1
|
Peng B, Yan MY, Chen YR, Sun F, Xiang XD, Liu D. The methyl-CpG binding domain 2 regulates peptidylarginine deiminase 4 expression and promotes neutrophil extracellular trap formation via the Janus kinase 2 signaling pathway in experimental severe asthma. Ann Med 2025; 57:2458207. [PMID: 39865866 PMCID: PMC11774153 DOI: 10.1080/07853890.2025.2458207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
OBJECTIVE The prognosis for severe asthma is poor, and the current treatment options are limited. The methyl-CpG binding domain protein 2 (MBD2) participates in neutrophil-mediated severe asthma through epigenetic regulation. Neutrophil extracellular traps (NETs) play a critical role in the pathogenesis of severe asthma. This study aims to detect if MBD2 can reduce NETs formation and the potential mechanism in severe asthma. METHODS A severe asthma model was established in C57BL/6 wild-type mice exposure to house dust mite (HDM), ovalbumin (OVA), and lipopolysaccharide (LPS). Enzyme-linked immunosorbent assay was used to measure the concentrations of IL-4, IL-17A, and IFN-γ in lung tissues. Flow cytometry was employed to determine the percentages of Th2, Th17, and Treg cells in lung tissues. Quantitative real-time polymerase chain reaction was utilized to assess the mRNA expression levels of MBD2, JAK2, and PAD4. Western blotting and immunofluorescence were conducted to detect the protein of MBD2, JAK2, PAD4, and CitH3. HL-60 cells were differentiated into neutrophil-like cells by culturing in a medium containing dimethyl sulfoxide and then stimulated with LPS. KCC-07, Ruxolitinib, and Cl-amidine were used to inhibit the expressions of MBD2, JAK2, and PAD4, respectively. RESULTS Severe asthma mice were characterized by pulmonary neutrophilic inflammation and increased formation of neutrophil extracellular traps (NETs). The expression of MBD2, JAK2, and PAD4 was elevated in severe asthma mice. Inhibiting the expression of MBD2, JAK2, and PAD4 reduced NETs formation and decreased airway inflammation scores, total cell counts and neutrophil counts in BALF, and percentage of Th2 and Th17 cell in lung tissues, whereas increasing Treg cell counts. In both severe asthma mice and HL-60-differentiated neutrophil-like cells in vitro, inhibiting MBD2 reduced the mRNA and protein expression of JAK2 and PAD4, and inhibiting JAK2 reduced the expression of PAD4 mRNA and protein. CONCLUSION MBD2 regulates PAD4 expression through the JAK2 signaling pathway to promote NETs formation in mice with severe asthma. Further bench-based and bedside-based studies targeting the MBD2, PAD4, and JAK2 signaling pathways will help open new avenues for drug development of severe asthma.
Collapse
Affiliation(s)
- Biao Peng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Mu-Yun Yan
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Yun-Rong Chen
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People’s Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Fei Sun
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xu-Dong Xiang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Da Liu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
2
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [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: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
3
|
Zheng L, Adili R, Wu Z, Zhang Q, Zhu G, Lei X, Liu Z, Neves MAD, Ma W, Slavkovic S, Xu XR, Ni H, Zheng XL. Preventative and therapeutic effects of a novel humanized anti-glycoprotein Ibα fragment of antigen-binding region in a murine model of thrombotic thrombocytopenic purpura. J Thromb Haemost 2025; 23:1596-1607. [PMID: 39956431 DOI: 10.1016/j.jtha.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/20/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Thrombotic thrombocytopenic purpura (TTP) is a potentially fatal blood disorder resulting from severe deficiency of plasma ADAMTS-13 activity. Current treatment for immune-mediated TTP includes daily therapeutic plasma exchange plus caplacizumab and immunosuppressives. For hereditary TTP resulting from mutations of ADAMTS-13, plasma infusion or recombinant ADAMTS-13 is the treatment of choice. However, there are still unmet needs for an effective alternative therapy for TTP. OBJECTIVES The present study aimed to evaluate the therapeutic efficacy of a novel humanized antibody fragment of antigen binding against platelet glycoprotein Ibα (CA1001) in a murine model of TTP. METHODS Platelet agglutination profiles, microfluidic shear-based assay, intravital microscopy thrombosis model, and lysine histone-induced murine "TTP-like" model were employed. RESULTS CA1001 exhibited potent inhibition of botrocetin-induced murine platelet agglutination in a dose- and time-dependent manner. CA1001 also significantly inhibited shear-dependent adhesion and aggregation of murine platelets to endothelial von Willebrand factor (VWF) released from calcium ionophore-activated cremaster venules in Adamts-13 null mice and blocked the formation of platelet-VWF rich thrombosis. More importantly, CA1001 appeared to be efficacious in preventing and treating a histone-induced "TTP-like" syndrome in Adamts-13 null mice, demonstrated by the alleviation of thrombocytopenia, prerenal injury, and formation of microvascular thrombosis in major organ tissues. CONCLUSION CA1001 can effectively inhibit VWF-platelet interaction and thrombus formation under various (patho)physiological conditions. Thus, CA1001 may be a potential candidate for further development as a novel therapeutic for immune-mediated and hereditary TTP and perhaps for other inflammatory thrombotic disorders such as ischemic stroke.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, Kansas, USA.
| | - Reheman Adili
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Bloodworks Research Institute, Seattle, Washington, USA
| | - Zhijian Wu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Zhenze Liu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Miguel A D Neves
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Sladjana Slavkovic
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
4
|
Fernandez-Flores A. Concepts in Cutaneous Vasculitis. Am J Dermatopathol 2025; 47:165-177. [PMID: 39950765 DOI: 10.1097/dad.0000000000002807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
ABSTRACT Skin biopsy plays a fundamental role in the diagnosis of vasculitis. However, the general pathologist or dermatopathologist who encounters these diagnostic findings in their early stages often faces the paradox that the clinician requests the exclusion of various systemic diseases, when the biopsy only shows leukocytoclastic vasculitis. In other cases, even though the affected vessels are small, some of them seem deep within the biopsy, raising differential diagnosis with several entities of systemic repercussion. Lastly, although the dermatopathologist has a histological picture before them, they are often required to correlate it with laboratory data such as the presence of antineutrophil antibodies, for example. Therefore, the objective of this article is conceptual, emphasizing those basic aspects that can contribute to a better understanding and diagnosis of skin biopsy in vasculitis.
Collapse
Affiliation(s)
- Angel Fernandez-Flores
- Pathologist, Department of Cellular Pathology, Hospital El Bierzo, Ponferrada, Spain
- Pathologist, Department of Cellular Pathology, Hospital de la Reina, Ponferrada, Spain; and
- Pathologist, Department of Research, Institute for Biomedical Research of A Coruña (INIBIC), University of A Coruña (UDC), A Coruña, Spain
| |
Collapse
|
5
|
Li X, Guo Y, Wang Z, Guo X, Wang J, Zhang J, Zhang T, Wang J, Li T, Zhou J, Zhang N, Amin B, Zhu B. Platelet-rich fibrin promotes mesothelial cell proliferation and peritoneal repair by up-regulating calretinin to prevent postoperative intestinal adhesion. Int J Med Sci 2025; 22:1254-1268. [PMID: 40084247 PMCID: PMC11898853 DOI: 10.7150/ijms.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction: As the most common postoperative complication, intestinal adhesions can cause intestinal obstruction, female infertility, and even endanger life. The currently developed materials for preventing intestinal adhesions mainly focus on physical barriers and reducing inflammatory reactions, while neglecting the importance of effectively promoting rapid repair of the peritoneum. We previously found that platelet-rich fibrin (PRF) can prevent postoperative intestinal adhesions. The proliferation of mesothelial cells may play a significant role in reducing intestinal adhesions, but the mechanism remains unclear. A study found a positive correlation between calretinin (CR) and mesothelial cell proliferation. Does CR play an important role in PRF promoting mesothelial cell proliferation? This study aims to further explore the mechanism of PRF in preventing intestinal adhesions. Methods: Primary mouse peritoneal mesothelial cells and mouse peritoneal fibroblasts were used in this study. The effects of PRF on the proliferation and attachment of mesothelial cells and fibroblasts were observed and compared using the CCK-8 assay, Edu assay, and laser scanning confocal microscope. The effects of PRF on the migration of mesothelial cells were examined using scratch and transwell migration assays. The effects of PRF on the mesothelial-mesenchymal transition (MMT) of mesothelial cells were examined using western blot. The expression level of CR in mesothelial cells was detected through immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot. Results: PRF promotes mesothelial cell proliferation from 1st day and significantly promotes fibroblast proliferation from 7th day. Meanwhile, PRF tends to promote the proliferation and attachment of mesothelial cells rather than fibroblasts. However, PRF had a limited regulatory effect on the MMT of mesothelial cells. In addition, PRF can promote mesothelial cell migration and upregulate the expression level of CR. Conclusion: PRF promotes mesothelial cell proliferation and migration, as well as peritoneal repair, by up-regulating CR in the early stages of peritoneal injury to prevent postoperative intestinal adhesion. Its mechanism is obviously different from that of traditional anti-adhesion materials, which will provide new strategies for the prevention of intestinal adhesions.
Collapse
Affiliation(s)
- Xinming Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yifan Guo
- Department of Thoracic Surgery, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Zhuoyin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xu Guo
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jia Wang
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Jianlu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Zhang
- Department of Endocrinology, Beijing Liangxiang Hospital, Capital Medical University, Beijing, 102401, China
| | - Jing Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Tianxiong Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jian Zhou
- Department of Science and Technology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Nengwei Zhang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Buhe Amin
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Bin Zhu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| |
Collapse
|
6
|
Tang W, Zhang Y, Lu S, Xue C. Association between ATG16L1 rs2241880(T300A) and rs4663421 and ANCA‑associated vasculitis in the Guangxi population of China: Propensity score matching analysis. Biomed Rep 2025; 22:3. [PMID: 39483332 PMCID: PMC11522951 DOI: 10.3892/br.2024.1880] [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: 04/12/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
Antineutrophil cytoplasmic antibody-associated vasculitis (AAV) is a rare autoimmune disease with an unclear pathogenesis. The present study investigated the associations between autophagy-related protein 16-like 1 (ATG16L1) rs2241880(T300A) and rs4663421 and AAV. A total of 177 patients with AAV and 216 healthy controls were included. Propensity score matching was used to match the two groups of subjects in terms of sex, age and ethnicity. Analyses of the relationships between these genetic polymorphisms and AAV susceptibility, including comparisons of allele and genotype frequency distribution, linkage disequilibrium analysis and analysis of single nucleotide polymorphism (SNP) interactions between two loci were performed. The association between the loci and laboratory test results and renal pathology were also analysed. A total of 154 pairs of patients with AAV and healthy controls was successfully matched. Neither polymorphism was associated with AAV susceptibility. However, SNP interaction in the model constructed with the two loci was statistically significant (P=0.018), and the combination of the AA genotype of rs2241880(T300A) and GG genotype of rs4663421 was associated the highest disease risk. The differences in the Birmingham Vasculitis Activity Score (BVAS), C-reactive protein (CRP) levels and 24-h urine protein level between patients with the rs2241880(T300A) AA + AG genotypes and the GG genotype were statistically significant (P<0.05). Furthermore, significant differences in the severity of glomerulosclerosis and global sclerosis were detected between individuals with the AA + AG genotype and those with the GG genotype at the rs2241880(T300A) locus (P<0.05). Similarly, there were statistically significant differences in degree of segmental sclerosis between individuals with CC + CG genotypes and those with GG genotypes at the rs2243421 locus (P<0.05). In summary, the single gene polymorphisms of these loci were not associated with genetic susceptibility to AAV. However, SNP interactions may serve a role in the risk of AAV. The rs2241880(T300A) polymorphism may be associated with BVAS, CRP levels and 24-h urine protein level in AAV. These SNPs may be associated with glomerulosclerosis and segmental sclerosis.
Collapse
Affiliation(s)
- Wenlv Tang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Yurong Zhang
- Department of Electrocardiographic Diagnosis, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530000, P.R. China
| | - Shurong Lu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Chao Xue
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| |
Collapse
|
7
|
Hua L, Xie M. Heterogeneity and individualized therapy for eosinophilic granulomatosis with polyangiitis. Ther Adv Respir Dis 2025; 19:17534666251318615. [PMID: 39980304 PMCID: PMC11843704 DOI: 10.1177/17534666251318615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA), as a heterogeneous component of antineutrophil cytoplasmic antibody-associated vasculitis, may be induced by a series of environmental and genetic factors, involved with a variety of immune cells and immune components, and presented with various clinical manifestations, with multiple organs and systems (respiratory, skin, heart, kidney, nerve, etc.) involved. The choice of glucocorticoid (GC) dosage and immunosuppressant in traditional treatment strategies varies greatly from individual to individual and is not universally applicable in all the EGPA phenotype spectrum, especially in relapsing or refractory diseases. With the understanding of the heterogeneity of EGPA, a variety of therapeutic approaches are emerging and improving the traditional treatment model. In this review, we summarized the heterogeneity of EGPA etiology and pathogenesis. Clinical and pathological manifestations of the same organ involved also show significant differences and there are even gender differences. Biological treatments that mainly target type 2 inflammatory pathways are widely used in clinical practice for remission induction and maintenance of EGPA. Targeted biological therapy has shown excellent performance in reducing GC dosage and controlling symptoms and recurrence. However, a large number of high-quality randomized controlled studies are still under research for relapsing or refractory EGPA with special organ involvement. We believe that EGPA has a highly heterogeneous phenotype spectrum, and the treatment patterns targeting key molecules in the pathogenesis are of great value for individual treatment of EGPA.
Collapse
Affiliation(s)
- Lijuan Hua
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Xie
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| |
Collapse
|
8
|
Hernández-López A, Reyna-Juárez Y, Ostos-Prado MJ, Alcalá-Carmona B, Torres-Ruiz J, Méndez-Flores S, Escobar-Ceballos S, Martínez-Benitez B, Gómez-Martín D. Anti-synthetase and myelodysplastic syndromes with deep morphea: an example of shared immunopathogenesis? A case-based review. Rheumatol Int 2024; 44:2645-2652. [PMID: 39249142 DOI: 10.1007/s00296-024-05717-y] [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: 06/24/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
Anti-synthetase syndrome (AS) is a subset of idiopathic inflammatory myopathy (IIM) characterized by the presence of anti-aminoacyl-transfer RNA synthetase accompanied by myositis, interstitial lung disease and other clinical features. According to a recent multicentric study, 31% of AS patients present skin lesions compatible with dermatomyositis, but sclerodermiform features are rare. Therefore, we aimed to report the case of a patient with simultaneous diagnosis of AS, deep morphea, vasculitic neuropathy, and myelodysplastic syndrome and review the current literature regarding these uncommon associations. A 57 year old man with axial and symmetrical proximal muscle weakness, skin thickening and B symptoms, later diagnosed with PL7 + AS, deep morphea, myelodysplastic syndrome (MDS) and vasculitic neuropathy documented by histopathologic studies and immunologic assessments. Since both AS and deep morphea share the vasculopathic changes and type II interferon-induced inflammation, we hypothesize that they may share pathogenic mechanisms. The muscle biopsy of the patient was consistent with AS and showed focal neutrophil infiltration. The patient received intensive immunosuppressive therapy for AS and vasculitic neuropathy, with high dose steroids, intravenous immunoglobulin (IVIg) and rituximab. Nonetheless, he suffered an unfavorable evolution with a fatal outcome due to septic shock. Albeit sclerodermiform features are rare in patients with AS, we propose a pathogenic link among AS, deep morphea and the autoimmune/autoinflammatory signs of MDS. The vasculopathic changes along with the activation of the innate and adaptive immune system leading to the production of proinflammatory cytokines may have been one of the contributing factors for the coexisting diagnosis of the patient.
Collapse
Affiliation(s)
- Agustín Hernández-López
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Yatzil Reyna-Juárez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - María José Ostos-Prado
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Beatriz Alcalá-Carmona
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Silvia Méndez-Flores
- Department of Dermatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Salvador Escobar-Ceballos
- Department of Dermatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Braulio Martínez-Benitez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
9
|
Osorio M, Velásquez I, Vargas R, Vanegas-García A, Rojas M, Vásquez G, Muñoz-Vahos C. NETosis Secondary to the Use of Levamisole-Adulterated Cocaine: A Likely Underlying Mechanism of Vasculopathy. J Toxicol 2024; 2024:7388799. [PMID: 38434602 PMCID: PMC10904679 DOI: 10.1155/2024/7388799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 03/05/2024] Open
Abstract
Background Since 2010, several cases of a new vasculopathy induced by the use of levamisole-adulterated cocaine (LAC) have been reported. This vasculopathy is characterized by retiform purpura, earlobe necrosis, multisystem compromise, and multiple autoantibodies. Given its similarity to antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis, LAC-associated vasculopathy is postulated to be mediated by pathophysiologic processes resulting from neutrophil cell death by NETosis, a phenomenon previously described in ANCA vasculitis. This study tries to establish the presence of NETosis induced by cocaine, levamisole, or both. Methodology. Neutrophils were isolated from the peripheral blood of healthy controls by Ficoll-Hystopaque density gradient centrifugation followed by dextran sedimentation. Cell viability and purity were evaluated by flow cytometry after staining with PI/DiOC6 and labeling with fluorescent anti-CD45/anti-CD3 monoclonal antibodies (mAbs), respectively. Neutrophils were exposed to levamisole, cocaine, a cocaine-levamisole mixture, and sera pools from healthy controls and patients with LAC-associated vasculopathy. NETosis was then assessed by flow cytometry after staining cells with Sytox Green, Hoechst-33342, and fluorescent antineutrophil elastase (NE) and antimyeloperoxidase (MPO) mAbs. In addition, NETosis was morphologically confirmed by fluorescence microscopy. Proinflammatory cytokine levels in culture supernatants and reactive oxygen species (ROS) synthesis were determined by flow cytometry. The involvement of calcium and muscarinic receptors in cell death induction was evaluated in parallel experiments carried out in the presence of 1,2-bis (o-aminophenoxy) ethane-N, N, N', N'-tetraacetic acid (BAPTA) and hyoscine butylbromide (HBB), their respective inhibitors. Results Cocaine, levamisole, and a cocaine-levamisole mixture induced neutrophil cell death. DNA/MPO extrusion and cell morphology patterns were consistent with NETosis. Neither proinflammatory cytokines nor ROS behaved as proNETotic factors. Preliminary results suggested that muscarinic receptors and calcium-dependent signals were involved in LAC-induced NETosis. Conclusions Cocaine, levamisole, and a cocaine-levamisole mixture can induce NETosis through mechanisms involving muscarinic receptors and calcium-dependent pathways.
Collapse
Affiliation(s)
- Manuela Osorio
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Isabel Velásquez
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Ruben Vargas
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Adriana Vanegas-García
- Hospital Universitario San Vicente Fundación, Medellín, Colombia
- Sección de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Gloria Vásquez
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Carlos Muñoz-Vahos
- Hospital Universitario San Vicente Fundación, Medellín, Colombia
- Sección de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
10
|
Yoshida Y, Nakamoto N, Oka N, Kidoguchi G, Hosokawa Y, Araki K, Ishitoku M, Watanabe H, Sugimoto T, Mokuda S, Kida T, Yajima N, Omura S, Nakagomi D, Abe Y, Kadoya M, Takizawa N, Nomura A, Kukida Y, Kondo N, Yamano Y, Yanagida T, Endo K, Matsui K, Takeuchi T, Ichinose K, Kato M, Yanai R, Matsuo Y, Shimojima Y, Nishioka R, Okazaki R, Takata T, Ito T, Moriyama M, Takatani A, Miyawaki Y, Ito-Ihara T, Kawaguchi T, Kawahito Y, Hirata S. Seasonal Influence on Development of Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: A Retrospective Cohort Study Conducted at Multiple Institutions in Japan (J-CANVAS). J Rheumatol 2023; 50:1152-1158. [PMID: 37263656 DOI: 10.3899/jrheum.2023-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVE To clarify seasonal and other environmental effects on the onset of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). METHODS We enrolled patients with new-onset eosinophilic granulomatosis with polyangiitis (EGPA), microscopic polyangiitis (MPA), and granulomatosis with polyangiitis (GPA) registered in the database of a Japanese multicenter cohort study. We investigated the relationship between environmental factors and clinical characteristics. Seasons were divided into 4 (spring, summer, autumn, and winter), and the seasonal differences in AAV onset were analyzed using Pearson chi-square test, with an expected probability of 25% for each season. RESULTS A total of 454 patients were enrolled, with a mean age of 70.9 years and a female proportion of 55.5%. Overall, 74, 291, and 89 patients were classified as having EGPA, MPA, and GPA, respectively. Positivity for myeloperoxidase (MPO)-ANCA and proteinase 3 (PR3)-ANCA was observed in 355 and 46 patients, respectively. Overall, the seasonality of AAV onset significantly deviated from the expected 25% for each season (P = 0.001), and its onset was less frequently observed in autumn. In ANCA serotypes, seasonality was significant in patients with MPO-ANCA (P < 0.001), but not in those with PR3-ANCA (P = 0.97). Additionally, rural residency of patients with AAV was associated with PR3-ANCA positivity and biopsy-proven pulmonary vasculitis. CONCLUSION The onset of AAV was influenced by seasonal variations and was less frequently observed in autumn. In contrast, the occurrence of PR3-ANCA was triggered, not by season, but by rural residency.
Collapse
Affiliation(s)
- Yusuke Yoshida
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima;
| | - Naoki Nakamoto
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Naoya Oka
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Genki Kidoguchi
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Yohei Hosokawa
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Kei Araki
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Michinori Ishitoku
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Hirofumi Watanabe
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Tomohiro Sugimoto
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Sho Mokuda
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| | - Takashi Kida
- T. Kida, MD, PhD, MPH, S. Omura, MD, Y. Kawahito, MD, PhD, Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto
| | - Nobuyuki Yajima
- N. Yajima, MD, PhD, MPH, R. Yanai, MD, Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo
| | - Satoshi Omura
- T. Kida, MD, PhD, MPH, S. Omura, MD, Y. Kawahito, MD, PhD, Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto
| | - Daiki Nakagomi
- D. Nakagomi, MD, PhD, Department of Rheumatology, University of Yamanashi Hospital, Chuo
| | - Yoshiyuki Abe
- Y. Abe, MD, PhD, Department of Internal Medicine and Rheumatology, Juntendo University, Tokyo
| | - Masatoshi Kadoya
- M. Kadoya, MD, PhD, Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto
| | - Naoho Takizawa
- N. Takizawa, MD, Department of Rheumatology, Chubu Rosai Hospital, Nagoya
| | - Atsushi Nomura
- A. Nomura, MD, PhD, Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo
| | - Yuji Kukida
- Y. Kukida, MD, PhD, Department of Rheumatology, Japanese Red Cross Society Kyoto Daini Hospital, Kyoto
| | - Naoya Kondo
- N. Kondo, PhD, Department of Nephrology, Kyoto Katsura Hospital, Kyoto
| | - Yasuhiko Yamano
- Y. Yamano, PhD, Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto
| | - Takuya Yanagida
- T. Yanagida, MD, Department of Hematology and Rheumatology, Kagoshima University Hospital, Kagoshima
| | - Koji Endo
- K. Endo, MD, Department of General Internal Medicine, Tottori Prefectural Central Hospital, Tottori, and Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto
| | - Kiyoshi Matsui
- K. Matsui, MD, PhD, Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya
| | - Tohru Takeuchi
- T. Takeuchi, MD, PhD, Department of Internal Medicine (IV), Osaka Medical and Pharmaceutical University, Osaka
| | - Kunihiro Ichinose
- K. Ichinose, MD, PhD, Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, and Department of Rheumatology, Shimane University Faculty of Medicine, Izumo
| | - Masaru Kato
- M. Kato, MD, PhD, Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine, Hokkaido University, Sapporo
| | - Ryo Yanai
- N. Yajima, MD, PhD, MPH, R. Yanai, MD, Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo
| | - Yusuke Matsuo
- Y. Matsuo, MD, PhD, Department of Rheumatology, Tokyo Kyosai Hospital, and Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo
| | - Yasuhiro Shimojima
- Y. Shimojima, MD, PhD, Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto
| | - Ryo Nishioka
- R. Nishioka, MD, Department of Rheumatology, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Ryota Okazaki
- R. Okazaki, MD, PhD, Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago
| | - Tomoaki Takata
- T. Takata, MD, PhD, Division of Gastroenterology and Nephrology, Tottori University, Yonago
| | - Takafumi Ito
- T. Ito, MD, PhD, Division of Nephrology, Shimane University Hospital, Izumo
| | - Mayuko Moriyama
- M. Moriyama, MD, Department of Rheumatology, Shimane University Faculty of Medicine, Izumo
| | - Ayuko Takatani
- A. Takatani, MD, PhD, Rheumatic Disease Center, Sasebo Chuo Hospital, Sasebo
| | - Yoshia Miyawaki
- Y. Miyawaki, MD, PhD, Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama
| | - Toshiko Ito-Ihara
- T. Ito-Ihara, MD, PhD, The Clinical and Translational Research Center, University Hospital, Kyoto Prefectural University of Medicine, Kyoto
| | - Takashi Kawaguchi
- T. Kawaguchi, PhD, Department of Practical Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yutaka Kawahito
- T. Kida, MD, PhD, MPH, S. Omura, MD, Y. Kawahito, MD, PhD, Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto
| | - Shintaro Hirata
- Y. Yoshida, MD, PhD, N. Nakamoto, MD, N. Oka, MD, G. Kidoguchi, MD, Y. Hosokawa, MD, K. Araki, MD, M. Ishitoku, MD, H. Watanabe, MD, T. Sugimoto, MD, PhD, S. Mokuda, MD, PhD, S. Hirata, MD, PhD, Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima
| |
Collapse
|
11
|
Poledniczek M, Neumayer C, Kopp CW, Schlager O, Gremmel T, Jozkowicz A, Gschwandtner ME, Koppensteiner R, Wadowski PP. Micro- and Macrovascular Effects of Inflammation in Peripheral Artery Disease-Pathophysiology and Translational Therapeutic Approaches. Biomedicines 2023; 11:2284. [PMID: 37626780 PMCID: PMC10452462 DOI: 10.3390/biomedicines11082284] [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: 06/25/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammation has a critical role in the development and progression of atherosclerosis. On the molecular level, inflammatory pathways negatively impact endothelial barrier properties and thus, tissue homeostasis. Conformational changes and destruction of the glycocalyx further promote pro-inflammatory pathways also contributing to pro-coagulability and a prothrombotic state. In addition, changes in the extracellular matrix composition lead to (peri-)vascular remodelling and alterations of the vessel wall, e.g., aneurysm formation. Moreover, progressive fibrosis leads to reduced tissue perfusion due to loss of functional capillaries. The present review aims at discussing the molecular and clinical effects of inflammatory processes on the micro- and macrovasculature with a focus on peripheral artery disease.
Collapse
Affiliation(s)
- Michael Poledniczek
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Thomas Gremmel
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria;
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Michael E. Gschwandtner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| |
Collapse
|
12
|
Li S, Wang H, Shao Q. The central role of neutrophil extracellular traps (NETs) and by-products in COVID-19 related pulmonary thrombosis. Immun Inflamm Dis 2023; 11:e949. [PMID: 37647446 PMCID: PMC10461423 DOI: 10.1002/iid3.949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
Extracellular trap networks (neutrophil extracellular traps [NETs]) of polymorphonuclear neutrophils are mesh-like substances that prevent the spread of pathogens. They primarily consist of DNA skeletons, histones, granule components, and cytoplasmic proteins. NETs formation requires a certain environment and there are different pathways for NETs production. However, it is still not clear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) promotes NETs. NETs exert antiinflammatory effects through immune response, while they can also lead to certain adverse outcomes, such as the development of immunothrombosis. Coronavirus disease 2019 (COVID-19) is an inflammatory reaction affecting various organs caused by SARS-CoV-2, especially the lungs. NETs production and disease severity are linked with unique neutrophil clusters by single-cell RNA sequencing. NETs might exert an anti-inflammatory role in the initial stage of lung tissue inflammation. Nevertheless, numerous studies and cases have shown that they can also result in pulmonary thrombosis. There is mounting evidence that NETs are tightly related with COVID-19 pulmonary thrombosis, and many studies on the mechanisms are involved. The role and mechanism of NETs in the development of pulmonary thrombosis will be the main topics of this manuscript. Additionally, we address the potential targeting of NETs in COVID-19 patients.
Collapse
Affiliation(s)
- Shi Li
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Hui Wang
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Qixiang Shao
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
- Department of Medical Microbiology and Immunology, Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory MedicineJiangsu College of NursingHuai'anJiangsuChina
| |
Collapse
|
13
|
Magaña-Guerrero FS, Aguayo-Flores JE, Buentello-Volante B, Zarco-Ávila K, Sánchez-Cisneros P, Castro-Salas I, De la Torre-Galván E, Rodríguez-Loaiza JL, Jiménez-Corona A, Garfias Y. Spontaneous Neutrophil Extracellular Traps Release Are Inflammatory Markers Associated with Hyperglycemia and Renal Failure on Diabetic Retinopathy. Biomedicines 2023; 11:1791. [PMID: 37509431 PMCID: PMC10376331 DOI: 10.3390/biomedicines11071791] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic retinopathy (DR) is the major microvascular complication of diabetes and causes vitreous traction and intraretinal hemorrhages leading to retinal detachment and total blindness. The evolution of diabetes is related to exacerbating inflammation caused by hyperglycemia and activation of inflammatory cells. Neutrophils are cells able to release structures of extracellular DNA and proteolytic enzymes called extracellular traps (NETs), which are associated with the persistence of inflammation in chronic pathologies. The purpose of the study was to determine the usefulness of neutrophil traps as indicators of DR progression in patients with type 2 diabetes (T2DM). We performed a case-control study of seventy-four cases classified into five groups (non-proliferative DR, mild, moderate, severe, and proliferative) and fifteen healthy controls. We found correlations between NETs and a diagnostic time of T2DM (r = 0.42; p < 0.0001), fasting glucose (r = 0.29; p < 0.01), glycated hemoglobin (HbA1c) (r = 0.31; p < 0.01), estimated glomerular filtration rate (eGFR) (r = -0.29; p < 0.01), and plasma osmolarity (r = 0.25; p < 0.01). These results suggest that due to NETs being associated with clinical indicators, such as HbA1c and eGFR, and that NETs are also associated with DR, clinical indicators might be explained in part through an NET-mediated inflammation process.
Collapse
Affiliation(s)
- Fátima Sofía Magaña-Guerrero
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - José Eduardo Aguayo-Flores
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Beatriz Buentello-Volante
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Karla Zarco-Ávila
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Paola Sánchez-Cisneros
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Ilse Castro-Salas
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Enya De la Torre-Galván
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | | | - Aida Jiménez-Corona
- Department of Ocular Epidemiology and Visual Health, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
- General Directorate of Epidemiology, Health Secretariat, Mexico City 01480, Mexico
| | - Yonathan Garfias
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
- Department of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
14
|
Fijolek J, Radzikowska E. Eosinophilic granulomatosis with polyangiitis - Advances in pathogenesis, diagnosis, and treatment. Front Med (Lausanne) 2023; 10:1145257. [PMID: 37215720 PMCID: PMC10193253 DOI: 10.3389/fmed.2023.1145257] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/13/2023] [Indexed: 05/24/2023] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare disease characterized by eosinophil-rich granulomatous inflammation and necrotizing vasculitis, pre-dominantly affecting small-to-medium-sized vessels. It is categorized as a primary antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) but also shares features of hypereosinophilic syndrome (HES); therefore, both vessel inflammation and eosinophilic infiltration are suggested to cause organ damage. This dual nature of the disease causes variable clinical presentation. As a result, careful differentiation from mimicking conditions is needed, especially from HES, given the overlapping clinical, radiologic, and histologic features, and biomarker profile. EGPA also remains a diagnostic challenge, in part because of asthma, which may pre-dominate for years, and often requires chronic corticosteroids (CS), which can mask other disease features. The pathogenesis is still not fully understood, however, the interaction between eosinophils and lymphocytes B and T seems to play an important role. Furthermore, the role of ANCA is not clear, and only up to 40% of patients are ANCA-positive. Moreover, two ANCA-dependent clinically and genetically distinct subgroups have been identified. However, a gold standard test for establishing a diagnosis is not available. In practice, the disease is mainly diagnosed based on the clinical symptoms and results of non-invasive tests. The unmet needs include uniform diagnostic criteria and biomarkers to help distinguish EGPA from HESs. Despite its rarity, notable progress has been made in understanding the disease and in its management. A better understanding of the pathophysiology has provided new insights into the pathogenesis and therapeutic targets, which are reflected in novel biological agents. However, there remains an ongoing reliance on corticosteroid therapy. Therefore, there is a significant need for more effective and better-tolerated steroid-sparing treatment schemes.
Collapse
|
15
|
Sen CK, Roy S, Khanna S. Diabetic Peripheral Neuropathy Associated with Foot Ulcer: One of a Kind. Antioxid Redox Signal 2023. [PMID: 35850520 DOI: 10.1089/ars.2022.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Significance: Diabetic peripheral neuropathy (DPN) associated with a diabetic foot ulcer (DFU) is likely to be complicated with critical factors such as biofilm infection and compromised skin barrier function of the diabetic skin. Repaired skin with a history of biofilm infection is known to be compromised in barrier function. Loss of barrier function is also observed in the oxidative stress affected diabetic and aged skin. Recent Advances: Loss of barrier function makes the skin prone to biofilm infection and cellulitis, which contributes to chronic inflammation and vasculopathy. Hyperglycemia favors biofilm formation as glucose lowering led to reduction in biofilm development. While vasculopathy limits oxygen supply, the O2 cost of inflammation is high increasing hypoxia severity. Critical Issues: The host nervous system can be inhabited by bacteria. Because electrical impulses are a part of microbial physiology, polymicrobial colonization of the host's neural circuit is likely to influence transmission of action potential. The identification of perineural apatite in diabetic patients with peripheral neuropathy suggests bacterial involvement. DPN starts in both feet at the same time. Future Directions: Pair-matched studies of DPN in the foot affected with DFU (i.e., DFU-DPN) compared with DPN in the without ulcer, and intact skin barrier function, are likely to provide critical insight that would help inform effective care strategies. This review characterizes DFU-DPN from a translational science point of view presenting a new paradigm that recognizes the current literature in the context of factors that are unique to DFU-DPN.
Collapse
Affiliation(s)
- Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Savita Khanna
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Ta H, Awada H, Kang P, Gilbert N, Haller N, Mostow E, Lane J, Singh I. Antineutrophil Cytoplasmic Autoantibody (ANCA)-Associated Vasculitis With Mucosal Involvement Following COVID-19 Pneumonia. Cureus 2022; 14:e31441. [DOI: 10.7759/cureus.31441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/14/2022] Open
|
17
|
Zhao WM, Wang ZJ, Shi R, Zhu YY, Zhang S, Wang RF, Wang DG. Environmental factors influencing the risk of ANCA-associated vasculitis. Front Immunol 2022; 13:991256. [PMID: 36119110 PMCID: PMC9479327 DOI: 10.3389/fimmu.2022.991256] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of diseases characterized by inflammation and destruction of small and medium-sized blood vessels. Clinical disease phenotypes include microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), and eosinophilic granulomatosis with polyangiitis (EGPA). The incidence of AAV has been on the rise in recent years with advances in ANCA testing. The etiology and pathogenesis of AAV are multifactorial and influenced by both genetic and environmental factors, as well as innate and adaptive immune system responses. Multiple case reports have shown that sustained exposure to silica in an occupational environment resulted in a significantly increased risk of ANCA positivity. A meta-analysis involving six case-control studies showed that silica exposure was positively associated with AAV incidence. Additionally, exposure to air pollutants, such as carbon monoxide (CO), is a risk factor for AAV. AAV has seasonal trends. Studies have shown that various environmental factors stimulate the body to activate neutrophils and expose their own antigens, resulting in the release of proteases and neutrophil extracellular traps, which damage vascular endothelial cells. Additionally, the activation of complement replacement pathways may exacerbate vascular inflammation. However, the role of environmental factors in the etiology of AAV remains unclear and has received little attention. In this review, we summarized the recent literature on the study of environmental factors, such as seasons, air pollution, latitude, silica, and microbial infection, in AAV with the aim of exploring the relationship between environmental factors and AAV and possible mechanisms of action to provide a scientific basis for the prevention and treatment of AAV.
Collapse
|
18
|
Surmiak M, Wawrzycka-Adamczyk K, Kosałka-Węgiel J, Polański S, Sanak M. Profile of circulating extracellular vesicles microRNA correlates with the disease activity in granulomatosis with polyangiitis. Clin Exp Immunol 2022; 208:103-113. [PMID: 35380163 PMCID: PMC9113355 DOI: 10.1093/cei/uxac022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 01/12/2023] Open
Abstract
Granulomatosis with polyangiitis is a chronic systemic inflammation of small vessels characterized by circulating anti-proteinase 3 antibodies. MicroRNAs are short transcripts specifically inhibiting protein translation. Neutrophils can release extracellular vesicles (EVs). In this study, we characterized profile of microRNA trafficked by EVs in GPA. Fifty patients with GPA were enrolled in the study, 25 at acute phase and 25 in remission. EVs were isolated from the blood serum, characterized by their number, size distribution. Following unbiased screening for microRNA expression, differentially expressed candidates were measured by quantitative real-time PCR. Circulating DNA-myeloperoxidase complexes and apoptosis-related transcripts in peripheral blood neutrophils were quantified. We identified four differentially expressed microRNAs from EVs in granulomatosis with polyangiitis (GPA). MirRs-223-3p, 664a-3p, and 200b-3p were overexpressed and miR-769-5p suppressed in the disease. A distinction between GPA and healthy controls was the best for miR-223-3p, whereas miR-664a-3p discriminated between active vs. remission of GPA. Correct classification of the disease based on multivariate discriminant analysis was between 92% for acute phase and 85% for all study participants. Bioinformatics tools identified genes transcripts potentially targeted by the microRNAs belonging to pathways of focal adhesion, mTOR signaling and neutrophil extracellular traps formation. Two microRNAs positively correlating with the disease activity were involved in neutrophil extracellular traps formation and apoptosis inhibition. A comprehensive characteristics of microRNAs trafficked in bloodstream inside EVs correlates well with our understanding of the mechanisms of GPA and suggests the importance of EVs in progression of the disease.
Collapse
Affiliation(s)
- Marcin Surmiak
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str., 31-066 Kraków, Poland
| | | | - Joanna Kosałka-Węgiel
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str., 31-066 Kraków, Poland
| | - Stanisław Polański
- Division of Biochemical and Molecular Diagnostics, University Hospital, 8 Skawinska Str., 31-066 Kraków, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str., 31-066 Kraków, Poland
| |
Collapse
|
19
|
Xu S, Han S, Dai Y, Wang L, Zhang X, Ding Y. A Review of the Mechanism of Vascular Endothelial Injury in Immunoglobulin A Vasculitis. Front Physiol 2022; 13:833954. [PMID: 35370802 PMCID: PMC8966136 DOI: 10.3389/fphys.2022.833954] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022] Open
Abstract
Immunoglobulin A (IgA) vasculitis (IgAV), also known as Henoch-Schönlein purpura, is the most common form of childhood vasculitis. It is characterized by cutaneous hemorrhage, resulting from red blood cell leakage into the skin or mucosae, possibly caused by damage to small blood vessels. These acute symptoms usually disappear without treatment. Endothelial cells are distributed on the inner surfaces of blood vessels and lymphatic vessels, and have important functions in metabolism and endocrine function, as well as being the primary targets of external stimuli and endogenous immune activity. Injury to endothelial cells is a feature of IgA vasculitis. Endothelial cell damage may be related to the deposition of immune complexes, the activation of complement, inflammatory factors, and chemokines, oxidative stress, hemodynamics, and coagulation factors. Both epigenetic mechanisms and genetic diversity provide a genetic background for endothelial cell injury. Here, research on the role of endothelial cells in allergic IgA vasculitis is reviewed.
Collapse
Affiliation(s)
- Shanshan Xu
- Pediatric Kidney Disease Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Shanshan Han
- Pediatric Kidney Disease Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yanlin Dai
- Pediatric Kidney Disease Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Long Wang
- Pediatric Kidney Disease Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xia Zhang
- Pediatric Kidney Disease Center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ying Ding
- Pediatric Kidney Disease Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- *Correspondence: Ying Ding,
| |
Collapse
|
20
|
S100A8/A9 Is a Marker for the Release of Neutrophil Extracellular Traps and Induces Neutrophil Activation. Cells 2022; 11:cells11020236. [PMID: 35053354 PMCID: PMC8773660 DOI: 10.3390/cells11020236] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Neutrophils are the most abundant innate immune cells in the circulation and they are the first cells recruited to sites of infection or inflammation. Almost half of the intracellular protein content in neutrophils consists of S100A8 and S100A9, though there has been controversy about their actual localization. Once released extracellularly, these proteins are thought to act as damage-associated molecular patterns (DAMPs), though their mechanism of action is not well understood. These S100 proteins mainly form heterodimers (S100A8/A9, also known as calprotectin) and this heterocomplex is recognized as a useful biomarker for several inflammatory diseases. We observed that S100A8/A9 is highly present in the cytoplasmic fraction of neutrophils and is not part of the granule content. Furthermore, we found that S100A8/A9 was not released in parallel with granular content but upon the formation of neutrophil extracellular traps (NETs). Accordingly, neutrophils of patients with chronic granulomatous disease, who are deficient in phorbol 12-myristate 13-acetate (PMA)-induced NETosis, did not release S100A8/A9 upon PMA stimulation. Moreover, we purified S100A8/A9 from the cytoplasmic fraction of neutrophils and found that S100A8/A9 could induce neutrophil activation, including adhesion and CD11b upregulation, indicating that this DAMP might amplify neutrophil activation.
Collapse
|
21
|
Palamidas DA, Argyropoulou OD, Georgantzoglou N, Karatza E, Xingi E, Kapsogeorgou EK, Anagnostopoulos CD, Lazaris AC, Ritis K, Goules AV, Kambas K, Tzioufas AG. Neutrophil extracellular traps in giant cell arteritis biopsies: presentation, localization and co-expression with inflammatory cytokines. Rheumatology (Oxford) 2021; 61:1639-1644. [PMID: 34260696 DOI: 10.1093/rheumatology/keab505] [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: 02/15/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To explore the presence of neutrophil extracellular traps (NETs) in inflamed temporal artery biopsies (TABs) of patients with giant cell arteritis (GCA). METHODS Ten patients with GCA [5 with limited and 5 with associated generalized vascular involvement, as defined by 18F-fluorodeoxyglucose (FDG) positron-emission tomography with computed tomography (PET/CT)] and 8 with polymyalgia rheumatica (PMR) were studied. The presence, location, quantitation, and decoration of NETs with IL-6, IL-1β, and IL-17A were assessed in TABs at the time of disease diagnosis by tissue immunofluorescence and confocal microscopy. Paired serum levels of IL-6 and IL-17A were also evaluated in all patients. RESULTS All temporal artery biopsies from GCA, but not PMR patients, had NETs located mainly in the adventitia, adjacent to the vasa vasorum. NETs decorated with IL-6 were present in 8/10 TABs of GCA patients, of whom 5 were -PET/CT(+) and 3 PET/CT(-) patients. IL-17A(+) NETs were observed in all GCA patients. IL-1β(+)NETs were not detected in any GCA patient. No relation was found between serum IL-6 and IL-17A levels and NETs containing IL-6 and/or IL-17A. CONCLUSIONS NETs bearing pro-inflammatory cytokines are present in inflamed GCA-TABs. Future studies with a larger number of patients from different centers will show whether the findings regarding neutrophils/NETs in the TAB are consistent and disclose their clinical impact.
Collapse
Affiliation(s)
- Dimitris Anastasios Palamidas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Rheumatology Academic program, University of Athens, Athens, Greece
| | - Ourania D Argyropoulou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Rheumatology Academic program, University of Athens, Athens, Greece
| | - Natalia Georgantzoglou
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Elli Karatza
- Second Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Evangelia Xingi
- Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece
| | - Efstathia K Kapsogeorgou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Rheumatology Academic program, University of Athens, Athens, Greece
| | | | - Andreas C Lazaris
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Ritis
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Alexandroupolis, Greece.,Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Andreas V Goules
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Rheumatology Academic program, University of Athens, Athens, Greece
| | - Konstantinos Kambas
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Rheumatology Academic program, University of Athens, Athens, Greece
| |
Collapse
|