1
|
Porreca S, Mennella A, Frasca L. The Role of CXCL4 in Systemic Sclerosis: DAMP, Auto-Antigen and Biomarker. Int J Mol Sci 2025; 26:2421. [PMID: 40141068 PMCID: PMC11942444 DOI: 10.3390/ijms26062421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by specific autoantibodies, vasculopathy and fibrosis of the skin and internal organs. In SSc, chronic activation of the immune system is largely sustained by endogenous inflammatory mediators that act as damage-associated molecular patterns (DAMPs), which activate Toll-like receptors (TLRs). Major autoantigens are nucleic acids or molecules that are able to bind nucleic acids. It is important to identify solid and predictive biomarkers of both disease activity and disease subtype. CXCL4 has been regarded as a new biomarker for early SSc in recent years, and here, we discuss its modulation over the course of a disease and after pharmacological interventions. Moreover, we provide evidence that CXCL4, in addition to being a biomarker of SSc subtypes and a prognostic marker of disease severity, has a dual pathogenic role in SSc: on the one hand, in complex with self-nucleic acids, CXCL4 acts as a DAMP for IFN-I and pro-inflammatory cytokines' release by innate immune cells (such as dendritic cells); on the other hand, CXCL4 is a target of both antibodies and T cells, functioning as an autoantigen. CXCL4 is certainly an interesting molecule in inflammation and autoimmunity, not only in SSc, and it may also be considered as a therapy target.
Collapse
Affiliation(s)
| | | | - Loredana Frasca
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.P.); (A.M.)
| |
Collapse
|
2
|
Li Y, Chen S, Liu S. Immune Effect of Co-Culture of Dendritic Cells and Cytokine-Induced Killer Cells on Prostate Cancer Cells. Cell Biochem Biophys 2024:10.1007/s12013-024-01569-2. [PMID: 39448420 DOI: 10.1007/s12013-024-01569-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/26/2024]
Abstract
It was to explore the immune outcome of co-culture of dendritic cells (DC) and cytokine-induced killer cells (CIK) on prostate cancer (PCa) cells. Peripheral blood mononuclear cells (PBMCs) were extracted from healthy blood donors. DC and CIK cells were induced and co-cultured. The proliferation and phenotypic changes of DC, CIK, and DC-CIK cells/groups were observed. Model rats were constructed by injecting PC3 PCa cells into the abdominal cavity. The successful 50 cases were divided into a negative control group, a chemotherapy group, a DC group, a CIK group, and a DC-CIK treatment group (each consisting of 10 rats) to observe tumor progression. The secreted concentrations of interleukin-12 (IL-12) ((103.67 ± 2.77) pg/mL) and interferon-γ (IFN-γ) ((730.09 ± 23.52) pg/mL) were higher in DC-CIK group as against DC and CIK groups; the proliferation of CIK was higher in DC-CIK group as against CIK within 12 to 20 days of culture. The positive rate (PR) of CD3+/ CD56+ and CD8+ was higher and that of CD45RA+ was lower in DC-CIK group as against CIK.The killing rate of the DC-CIK group was higher than that of the DC and CIK groups at a target effect ratio of 10:1/20:1/50:1 (P < 0.05). After the treatment, the body weight of rats in the chemotherapy group, DC group, and CIK group was significantly reduced (P < 0.05), while no significant changes were observed in the negative control group and DC-CIK group (P > 0.05). After 25 days of treatment, the tumor size in the DC-CIK group was significantly smaller compared to the negative control group, chemotherapy group, DC group, and CIK group; the necrotic area of the tumor tissue in the DC-CIK group was also significantly larger than that in the negative control group, chemotherapy group, DC group, and CIK group (P < 0.05). Co-culture of DC and CIK is excellent in enhancing the proliferation of CIK cells, increasing the secretion of IL-12 and IFN-γ, and enhancing the activity of immune cells and anti-tumor ability, showing its potential in anti-PCa tumor immunotherapy.
Collapse
Affiliation(s)
- Yaojun Li
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China
| | - Shanmiao Chen
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China
| | - Shoulei Liu
- Department of Urology Surgery, Tongxiang First People's Hospital, Tongxiang, 314500, Zhejiang, China.
| |
Collapse
|
3
|
Wei J, Peng MY, Wang SN, Lu HX. CXCL4:NLRP3-mediated pyroptosis product that regulates cardiac fibrosis. Int Immunopharmacol 2024; 133:112096. [PMID: 38657496 DOI: 10.1016/j.intimp.2024.112096] [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: 01/17/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Severe myocarditis is often accompanied by cardiac fibrosis, but the underlying mechanism has not been fully elucidated. NOD-like receptor protein 3 (NLRP3) inflammation is involved in the development of myocarditis and is closely related to the form of cell death. Inhibiting pyroptosis mediated by NLRP3 inflammasome can reduce cardiac fibrosis, although its exact mechanism remains unknown. In this study, we induced Viral myocarditis (VMC) via infection of CVB3 to explore the relationship between pyroptosis and fibrosis. Our results showed that intraperitoneal injection of an NLRP3 inhibitor MCC950 or use of NLRP3-/- mice inhibited cardiac pyroptosis mediated by NLRP3 inflammasome in VMC. CXCL4 is a chemokine that has been reported to have pro-inflammatory and pro-fibrotic functions. In VMC, we further found that pyroptosis of Mouse myocardial fibroblasts (MCF) promoted the secretion of CXCL4 by activating Wnt/β-Catenin signaling. Subsequently, the transcriptome sequencing data showed that CXCL4 could promote cardiac fibrosis by activating PI3K/AKT pathway. In summary, infection of CVB3 induced host oxidative stress to further activate the NLRP3 inflammasome and ultimately lead to heart pyroptosis, in which MCF secreted CXCL4 by activating Wnt/β-Catenin signaling and CXCL4 participated in cardiac fibrosis by activating PI3K/AKT pathway. Therefore, our findings revealed the role of CXCL4 in VMC and unveiled its underlying mechanism. CXCL4 appears to be a potential target for the treatment of VMC.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing 210006, China
| | - Ming Yu Peng
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing 211100, China
| | - Sai Nan Wang
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing 211100, China
| | - Hong Xiang Lu
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing 211100, China; Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing 210006, China.
| |
Collapse
|
4
|
Ah Kioon MD, Laurent P, Chaudhary V, Du Y, Crow MK, Barrat FJ. Modulation of plasmacytoid dendritic cells response in inflammation and autoimmunity. Immunol Rev 2024; 323:241-256. [PMID: 38553621 DOI: 10.1111/imr.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The discovery of toll-like receptors (TLRs) and the subsequent recognition that endogenous nucleic acids (NAs) could serve as TLR ligands have led to essential insights into mechanisms of healthy immune responses as well as pathogenic mechanisms relevant to systemic autoimmune and inflammatory diseases. In systemic lupus erythematosus, systemic sclerosis, and rheumatoid arthritis, NA-containing immune complexes serve as TLR ligands, with distinct implications depending on the additional immune stimuli available. Plasmacytoid dendritic cells (pDCs), the robust producers of type I interferon (IFN-I), are providing critical insights relevant to TLR-mediated healthy immune responses and tissue repair, as well as generation of inflammation, autoimmunity and fibrosis, processes central to the pathogenesis of many autoimmune diseases. In this review, we describe recent data characterizing the role of platelets and NA-binding chemokines in modulation of TLR signaling in pDCs, as well as implications for how the IFN-I products of pDCs contribute to the generation of inflammation and wound healing responses by monocyte/macrophages. Chemokine modulators of TLR-mediated B cell tolerance mechanisms and interactions between TLR signaling and metabolic pathways are also considered. The modulators of TLR signaling and their contribution to the pathogenesis of systemic autoimmune diseases suggest new opportunities for identification of novel therapeutic targets.
Collapse
Affiliation(s)
| | - Paôline Laurent
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Vidyanath Chaudhary
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Yong Du
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Mary K Crow
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Franck J Barrat
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
5
|
Wei J, Wang D, Cui C, Tan J, Peng M, Lu H. CXCL4/CXCR3 axis regulates cardiac fibrosis by activating TGF-β1/Smad2/3 signaling in mouse viral myocarditis. Immun Inflamm Dis 2024; 12:e1237. [PMID: 38577984 PMCID: PMC10996374 DOI: 10.1002/iid3.1237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Severe myocarditis is often accompanied by cardiac fibrosis, but the underlying mechanism has not been fully elucidated. CXCL4 is a chemokine that has been reported to have pro-inflammatory and profibrotic functions. The exact role of CXCL4 in cardiac fibrosis remains unclear. METHODS Viral myocarditis (VMC) models were induced by intraperitoneal injection of Coxsackie B Type 3 (CVB3). In vivo, CVB3 (100 TCID50) and CVB3-AMG487 (CVB3: 100 TCID50; AMG487: 5 mg/kg) combination were administered in the VMC and VMC+AMG487 groups, respectively. Hematoxylin and eosin staining, severity score, Masson staining, and immunofluorescence staining were performed to measure myocardial morphology in VMC. Enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were performed to quantify inflammatory factors (IL-1β, IL-6, TNF-α, and CXCL4). Aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and creatine kinase-myocardial band (CK-MB) levels were analyzed by commercial kits. CXCL4, CXCR3B, α-SMA, TGF-β1, Collagen I, and Collagen III were determined by Western blot and immunofluorescence staining. RESULTS In vivo, CVB3-AMG487 reduced cardiac injury, α-SMA, Collagen I and Collagen III levels, and collagen deposition in VMC+AMG487 group. Additionally, compared with VMC group, VMC+AMG group decreased the levels of inflammatory factors (IL-1β, IL-6, and TNF-α). In vitro, CXCL4/CXCR3B axis activation TGF-β1/Smad2/3 pathway promote mice cardiac fibroblasts differentiation. CONCLUSION CXCL4 acts as a profibrotic factor in TGF-β1/Smad2/3 pathway-induced cardiac fibroblast activation and ECM synthesis, and eventually progresses to cardiac fibrosis. Therefore, our findings revealed the role of CXCL4 in VMC and unveiled its underlying mechanism. CXCL4 appears to be a potential target for the treatment of VMC.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Dan‐feng Wang
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Cong‐cong Cui
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Jia‐jia Tan
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Ming‐yu Peng
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Hong‐xiang Lu
- Department of Laboratory Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingChina
- Department of Laboratory MedicineJiangning Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Platelet factor 4 (PF4, CXCL4), the most abundant α-granule platelet-specific chemokine, forms tetramers with an equatorial ring of high positive charge that bind to a wide range of polyanions, after which it changes conformation to expose antigenic epitopes. Antibodies directed against PF4 not only help to clear infection but can also lead to the development of thrombotic disorders such as heparin-induced thrombocytopenia (HIT) and vaccine-induced thrombocytopenia and thrombosis (VITT). This review will outline the different mechanisms through which PF4 engagement with polyanions combats infection but also contributes to the pathogenesis of inflammatory and thrombotic disease states. RECENT FINDINGS Recent work has shown that PF4 binding to microbial polyanions may improve outcomes in infection by enhancing leukocyte-bacterial binding, tethering pathogens to neutrophil extracellular traps (NETs), decreasing the thrombotic potential of NET DNA, and modulating viral infectivity. However, PF4 binding to nucleic acids may enhance their recognition by innate immune receptors, leading to autoinflammation. Lastly, while HIT is induced by platelet activating antibodies that bind to PF4/polyanion complexes, VITT, which occurs in a small subset of patients treated with COVID-19 adenovirus vector vaccines, is characterized by prothrombotic antibodies that bind to PF4 alone. SUMMARY Investigating the complex interplay of PF4 and polyanions may provide insights relevant to the treatment of infectious disease while also improving our understanding of the pathogenesis of thrombotic disorders driven by anti-PF4/polyanion and anti-PF4 antibodies.
Collapse
Affiliation(s)
- Anh T P Ngo
- Division of Hematology, Children's Hospital of Philadelphia
| | | | - Kandace Gollomp
- Division of Hematology, Children's Hospital of Philadelphia
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Muruganandam M, Ariza-Hutchinson A, Patel RA, Sibbitt WL. Biomarkers in the Pathogenesis, Diagnosis, and Treatment of Systemic Sclerosis. J Inflamm Res 2023; 16:4633-4660. [PMID: 37868834 PMCID: PMC10590076 DOI: 10.2147/jir.s379815] [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: 04/15/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by vascular damage, vasoinstability, and decreased perfusion with ischemia, inflammation, and exuberant fibrosis of the skin and internal organs. Biomarkers are analytic indicators of the biological and disease processes within an individual that can be accurately and reproducibly measured. The field of biomarkers in SSc is complex as recent studies have implicated at least 240 pathways and dysregulated proteins in SSc pathogenesis. Anti-nuclear antibodies (ANA) are classical biomarkers with well-described clinical classifications and are present in more than 90% of SSc patients and include anti-centromere, anti-Th/To, anti-RNA polymerase III, and anti-topoisomerase I antibodies. Transforming growth factor-β (TGF-β) is central to the fibrotic process of SSc and is intimately intertwined with other biomarkers. Tyrosine kinases, interferon-1 signaling, IL-6 signaling, endogenous thrombin, peroxisome proliferator-activated receptors (PPARs), lysophosphatidic acid receptors, and amino acid metabolites are new biomarkers with the potential for developing new therapeutic agents. Other biomarkers implicated in SSc-ILD include signal transducer and activator of transcription 4 (STAT4), CD226 (DNAX accessory molecule 1), interferon regulatory factor 5 (IRF5), interleukin-1 receptor-associated kinase-1 (IRAK1), connective tissue growth factor (CTGF), pyrin domain containing 1 (NLRP1), T-cell surface glycoprotein zeta chain (CD3ζ) or CD247, the NLR family, SP-D (surfactant protein), KL-6, leucine-rich α2-glycoprotein-1 (LRG1), CCL19, genetic factors including DRB1 alleles, the interleukins (IL-1, IL-4, IL-6, IL-8, IL-10 IL-13, IL-16, IL-17, IL-18, IL-22, IL-32, and IL-35), the chemokines CCL (2,3,5,13,20,21,23), CXC (8,9,10,11,16), CX3CL1 (fractalkine), and GDF15. Adiponectin (an indicator of PPAR activation) and maresin 1 are reduced in SSc patients. A new trend has been the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy. The present review is an update of the various biomarker molecules, pathways, and receptors involved in the pathology of SSc.
Collapse
Affiliation(s)
- Maheswari Muruganandam
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Angie Ariza-Hutchinson
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Rosemina A Patel
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Wilmer L Sibbitt
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
8
|
Blanchet X, Weber C, von Hundelshausen P. Chemokine Heteromers and Their Impact on Cellular Function-A Conceptual Framework. Int J Mol Sci 2023; 24:10925. [PMID: 37446102 DOI: 10.3390/ijms241310925] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Chemoattractant cytokines or chemokines are proteins involved in numerous biological activities. Their essential role consists of the formation of gradient and (immune) cell recruitment. Chemokine biology and its related signaling system is more complex than simple ligand-receptor interactions. Beside interactions with their cognate and/or atypical chemokine receptors, and glycosaminoglycans (GAGs), chemokines form complexes with themselves as homo-oligomers, heteromers and also with other soluble effector proteins, including the atypical chemokine MIF, carbohydrate-binding proteins (galectins), damage-associated molecular patterns (DAMPs) or with chemokine-binding proteins such as evasins. Likewise, nucleic acids have been described as binding targets for the tetrameric form of CXCL4. The dynamic balance between monomeric and dimeric structures, as well as interactions with GAGs, modulate the concentrations of free chemokines available along with the nature of the gradient. Dimerization of chemokines changes the canonical monomeric fold into two main dimeric structures, namely CC- and CXC-type dimers. Recent studies highlighted that chemokine dimer formation is a frequent event that could occur under pathophysiological conditions. The structural changes dictated by chemokine dimerization confer additional biological activities, e.g., biased signaling. The present review will provide a short overview of the known functionality of chemokines together with the consequences of the interactions engaged by the chemokines with other proteins. Finally, we will present potential therapeutic tools targeting the chemokine multimeric structures that could modulate their biological functions.
Collapse
Affiliation(s)
- Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| |
Collapse
|
9
|
Molecular Mechanisms Behind the Role of Plasmacytoid Dendritic Cells in Systemic Sclerosis. BIOLOGY 2023; 12:biology12020285. [PMID: 36829561 PMCID: PMC9953616 DOI: 10.3390/biology12020285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
Systemic sclerosis (SSc) is a debilitating autoimmune disease that affects multiple systems. It is characterized by immunological deregulation, functional and structural abnormalities of small blood vessels, and fibrosis of the skin, and, in some cases, internal organs. Fibrosis has a devastating impact on a patient's life and lung fibrosis is associated with high morbimortality. Several immune populations contribute to the progression of SSc, and plasmacytoid dendritic cells (pDCs) have been identified as crucial mediators of fibrosis. Research on murine models of lung and skin fibrosis has shown that pDCs are essential in the development of fibrosis, and that removing pDCs improves fibrosis. pDCs are a subset of dendritic cells (DCs) that are specialized in anti-viral responses and are also involved in autoimmune diseases, such as SSc, systemic lupus erythematosus (SLE) and psoriasis, mostly due to their capacity to produce type I interferon (IFN). A type I IFN signature and high levels of CXCL4, both derived from pDCs, have been associated with poor prognosis in patients with SSc and are correlated with fibrosis. This review will examine the recent research on the molecular mechanisms through which pDCs impact SSc.
Collapse
|