recurrent attacks, seriously affecting the physical and mental of patients. The exact etiology of the disease is still unclear. Material/Methods There are 7 datasets on atopic dermatitis in the Gene Expression Omnibus database, including 142 lesional and 134 non-lesional skin biopsy samples. Differential analysis was performed after datasets were integrated by robust multi-array average method. Functional modules of GSE99802 were explored by weighted gene co-expression network analysis. The 4 most important modules were enriched into the pathways by Metascape. Results Significantly differentially expressed genes included 41 upregulated and 10 downregulated genes. The following 5 of the most important upregulated genes had the strongest association with atopic dermatitis. SERPINB3&4 promote inflammation and impaired skin barrier function in the early stage of atopic dermatitis. S100A9 aggravates the inflammatory response by inducing the activation of toll-like receptor 4, neutrophil chemotaxis, neutrophilic inflammation, and the amplification of interleukin-8. MMP1 is the key protease of skin collagen degradation, keeping the extracellular matrix in dynamic balance. MMP12 induces the aggregation of various inflammatory cells into inflammatory tissue. The enriched pathways of each module mainly include Cellular responses to external stimuli, Metabolism of RNA and Translation, and Infectious disease. Conclusions The associated pathways and genes not only help us understand the molecular mechanism of the disease, but also provide research directions or targets for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|