1
|
Yang X, Ren Y, Li X, Xia L, Wan J. MiR-146a Reduces Inflammation in Experimental Pancreatitis via the TRAF6-NF-κB Signaling Pathway in Mice. Immun Inflamm Dis 2025; 13:e70163. [PMID: 40018991 PMCID: PMC11868994 DOI: 10.1002/iid3.70163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND The initial inflammatory response plays a pivotal role in the development of acute pancreatitis. MiR-146a is believed to play a key role in negatively regulating inflammation and potentially contributes to anti-inflammatory activity in acute pancreatitis, though its mechanism remains largely unexplored. OBJECTIVES This study aimed to explore the effects of miR-146a on AP in mice and clarify its regulatory mechanisms in pancreatic inflammation and damage. METHODS Adult male BALB/C mice were used. Adeno-associated virus (AAV) vectors were used to modulate miR-146a expression in mice via tail vein injection. AP was induced by intraperitoneal injection of caerulein, caerulein + LPS, or l-arginine. Histological analysis, immunohistochemistry staining, immunofluorescence staining, measurements of amylase and lipase activities, and qRT-PCR were performed. RESULTS Overexpression of miR-146a reduced pancreatic damage and inflammation in caerulein-induced AP. It decreased serum amylase and lipase levels, mitigated pathological features such as interstitial edema and inflammatory cell infiltration in the pancreas and lung, and reduced neutrophil infiltration and proinflammatory cytokine expression. MiR-146a attenuated the activation of the NF-κB signaling pathway by inhibiting the degradation of IκBα and the expression of phosphorylated-p65 and reducing the nuclear translocation of NF-κB p65. Similar protective effects of miR-146a were observed in AP models induced by l-arginine and caerulein combined with LPS. CONCLUSIONS MiR-146a alleviates acute pancreatitis in mice by targeting TRAF6 and suppressing the activation of the NF-κB signaling pathway. These findings suggest that miR-146a could be a potential therapeutic target for AP.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Yuping Ren
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Xueyang Li
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Liang Xia
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Jianhua Wan
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| |
Collapse
|
2
|
Kundu D, Shin SY, Chilian WM, Dong F. The Potential of Mesenchymal Stem Cell-Derived Exosomes in Cardiac Repair. Int J Mol Sci 2024; 25:13494. [PMID: 39769256 PMCID: PMC11727646 DOI: 10.3390/ijms252413494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, and effectively repairing the heart following myocardial injuries remains a significant challenge. Research has increasingly shown that exosomes derived from mesenchymal stem cells (MSC-Exo) can ameliorate myocardial injuries and improve outcomes after such injuries. The therapeutic benefits of MSC-Exo are largely due to their capacity to deliver specific cargo, including microRNAs and proteins. MSC-Exo can modulate various signaling pathways and provide several beneficial effects, including cytoprotection, inflammation modulation, and angiogenesis promotion to help repair the damaged myocardium. In this review, we summarize the cardioprotective effects of MSC-Exo in myocardial injury, the underlying molecular mechanism involved in the process, and various approaches studied to enhance their efficacy based on recent findings.
Collapse
Affiliation(s)
| | | | | | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (D.K.); (S.Y.S.); (W.M.C.)
| |
Collapse
|
3
|
Martinod K, Denorme F, Meyers S, Crescente M, Van Bruggen S, Stroobants M, Siegel PM, Grandhi R, Glatz K, Witsch T. Involvement of peptidylarginine deiminase 4 in eosinophil extracellular trap formation and contribution to citrullinated histone signal in thrombi. J Thromb Haemost 2024; 22:1649-1659. [PMID: 38395360 DOI: 10.1016/j.jtha.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Extracellular traps formed by neutrophils (NETs) and eosinophils (EETs) have been described in coronary thrombi, contributing to thrombus stability. A key mechanism during NET formation is histone modification by the enzyme PAD4. Citrullinated histones, the product of PAD4 activity, are often attributed to neutrophils. Eosinophils also express high levels of PAD4. OBJECTIVES We aimed to explore the contribution of PAD4 to EET formation. METHODS We performed immunohistological analyses on thrombi, including a large, intact, and eosinophil-containing thrombus retrieved from the right coronary artery using an aspiration catheter and stroke thrombi from thrombectomy retrieval. We studied eosinophils for their capability to form PAD4-dependent EETs in response to strong ET-inducing agonists as well as activated platelets and bacteria. RESULTS Histopathology and immunofluorescence microscopy identified a coronary thrombus rich in platelets and neutrophils, with distinct areas containing von Willebrand factor and citrullinated histone H3 (H3Cit). Eosinophils were also identified in leukocyte-rich areas. The majority of the H3Cit+ signal colocalized with myeloperoxidase, but some colocalized with eosinophil peroxidase, indicating EETs. Eosinophils isolated from healthy volunteers produced H3Cit+ EETs, indicating an involvement of PAD4 activity. The selective PAD4 inhibitor GSK484 blocked this process, supporting PAD4 dependence of H3Cit+ EET release. Citrullinated histones were also present in EETs produced in response to live Staphylococci. However, limited evidence for EETs was found in mouse models of venous thrombosis or infective endocarditis. CONCLUSION As in NETosis, PAD4 can catalyze the formation of EETs. Inhibition of PAD4 decreases EET formation, supporting the future utility of PAD4 inhibitors as possible antithrombotic agents.
Collapse
Affiliation(s)
- Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Frederik Denorme
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Severien Meyers
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marilena Crescente
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Stijn Van Bruggen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Mathias Stroobants
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Patrick M Siegel
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ramesh Grandhi
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, Utah, USA
| | - Katharina Glatz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Thilo Witsch
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Xu X, Xu S, Zhang Y, Wang L, Yan C, Xu Z, Zhao Q, Qi X. Neutrophil extracellular traps formation may be involved in the association of propranolol with the development of portal vein thrombosis. Thromb Res 2024; 238:208-221. [PMID: 38733693 DOI: 10.1016/j.thromres.2024.04.030] [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: 10/19/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND & AIMS Nonselective β blockers (NSBBs) facilitate the development of portal vein thrombosis (PVT) in liver cirrhosis. Considering the potential effect of NSBBs on neutrophils and neutrophil extracellular traps (NETs), we speculated that NSBBs might promote the development of PVT by stimulating neutrophils to release NETs. MATERIALS AND METHODS Serum NETs biomarkers were measured, use of NSBBs was recorded, and PVT was evaluated in cirrhotic patients. Carbon tetrachloride and ferric chloride (FeCl3) were used to induce liver fibrosis and PVT in mice, respectively. After treatment with propranolol and DNase I, neutrophils in peripheral blood, colocalization and expression of NETs in PVT specimens, and NETs biomarkers in serum were measured. Ex vivo clots lysis analysis was performed and portal vein velocity and coagulation parameters were tested. RESULTS Serum MPO-DNA level was significantly higher in cirrhotic patients treated with NSBBs, and serum H3Cit and MPO-DNA levels were significantly higher in those with PVT. In fibrotic mice, following treatment with propranolol, DNase I significantly shortened the time of FeCl3-induced PVT formation, lowered the peripheral blood neutrophils labelled by CD11b/Ly6G, inhibited the positive staining of H3Cit and the expression of H3Cit and MPO proteins in PVT tissues, and reduced serum nucleosome level. Furthermore, the addition of DNase I to tissue plasminogen activator (tPA) significantly accelerated clots lysis as compared with tPA alone. Propranolol reduced portal vein velocity in fibrotic mice, but did not influence coagulation parameters. CONCLUSION Our study provides a clue to the potential impact of NETs formation on the association of NSBBs with the development of PVT.
Collapse
Affiliation(s)
- Xiangbo Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shixue Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yiyan Zhang
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
| | - Le Wang
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang, China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
5
|
Garley M, Nowak K, Jabłońska E. Neutrophil microRNAs. Biol Rev Camb Philos Soc 2024; 99:864-877. [PMID: 38148491 DOI: 10.1111/brv.13048] [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: 02/01/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
Neutrophils are considered 'first-line defence' cells as they can be rapidly recruited to the site of the immune response. As key components of non-specific immune mechanisms, neutrophils use phagocytosis, degranulation, and formation of neutrophil extracellular traps (NETs) to fight pathogens. Recently, immunoregulatory abilities of neutrophils associated with the secretion of several mediators, including cytokines and extracellular vesicles (EVs) containing, among other components, microRNAs (miRNAs), have also been reported. EVs are small structures released by cells into the extracellular space and are present in all body fluids. Microvesicles show the composition and status of the releasing cell, its physiological state, and pathological changes. Currently, EVs have gained immense scientific interest as they act as transporters of epigenetic information in intercellular communication. This review summarises findings from recent scientific reports that have evaluated the utility of miRNA molecules as biomarkers for effective diagnostics or even as start-points for new therapeutic strategies in neutrophil-mediated immune reactions. In addition, this review describes the current state of knowledge on miRNA molecules, which are endogenous regulators of gene expression besides being involved in the regulation of the immune response.
Collapse
Affiliation(s)
- Marzena Garley
- Department of Immunology, Medical University of Bialystok, Waszyngtona 15A, Bialystok, 15-269, Poland
| | - Karolina Nowak
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Ewa Jabłońska
- Department of Immunology, Medical University of Bialystok, Waszyngtona 15A, Bialystok, 15-269, Poland
| |
Collapse
|
6
|
Águila S, González-Conejero R, Martínez C. microRNAs and thrombo-inflammation: relationship in sight. Curr Opin Hematol 2024; 31:140-147. [PMID: 38277182 DOI: 10.1097/moh.0000000000000803] [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: 01/27/2024]
Abstract
PURPOSE OF REVIEW Thrombo-inflammation is a multifaceted pathologic process involving various cells such as platelets, neutrophils, and monocytes. In recent years, microRNAs have been consistently implicated as regulators of these cells. RECENT FINDINGS MicroRNAs play a regulatory role in several platelet receptors that have recently been identified as contributing to thrombo-inflammation and neutrophil extracellular trap (NET) formation. In addition, a growing body of evidence has shown that several intracellular and extracellular microRNAs directly promote NET formation. SUMMARY Targeting microRNAs is a promising therapeutic approach to control thrombosis in patients with both infectious and noninfectious inflammatory diseases. Future research efforts should focus on elucidating the specific roles of microRNAs in thrombo-inflammation and translating these findings into tangible benefits for patients.
Collapse
Affiliation(s)
- Sonia Águila
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, UCAM
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, Murcia, Spain
| | - Rocío González-Conejero
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, UCAM
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, Murcia, Spain
| | - Constantino Martínez
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, UCAM
- Department of Hematology, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, Murcia, Spain
| |
Collapse
|
7
|
Vardas EP, Theofilis P, Oikonomou E, Vardas PE, Tousoulis D. MicroRNAs in Atrial Fibrillation: Mechanisms, Vascular Implications, and Therapeutic Potential. Biomedicines 2024; 12:811. [PMID: 38672166 PMCID: PMC11048414 DOI: 10.3390/biomedicines12040811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Atrial fibrillation (AFib), the most prevalent arrhythmia in clinical practice, presents a growing global health concern, particularly with the aging population, as it is associated with devastating complications and an impaired quality of life. Its pathophysiology is multifactorial, including the pathways of fibrosis, inflammation, and oxidative stress. MicroRNAs (miRNAs), small non-coding RNA molecules, have emerged as substantial contributors in AFib pathophysiology, by affecting those pathways. In this review, we explore the intricate relationship between miRNAs and the aforementioned aspects of AFib, shedding light on the molecular pathways as well as the potential diagnostic applications. Recent evidence also suggests a possible role of miRNA therapeutics in maintenance of sinus rhythm via the antagonism of miR-1 and miR-328, or the pharmacological upregulation of miR-27b and miR-223-3p. Unraveling the crosstalk between specific miRNA profiles and genetic predispositions may pave the way for personalized therapeutic approaches, setting the tone for precision medicine in atrial fibrillation.
Collapse
Affiliation(s)
- Emmanouil P. Vardas
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
- Department of Cardiology, General Hospital of Athens “G. Gennimatas”, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
| | - Evangelos Oikonomou
- 3rd Cardiology Department, Sotiria Regional Hospital for Chest Diseases, University of Athens Medical School, 11527 Athens, Greece;
| | - Panos E. Vardas
- Biomedical Research Foundation Academy of Athens, Heart Sector, Hygeia Hospitals Group, Attica, 15123 Athens, Greece;
| | - Dimitris Tousoulis
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
| |
Collapse
|
8
|
Jiang W, Jia W, Dong C. Under the dual effect of inflammation and pulmonary fibrosis, CTD-ILD patients possess a greater susceptibility to VTE. Thromb J 2024; 22:34. [PMID: 38576023 PMCID: PMC10993540 DOI: 10.1186/s12959-024-00599-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
As an autoimmune disease, the persistent systemic inflammatory response associated with connective tissue disease (CTD) is involved in the development of venous thromboembolism (VTE). However, clinical data showed that the risk of VTE in patients differed between subtypes of CTD, suggesting that different subtypes may have independent mechanisms to promote the development of VTE, but the specific mechanism lacks sufficient research at present. The development of pulmonary fibrosis also contributes to the development of VTE, and therefore, patients with CTD-associated interstitial lung disease (CTD-ILD) may be at higher risk of VTE than patients with CTD alone or patients with ILD alone. In addition, the activation of the coagulation cascade response will drive further progression of the patient's pre-existing pulmonary fibrosis, which will continue to increase the patient's risk of VTE and adversely affect prognosis. Currently, the treatment for CTD-ILD is mainly immunosuppressive and antirheumatic therapy, such as the use of glucocorticoids and janus kinase-inhibitors (JAKis), but, paradoxically, these drugs are also involved in the formation of patients' coagulation tendency, making the clinical treatment of CTD-ILD patients with a higher risk of developing VTE challenging. In this article, we review the potential risk factors and related mechanisms for the development of VTE in CTD-ILD patients to provide a reference for clinical treatment and prevention.
Collapse
Affiliation(s)
- Wenli Jiang
- Department of Pulmonary and Critical Care Medicine, Second Hospital, Jilin University, 130041, Changchun, China
| | - Wenhui Jia
- Department of Pulmonary and Critical Care Medicine, Second Hospital, Jilin University, 130041, Changchun, China
| | - Chunling Dong
- Department of Pulmonary and Critical Care Medicine, Second Hospital, Jilin University, 130041, Changchun, China.
| |
Collapse
|
9
|
Manso J, Censi S, Clausi C, Piva I, Zhu YH, Mondin A, Pedron MC, Barollo S, Bertazza L, Midena G, Parrozzani R, Mian C. Circulating miR-146a predicts glucocorticoid response in thyroid eye disease. Eur Thyroid J 2023; 12:e230083. [PMID: 37606076 PMCID: PMC10563606 DOI: 10.1530/etj-23-0083] [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: 04/17/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023] Open
Abstract
Objective Thyroid eye disease (TED) is an immune-mediated disorder of the eye. Intravenous glucocorticoid (GC) is the first-line treatment for patients with active moderate-to-severe TED. However, the response rate is between 50% and 80%. There are still no simple and reliable markers of responsiveness to GC therapy. We aimed to explore the possible role of miR-146a and miR-21 as predictors of responsiveness to GC treatment in TED. Methods We carried out a prospective longitudinal study on 30 consecutive adult patients with active moderate-to-severe TED and eligible for GC therapy. All patients received the standard GC treatment with methylprednisolone i.v. In cases of progressive worsening of Gorman Score for diplopia or with duction restriction <30° in at least two consecutive controls, patients also underwent orbital radiotherapy. Response to GC treatment was defined as a decrease of two or more points in the clinical activity score (CAS) or CAS <4/10 at 24 weeks. Circulating miRNAs were extracted from patients' serum and quantified by real-time PCR. Results Twenty-three (77%) patients responded to GC. Thyroid surgery, higher CAS, greater proptosis and higher pre-treatment circulating levels of miR-146a emerged as predictive factors of responsiveness to GC. A ROC analysis revealed that miR-146a could predict responsiveness to GC with a positive predictive value of 100%. Conclusion This is the first study investigating the role of pre-treatment circulating miR-21 and miR-146a to predict responsiveness to GC in TED. miR-146a emerged as a simple, objective, new marker of GC sensitivity that could be used to avoid ineffective administration of GC therapy to TED patients.
Collapse
Affiliation(s)
- Jacopo Manso
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
- Department of Women’s and Children’s Health, Pediatric Endocrinology Unit, Padua University Hospital, Padua, Italy
| | - Simona Censi
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Cristina Clausi
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Ilaria Piva
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Yi Hang Zhu
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Alberto Mondin
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Maria Chiara Pedron
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Susi Barollo
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Loris Bertazza
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| | - Giulia Midena
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Caterina Mian
- Department of Medicine (DIMED), Endocrinology Unit, University of Padua, Padua, Italy
| |
Collapse
|
10
|
Gilyazova I, Asadullina D, Kagirova E, Sikka R, Mustafin A, Ivanova E, Bakhtiyarova K, Gilyazova G, Gupta S, Khusnutdinova E, Gupta H, Pavlov V. MiRNA-146a-A Key Player in Immunity and Diseases. Int J Mol Sci 2023; 24:12767. [PMID: 37628949 PMCID: PMC10454149 DOI: 10.3390/ijms241612767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
miRNA-146a, a single-stranded, non-coding RNA molecule, has emerged as a valuable diagnostic and prognostic biomarker for numerous pathological conditions. Its primary function lies in regulating inflammatory processes, haemopoiesis, allergic responses, and other key aspects of the innate immune system. Several studies have indicated that polymorphisms in miRNA-146a can influence the pathogenesis of various human diseases, including autoimmune disorders and cancer. One of the key mechanisms by which miRNA-146a exerts its effects is by controlling the expression of certain proteins involved in critical pathways. It can modulate the activity of interleukin-1 receptor-associated kinase, IRAK1, IRAK2 adaptor proteins, and tumour necrosis factor (TNF) targeting protein receptor 6, which is a regulator of the TNF signalling pathway. In addition, miRNA-146a affects gene expression through multiple signalling pathways, such as TNF, NF-κB and MEK-1/2, and JNK-1/2. Studies have been carried out to determine the effect of miRNA-146a on cancer pathogenesis, revealing its involvement in the synthesis of stem cells, which contributes to tumourigenesis. In this review, we focus on recent discoveries that highlight the significant role played by miRNA-146a in regulating various defence mechanisms and oncogenesis. The aim of this review article is to systematically examine miRNA-146a's impact on the control of signalling pathways involved in oncopathology, immune system development, and the corresponding response to therapy.
Collapse
Affiliation(s)
- Irina Gilyazova
- Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Institute of Biochemistry and Genetics, 450054 Ufa, Russia (E.K.)
| | - Dilara Asadullina
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Evelina Kagirova
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Ruhi Sikka
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura 281406, India
| | - Artur Mustafin
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Elizaveta Ivanova
- Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Institute of Biochemistry and Genetics, 450054 Ufa, Russia (E.K.)
| | - Ksenia Bakhtiyarova
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Gulshat Gilyazova
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Saurabh Gupta
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura 281406, India
| | - Elza Khusnutdinova
- Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, Institute of Biochemistry and Genetics, 450054 Ufa, Russia (E.K.)
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| | - Himanshu Gupta
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura 281406, India
| | - Valentin Pavlov
- Institute of Urology and Clinical Oncology, Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, 450008 Ufa, Russia (A.M.); (G.G.)
| |
Collapse
|
11
|
Kanduła Z, Janowski M, Więckowska B, Paczkowska E, Lewandowski K. JAK2V617F variant allele frequency, non-driver mutations, single-nucleotide variants and polycythemia vera outcome. J Cancer Res Clin Oncol 2023; 149:4789-4803. [PMID: 36242602 PMCID: PMC10349754 DOI: 10.1007/s00432-022-04327-0] [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: 06/14/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Despite comparatively favourable prognosis in polycythemia vera (PV) patients (pts), the overall survival is shorter compared to the age-matched general population. The aim of the study was to evaluate the impact of chosen laboratory and genetic factors on the individual disease outcome, i.e. risk of thrombosis, myelofibrosis/blastic transformation and death. MATERIALS AND METHODS The study group consisted of 151 pts and 57 healthy donors (HD). RESULTS JAK2V617F mutation was found in 96.7% (146/151) of the studied pts. JAK2 exon 12 mutations were identified in 2 individuals. The coexistence of JAK2V617F and JAK2 exon 12 mutation was confirmed in 2 other pts. In one case, neither JAK2V617F nor JAK2 exon 12 mutation was found. The presence of ten different non-driver mutations (ASXL1, SRSF2, U2AF1, IDH2) in eight of the analyzed pts (5.3%) was confirmed. The overall frequency of thrombotic events (TE) in the studied PV group was 23.8% (36/151). In patients with TE, median platelet count was lower than in pts without TE. Thrombotic risk did not depend on JAK2 rs12343867, TERT rs2736100, OBFC1 rs9420907 SNV, however, we found a novel strong tendency towards statistical significance between the CC genotype miR-146a rs2431697 and thrombosis. The disease progression to fibrotic phase was confirmed in 9% of the pts. Fibrotic transformation in PV pts was affected mainly by JAK2V617F variant allele frequency (VAF) and the presence of coexisting non-driver variants. The high JAK2V617F VAF and elevated white blood cell (WBC) count at the time of diagnosis were associated with an increased risk of death. CONCLUSION Therefore, in our opinion, complex, laboratory and genetic PV pts evaluation at the time of diagnosis should be incorporated into a new prognostic scoring system to more precisely define the PV prognosis and to optimize the therapeutic decision-making process.
Collapse
Affiliation(s)
- Zuzanna Kanduła
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| | - Michał Janowski
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Barbara Więckowska
- Department of Computer Science and Statistics, Poznań University of Medical Sciences, Poznań, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Lewandowski
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
12
|
Spinetti G, Mutoli M, Greco S, Riccio F, Ben-Aicha S, Kenneweg F, Jusic A, de Gonzalo-Calvo D, Nossent AY, Novella S, Kararigas G, Thum T, Emanueli C, Devaux Y, Martelli F. Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems. Cardiovasc Diabetol 2023; 22:122. [PMID: 37226245 PMCID: PMC10206598 DOI: 10.1186/s12933-023-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by high levels of blood glucose caused by insulin defect or impairment, is a major risk factor for cardiovascular diseases and related mortality. Patients with diabetes experience a state of chronic or intermittent hyperglycemia resulting in damage to the vasculature, leading to micro- and macro-vascular diseases. These conditions are associated with low-grade chronic inflammation and accelerated atherosclerosis. Several classes of leukocytes have been implicated in diabetic cardiovascular impairment. Although the molecular pathways through which diabetes elicits an inflammatory response have attracted significant attention, how they contribute to altering cardiovascular homeostasis is still incompletely understood. In this respect, non-coding RNAs (ncRNAs) are a still largely under-investigated class of transcripts that may play a fundamental role. This review article gathers the current knowledge on the function of ncRNAs in the crosstalk between immune and cardiovascular cells in the context of diabetic complications, highlighting the influence of biological sex in such mechanisms and exploring the potential role of ncRNAs as biomarkers and targets for treatments. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.
Collapse
Affiliation(s)
- Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Martina Mutoli
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Federica Riccio
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Soumaya Ben-Aicha
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susana Novella
- Department of Physiology, University of Valencia - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Georgios Kararigas
- Department of Physiology, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Costanza Emanueli
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy.
| |
Collapse
|
13
|
Zapata-Martínez L, Águila S, de los Reyes-García AM, Carrillo-Tornel S, Lozano ML, González-Conejero R, Martínez C. Inflammatory microRNAs in cardiovascular pathology: another brick in the wall. Front Immunol 2023; 14:1196104. [PMID: 37275892 PMCID: PMC10233054 DOI: 10.3389/fimmu.2023.1196104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
The regulatory role of microRNAs (miRNAs) is mainly mediated by their effect on protein expression and is recognized in a multitude of pathophysiological processes. In recent decades, accumulating evidence has interest in these factors as modulatory elements of cardiovascular pathophysiology. Furthermore, additional biological processes have been identified as new components of cardiovascular disease etiology. In particular, inflammation is now considered an important cardiovascular risk factor. Thus, in the present review, we will focus on the role of a subset of miRNAs called inflamma-miRs that may regulate inflammatory status in the development of cardiovascular pathology. According to published data, the most representative candidates that play functional roles in thromboinflammation are miR-21, miR-33, miR-34a, miR-146a, miR-155, and miR-223. We will describe the functions of these miRNAs in several cardiovascular pathologies in depth, with specific emphasis on the molecular mechanisms related to atherogenesis. We will also discuss the latest findings on the role of miRNAs as regulators of neutrophil extracellular traps and their impact on cardiovascular diseases. Overall, the data suggest that the use of miRNAs as therapeutic tools or biomarkers may improve the diagnosis or prognosis of adverse cardiovascular events in inflammatory diseases. Thus, targeting or increasing the levels of adequate inflamma-miRs at different stages of disease could help mitigate or avoid the development of cardiovascular morbidities.
Collapse
|
14
|
Maneta E, Aivalioti E, Tual-Chalot S, Emini Veseli B, Gatsiou A, Stamatelopoulos K, Stellos K. Endothelial dysfunction and immunothrombosis in sepsis. Front Immunol 2023; 14:1144229. [PMID: 37081895 PMCID: PMC10110956 DOI: 10.3389/fimmu.2023.1144229] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Sepsis is a life-threatening clinical syndrome characterized by multiorgan dysfunction caused by a dysregulated or over-reactive host response to infection. During sepsis, the coagulation cascade is triggered by activated cells of the innate immune system, such as neutrophils and monocytes, resulting in clot formation mainly in the microcirculation, a process known as immunothrombosis. Although this process aims to protect the host through inhibition of the pathogen’s dissemination and survival, endothelial dysfunction and microthrombotic complications can rapidly lead to multiple organ dysfunction. The development of treatments targeting endothelial innate immune responses and immunothrombosis could be of great significance for reducing morbidity and mortality in patients with sepsis. Medications modifying cell-specific immune responses or inhibiting platelet–endothelial interaction or platelet activation have been proposed. Herein, we discuss the underlying mechanisms of organ-specific endothelial dysfunction and immunothrombosis in sepsis and its complications, while highlighting the recent advances in the development of new therapeutic approaches aiming at improving the short- or long-term prognosis in sepsis.
Collapse
Affiliation(s)
- Eleni Maneta
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
- *Correspondence: Eleni Maneta, ; Konstantinos Stellos, ;
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Besa Emini Veseli
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
- *Correspondence: Eleni Maneta, ; Konstantinos Stellos, ;
| |
Collapse
|
15
|
Meregildo-Rodriguez ED, Asmat-Rubio MG, Rojas-Benites MJ, Vásquez-Tirado GA. Acute Coronary Syndrome, Stroke, and Mortality after Community-Acquired Pneumonia: Systematic Review and Meta-Analysis. J Clin Med 2023; 12:2577. [PMID: 37048661 PMCID: PMC10095577 DOI: 10.3390/jcm12072577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/31/2023] Open
Abstract
One-third of adult inpatients with community-acquired pneumonia (CAP) develop acute coronary syndrome (ACS), stroke, heart failure (HF), arrhythmias, or die. The evidence linking CAP to cardiovascular disease (CVD) events is contradictory. We aimed to systematically review the role of CAP as a CVD risk factor. We registered the protocol (CRD42022352910) and searched for six databases from inception to 31 December 2022. We included 13 observational studies, 276,109 participants, 18,298 first ACS events, 12,421 first stroke events, 119 arrhythmic events, 75 episodes of new onset or worsening HF, 3379 deaths, and 218 incident CVD events. CAP increased the odds of ACS (OR 3.02; 95% CI 1.88-4.86), stroke (OR 2.88; 95% CI 2.09-3.96), mortality (OR 3.22; 95% CI 2.42-4.27), and all CVD events (OR 3.37; 95% CI 2.51-4.53). Heterogeneity was significant (I2 = 97%, p < 0.001). Subgroup analysis found differences according to the continent of origin of the study, the follow-up length, and the sample size (I2 > 40.0%, p < 0.10). CAP is a significant risk factor for all major CVD events including ACS, stroke, and mortality. However, these findings should be taken with caution due to the substantial heterogeneity and the possible publication bias.
Collapse
|
16
|
Tomás-Pérez S, Oto J, Aghababyan C, Herranz R, Cuadros-Lozano A, González-Cantó E, Mc Cormack B, Arrés J, Castaño M, Cana F, Martínez-Fernández L, Santonja N, Ramírez R, Herreros-Pomares A, Cañete-Mota S, Llueca A, Marí-Alexandre J, Medina P, Gilabert-Estellés J. Increased levels of NETosis biomarkers in high-grade serous ovarian cancer patients' biofluids: Potential role in disease diagnosis and management. Front Immunol 2023; 14:1111344. [PMID: 36817483 PMCID: PMC9936152 DOI: 10.3389/fimmu.2023.1111344] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction High-grade serous ovarian cancer (HGSOC) is the second most frequent gynecological malignancy but the most lethal, partially due to the spread of the disease through the peritoneal cavity. Recent evidence has shown that, apart from their role in immune defense through phagocytosis and degranulation, neutrophils are able to participate in cancer progression through the release of neutrophil extracellular traps (NETs) in a process called NETosis. NETs are composed of DNA, histones, calprotectin, myeloperoxidase (MPO) and elastase and the NETosis process has been proposed as a pre-requisite for the establishment of omental metastases in early stages of HGSOC. Nevertheless, its role in advanced stages remains to be elucidated. Therefore, our principal aim is to characterize a NETosis biomarker profile in biofluids from patients with advanced HGSOC and control women. Methods Specifically, five biomarkers of NETosis (cell-free DNA (cfDNA), nucleosomes, citrullinated histone 3 (citH3), calprotectin and MPO) were quantified in plasma and peritoneal fluid (PF) samples from patients (n=45) and control women (n=40). Results Our results showed that HGSOC patients presented a higher concentration of cfDNA, citH3 and calprotectin in plasma and of all five NETosis biomarkers in PF than control women. Moreover, these biomarkers showed a strong ability to differentiate the two clinical groups. Interestingly, neoadjuvant treatment (NT) seemed to reduce NETosis biomarkers mainly systemically (plasma) compared to the tumor environment (PF). Discussion In conclusion, NETosis biomarkers are present in the tumor environment of patients with advanced HGSOC, which might contribute to the progression of the disease. Besides, plasma cfDNA and calprotectin could represent minimally invasive surrogate biomarkers for HGSOC. Finally, NT modifies NETosis biomarkers levels mainly at the systemic level.
Collapse
Affiliation(s)
- Sarai Tomás-Pérez
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain
| | - Julia Oto
- Hemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Cristina Aghababyan
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain,Department of Obstetrics and Gynecology, General University Hospital of Valencia Consortium, Valencia, Spain
| | - Raquel Herranz
- Hemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Aitor Cuadros-Lozano
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain,Department of Obstetrics and Gynecology, General University Hospital of Valencia Consortium, Valencia, Spain
| | - Eva González-Cantó
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain
| | - Bárbara Mc Cormack
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain
| | - Judith Arrés
- Department of Statistics and Operational Research, University of Valencia, Valencia, Spain
| | - María Castaño
- Hemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Fernando Cana
- Hemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Laura Martínez-Fernández
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain,Department of Obstetrics and Gynecology, General University Hospital of Valencia Consortium, Valencia, Spain
| | - Núria Santonja
- Department of Pathology, General University Hospital of Valencia Consortium, Valencia, Spain
| | - Rocío Ramírez
- Department of Medical Oncology, General University Hospital of Valencia Consortium, Valencia, Spain
| | - Alejandro Herreros-Pomares
- Department of Biotechnology, Polytechnic University of Valencia, Valencia, Spain,Cancer Biomedical Research Network Center, CIBERONC, Madrid, Spain
| | - Sarai Cañete-Mota
- Department of Obstetrics and Gynecology, General University Hospital of Castellon, Castellón, Spain
| | - Antoni Llueca
- Department of Obstetrics and Gynecology, General University Hospital of Castellon, Castellón, Spain,Multidisciplinary Unit of Abdominal Pelvic Oncology Surgery (MUAPOS), General University Hospital of Castellon, Castellón, Spain,Department of Medicine, University Jaume I, Castellón, Spain
| | - Josep Marí-Alexandre
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain,Department of Pathology, General University Hospital of Valencia Consortium, Valencia, Spain,*Correspondence: Josep Marí-Alexandre, ; Pilar Medina,
| | - Pilar Medina
- Hemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain,*Correspondence: Josep Marí-Alexandre, ; Pilar Medina,
| | - Juan Gilabert-Estellés
- Research Laboratory in Biomarkers in Reproduction, Obstetrics and Gynecology, Research Foundation of the General University Hospital of Valencia, Valencia, Spain,Department of Obstetrics and Gynecology, General University Hospital of Valencia Consortium, Valencia, Spain,Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| |
Collapse
|
17
|
Bioinformatic Analyses of Peripheral Blood Transcriptome Identify Altered Neutrophil-Related Pathway and Different Transcriptomic Profiles for Acute Pancreatitis in Patients with and without Chylomicronemia Syndrome. Biomolecules 2023; 13:biom13020284. [PMID: 36830652 PMCID: PMC9953624 DOI: 10.3390/biom13020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Acute pancreatitis (AP) is a serious inflammatory condition of the pancreas that can be associated with chylomicronemia syndrome (CS). Currently, no study has explored the differences between non-CS-associated AP and CS-associated AP in terms of gene expression. Transcriptomic profiles of blood samples from patients with AP were retrieved from GSE194331 (non-CS-associated) and GSE149607 (CS-associated). GSE31568 was used to examine the linkage between non-CS-associated AP and the expression of micro RNAs (miRNAs). Differentially expressed genes (DEGs) were identified, a gene regulatory network was constructed, and hub genes were defined. Subsequently, single-sample gene set enrichment analysis (ssGSEA) scores of hub genes were calculated to represent their regulatory-level activity. A total of 1851 shared DEGs were identified between non-CS-associated and CS-associated AP. Neutrophils were significantly enriched in both conditions. In non-CS-associated AP, miRNAs including hsa-miR-21, hsa-miR-146a, and hsa-miR-106a demonstrated a lower expression level as compared with the healthy control. Furthermore, the expression patterns and regulatory activities were largely opposite between non-CS-associated and CS-associated AP, with significantly lower estimated neutrophils in the latter case. In summary, we found that the regulation of neutrophils was altered in AP. There was a different gene expression pattern and lower estimated neutrophil infiltration in CS-associated AP. Whether these findings are clinically significant requires further investigation.
Collapse
|
18
|
Liang Y, Fang D, Gao X, Deng X, Chen N, Wu J, Zeng M, Luo M. Circulating microRNAs as emerging regulators of COVID-19. Theranostics 2023; 13:125-147. [PMID: 36593971 PMCID: PMC9800721 DOI: 10.7150/thno.78164] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/06/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), an infectious disease caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global pandemic that has high incidence rates, spreads rapidly, and has caused more than 6.5 million deaths globally to date. Currently, several drugs have been used in the clinical treatment of COVID-19, including antivirals (e.g., molnupiravir, baricitinib, and remdesivir), monoclonal antibodies (e.g., etesevimab and tocilizumab), protease inhibitors (e.g., paxlovid), and glucocorticoids (e.g., dexamethasone). Increasing evidence suggests that circulating microRNAs (miRNAs) are important regulators of viral infection and antiviral immune responses, including the biological processes involved in regulating COVID-19 infection and subsequent complications. During viral infection, both viral genes and host cytokines regulate transcriptional and posttranscriptional steps affecting viral replication. Virus-encoded miRNAs are a component of the immune evasion repertoire and function by directly targeting immune functions. Moreover, several host circulating miRNAs can contribute to viral immune escape and play an antiviral role by not only promoting nonstructural protein (nsp) 10 expression in SARS coronavirus, but among others inhibiting NOD-like receptor pyrin domain-containing (NLRP) 3 and IL-1β transcription. Consequently, understanding the expression and mechanism of action of circulating miRNAs during SARS-CoV-2 infection will provide unexpected insights into circulating miRNA-based studies. In this review, we examined the recent progress of circulating miRNAs in the regulation of severe inflammatory response, immune dysfunction, and thrombosis caused by SARS-CoV-2 infection, discussed the mechanisms of action, and highlighted the therapeutic challenges involving miRNA and future research directions in the treatment of COVID-19.
Collapse
Affiliation(s)
- Yu Liang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- College of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaojun Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Deng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ni Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- College of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
19
|
de Los Reyes-García AM, Zapata-Martínez L, Águila S, Lozano ML, Martínez C, González-Conejero R. microRNAs as biomarkers of risk of major adverse cardiovascular events in atrial fibrillation. Front Cardiovasc Med 2023; 10:1135127. [PMID: 36895835 PMCID: PMC9988920 DOI: 10.3389/fcvm.2023.1135127] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Atrial fibrillation is a complex and multifactorial disease. Although prophylactic anticoagulation has great benefits in avoiding comorbidities, adverse cardiovascular events still occur and thus in recent decades, many resources have been invested in the identification of useful markers in the prevention of the risk of MACE in these patients. As such, microRNAs, that are small non-coding RNAs whose function is to regulate gene expression post-transcriptionally, have a relevant role in the development of MACE. miRNAs, have been investigated for many years as potential non-invasive biomarkers of several diseases. Different studies have shown their utility in the diagnosis and prognosis of cardiovascular diseases. In particular, some studies have associated the presence of certain miRNAs in plasma with the development of MACE in AF. Despite these results, there are still many efforts to be done to allow the clinical use of miRNAs. The lack of standardization concerning the methodology in purifying and detecting miRNAs, still provides contradictory results. miRNAs also have a functional impact in MACE in AF through the dysregulation of immunothrombosis. Indeed, miRNAs may be a link between MACE and inflammation, through the regulation of neutrophil extracellular traps that are a key element in the establishment and evolution of thrombotic events. The use of miRNAs as therapy against thromboinflammatory processes should also be a future approach to avoid the occurrence of MACE in atrial fibrillation.
Collapse
Affiliation(s)
- Ascensión M de Los Reyes-García
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Laura Zapata-Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Sonia Águila
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - María L Lozano
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Constantino Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Rocío González-Conejero
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| |
Collapse
|
20
|
Song Y, Zhang L, Huang Y. Differential Expression of Peripheral Circulating MicroRNA-146a Between Patients with Atherosclerotic Vulnerable Plaque and Stable Plaque. Int Heart J 2023; 64:847-855. [PMID: 37778988 DOI: 10.1536/ihj.23-006] [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] [Indexed: 10/03/2023]
Abstract
Atherosclerotic plaque rupture and subsequent cardiovascular complications threaten the population's health worldwide. The polymorphism of miR-146a rs2910164 was significantly associated with the risk of vulnerable plaques. However, it remains unclear whether the circulating miR-146a is differentially expressed in stable and vulnerable plaques and thus, serves as a potential biomarker.This study aims to analyze the differential expression of circulating miR-146a between patients with stable and vulnerable plaques to explore the potential molecular mechanisms.Public databases were searched from their inception up to November 2022. A study reporting the specific circulating miR-146a levels between patients with stable and vulnerable plaques was included. The study quality was assessed using the modified genetic 8-stars Newcastle-Ottawa scale. The differential expression levels of miR-146a were evaluated using the standardized mean difference (SMD).Eight studies with 978 patients were included and analyzed. The results showed that miR-146a expression levels were significantly higher in the vulnerable plaque population than in the stable plaque population (SMD: 1.91; 95% confidence interval: 1.35, 2.47; P < 0.01). A similar statistical significance was found in subgroup analyses regarding sample source, disease type, and vulnerable plaque characteristics. Sensitivity analysis suggested the robustness of the results. Analysis of downstream genes suggested that miR-146a-targeted regulation of ACTN4, SARM1, and ULK2 may affect intraplaque hemorrhage.Patients with vulnerable plaque have higher circulating miR-146a levels than those with stable plaque. However, based on the limitations of this study, high-quality studies are still needed to confirm the results.
Collapse
Affiliation(s)
- Yenwen Song
- Department of Emergency, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine
| | - Lei Zhang
- Department of Emergency, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine
| | - Ye Huang
- Department of Emergency, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine
| |
Collapse
|
21
|
Gaytán-Pacheco N, Ibáñez-Salazar A, Herrera-Van Oostdam AS, Oropeza-Valdez JJ, Magaña-Aquino M, Adrián López J, Monárrez-Espino J, López-Hernández Y. miR-146a, miR-221, and miR-155 are Involved in Inflammatory Immune Response in Severe COVID-19 Patients. Diagnostics (Basel) 2022; 13:133. [PMID: 36611425 PMCID: PMC9818442 DOI: 10.3390/diagnostics13010133] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
COVID-19 infection triggered a global public health crisis during the 2020-2022 period, and it is still evolving. This highly transmissible respiratory disease can cause mild symptoms up to severe pneumonia with potentially fatal respiratory failure. In this cross-sectional study, 41 PCR-positive patients for SARS-CoV-2 and 42 healthy controls were recruited during the first wave of the pandemic in Mexico. The plasmatic expression of five circulating miRNAs involved in inflammatory and pathological host immune responses was assessed using RT-qPCR (Reverse Transcription quantitative Polymerase Chain Reaction). Compared with controls, a significant upregulation of miR-146a, miR-155, and miR-221 was observed; miR-146a had a positive correlation with absolute neutrophil count and levels of brain natriuretic propeptide (proBNP), and miR-221 had a positive correlation with ferritin and a negative correlation with total cholesterol. We found here that CDKN1B gen is a shared target of miR-146a, miR-221-3p, and miR-155-5p, paving the way for therapeutic interventions in severe COVID-19 patients. The ROC curve built with adjusted variables (miR-146a, miR-221-3p, miR-155-5p, age, and male sex) to differentiate individuals with severe COVID-19 showed an AUC of 0.95. The dysregulation of circulating miRNAs provides new insights into the underlying immunological mechanisms, and their possible use as biomarkers to discriminate against patients with severe COVID-19. Functional analysis showed that most enriched pathways were significantly associated with processes related to cell proliferation and immune responses (innate and adaptive). Twelve of the predicted gene targets have been validated in plasma/serum, reflecting their potential use as predictive prognosis biomarkers.
Collapse
Affiliation(s)
- Noemí Gaytán-Pacheco
- Clinical Analysis Laboratory UAZ-Siglo-XXI, Academic Unit of Chemical Sciences, Autonomous University of Zacatecas, Zacatecas 98000, Mexico
| | - Alejandro Ibáñez-Salazar
- Clinical Analysis Laboratory UAZ-Siglo-XXI, Academic Unit of Chemical Sciences, Autonomous University of Zacatecas, Zacatecas 98000, Mexico
| | | | - Juan José Oropeza-Valdez
- Metabolomics and Proteomics Laboratory, Academic Unit of Biological Sciences, Autonomous University of Zacatecas, Zacatecas 98600, Mexico
| | | | - Jesús Adrián López
- MicroRNAs and Cancer Laboratory, Academic Unit of Biological Sciences, Autonomous University of Zacatecas, Zacatecas 98000, Mexico
| | - Joel Monárrez-Espino
- Department of Health Research, Christus Muguerza del Parque Hospital Chihuahua, University of Monterrey, San Pedro Garza García 66238, Mexico
| | - Yamilé López-Hernández
- CONACyT-Metabolomics and Proteomics Laboratory, Autonomous University of Zacatecas, Zacatecas 98000, Mexico
| |
Collapse
|
22
|
Bifunctional fusion protein targeting both FXIIa and FXIa displays potent anticoagulation effects. Life Sci 2022; 309:121021. [DOI: 10.1016/j.lfs.2022.121021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/21/2022]
|
23
|
Dhuppar S, Murugaiyan G. miRNA effects on gut homeostasis: therapeutic implications for inflammatory bowel disease. Trends Immunol 2022; 43:917-931. [PMID: 36220689 PMCID: PMC9617792 DOI: 10.1016/j.it.2022.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 01/12/2023]
Abstract
Inflammatory bowel disease (IBD) spans a range of chronic conditions affecting the gastrointestinal (GI) tract, which are marked by intermittent flare-ups and remissions. IBD results from microbial dysbiosis or a defective mucosal barrier in the gut that triggers an inappropriate immune response in a genetically susceptible person, altering the immune-microbiome axis. In this review, we discuss the regulatory roles of miRNAs, small noncoding RNAs with gene regulatory functions, in the stability and maintenance of the gut immune-microbiome axis, and detail the challenges and recent advances in the use of miRNAs as putative therapeutic agents for treating IBD.
Collapse
Affiliation(s)
- Shivnarayan Dhuppar
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Current address: Centre for Business Innovation, The Indian School of Business, Hyderabad 500111, India
| | - Gopal Murugaiyan
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Xu X, Wu Y, Xu S, Yin Y, Ageno W, De Stefano V, Zhao Q, Qi X. Clinical significance of neutrophil extracellular traps biomarkers in thrombosis. Thromb J 2022; 20:63. [PMID: 36224604 PMCID: PMC9555260 DOI: 10.1186/s12959-022-00421-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Neutrophil extracellular traps (NETs) may be associated with the development of thrombosis. Experimental studies have confirmed the presence of NETs in thrombi specimens and potential role of NETs in the mechanisms of thrombosis. Clinical studies also have demonstrated significant changes in the levels of serum or plasma NETs biomarkers, such as citrullinated histones, myeloperoxidase, neutrophil elastase, nucleosomes, DNA, and their complexes in patients with thrombosis. This paper aims to comprehensively review the currently available evidence regarding the change in the levels of NETs biomarkers in patients with thrombosis, summarize the role of NETs and its biomarkers in the development and prognostic assessment of venous thromboembolism, coronary artery diseases, ischemic stroke, cancer-associated thromboembolism, and coronavirus disease 2019-associated thromboembolism, explore the potential therapeutic implications of NETs, and further discuss the shortcomings of existing NETs biomarkers in serum and plasma and their detection methods.
Collapse
Affiliation(s)
- Xiangbo Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Shixue Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Yue Yin
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Valerio De Stefano
- Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli IRCCS, Section of Hematology, Rome, Italy
| | - Qingchun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.
- Department of Pharmacy, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China.
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (the Teaching School of Shenyang Pharmaceutical University), Shenyang, China.
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
25
|
Ngo ATP, Gollomp K. Building a better
NET
: Neutrophil extracellular trap targeted therapeutics in the treatment of infectious and inflammatory disorders. Res Pract Thromb Haemost 2022. [DOI: 10.1002/rth2.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Anh T. P. Ngo
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Kandace Gollomp
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
- Department of Pediatrics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
26
|
Scavenger receptor-targeted plaque delivery of microRNA-coated nanoparticles for alleviating atherosclerosis. Proc Natl Acad Sci U S A 2022; 119:e2201443119. [PMID: 36122215 PMCID: PMC9522431 DOI: 10.1073/pnas.2201443119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis treatments by gene regulation are garnering attention, yet delivery of gene cargoes to atherosclerotic plaques remains inefficient. Here, we demonstrate that assembly of therapeutic oligonucleotides into a three-dimensional spherical nucleic acid nanostructure improves their systemic delivery to the plaque and the treatment of atherosclerosis. This noncationic nanoparticle contains a shell of microRNA-146a oligonucleotides, which regulate the NF-κB pathway, for achieving transfection-free cellular entry. Upon an intravenous injection into apolipoprotein E knockout mice fed with a high-cholesterol diet, this nanoparticle naturally targets class A scavenger receptor on plaque macrophages and endothelial cells, contributing to elevated delivery to the plaques (∼1.2% of the injected dose). Repeated injections of the nanoparticle modulate genes related to immune response and vascular inflammation, leading to reduced and stabilized plaques but without inducing severe toxicity. Our nanoparticle offers a safe and effective treatment of atherosclerosis and reveals the promise of nucleic acid nanotechnology for cardiovascular disease.
Collapse
|
27
|
Mortazavi-Jahromi SS, Aslani M. Dysregulated miRNAs network in the critical COVID-19: An important clue for uncontrolled immunothrombosis/thromboinflammation. Int Immunopharmacol 2022; 110:109040. [PMID: 35839566 PMCID: PMC9271492 DOI: 10.1016/j.intimp.2022.109040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
Known as a pivotal immunohemostatic response, immunothrombosis is activated to restrict the diffusion of pathogens. This beneficial intravascular defensive mechanism represents the close interaction between the immune and coagulation systems. However, its uncontrolled form can be life-threatening to patients with the critical coronavirus disease 2019 (COVID-19). Hyperinflammation and ensuing cytokine storm underlie the activation of the coagulation system, something which results in the provocation of more immune-inflammatory responses by the thrombotic mediators. This vicious cycle causes grave clinical complications and higher risks of mortality. Classified as an evolutionarily conserved family of the small non-coding RNAs, microRNAs (miRNAs) serve as the fine-tuners of genes expression and play a key role in balancing the pro/anticoagulant and pro-/anti-inflammatory factors maintaining homeostasis. Therefore, any deviation from their optimal expression levels or efficient functions can lead to severe complications. Despite their extensive effects on the molecules and processes involved in uncontrolled immunothrombosis, some genetic agents and uncontrolled immunothrombosis-induced interfering factors (e.g., miRNA-single nucleotide polymorphysms (miR-SNPs), the complement system components, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, and reactive oxygen species (ROS)) have apparently disrupted their expressions/functions. This review study aims to give an overview of the role of miRNAs in the context of uncontrolled immunothrombosis/thromboinflammation accompanied by some presumptive interfering factors affecting their expressions/functions in the critical COVID-19. Detecting, monitoring, and resolving these interfering agents mafy facilitate the design and development of the novel miRNAs-based therapeutic approaches to the reduction of complications incidence and mortality in patients with the critical COVID-19.
Collapse
Affiliation(s)
- Seyed Shahabeddin Mortazavi-Jahromi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Cellular and Molecular Biology, Kish International Campus, University of Tehran, Kish, Iran.
| | - Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Hsieh YT, Chou YC, Kuo PY, Tsai HW, Yen YT, Shiau AL, Wang CR. Down-regulated miR-146a expression with increased neutrophil extracellular traps and apoptosis formation in autoimmune-mediated diffuse alveolar hemorrhage. J Biomed Sci 2022; 29:62. [PMID: 36028828 PMCID: PMC9413930 DOI: 10.1186/s12929-022-00849-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing evidences have suggested an important role of microRNAs (miRNAs) in regulating cell death processes including NETosis and apoptosis. Dysregulated expression of miRNAs and increased formation of neutrophil extracellular traps (NETs) and apoptosis participate in autoimmune-mediated diffuse alveolar hemorrhage (DAH), mostly associated with pulmonary capillaritis in systemic lupus erythematosus (SLE) patients. In particular, besides the inhibition of apoptosis, miR-146a can control innate and acquired immune responses, and regulate the toll-like receptor pathway through targeting TRAF6 to reduce the expression of pro-inflammatory cytokines/chemokines like IL-8, a NETosis inducer. METHODS Expression of miR-146a, TRAF6 and NETs were examined in peripheral blood neutrophils (PBNs) and lung tissues from SLE-associated DAH patients, and in neutrophils and pristane-induced DAH lung tissues from C57BL/6 mice. To assess NETs formation, we examined NETosis-related DNAs morphology and crucial mediators including protein arginine deiminase 4 and citrullinated Histone 3. Expression of miR-146a and its endogenous RNA SNHG16 were studied in HL-60 promyelocytic cells and MLE-12 alveolar cells during NETosis and apoptosis processes, respectively. MiR-146a-overexpressed and CRISPR-Cas13d-mediated SNHG16-silenced HL-60 cells were investigated for NETosis. MiR-146a-overexpressed MLE-12 cells were analyzed for apoptosis. Pristane-injected mice received intra-pulmonary miR-146a delivery to evaluate therapeutic efficacy in DAH. RESULTS In DAH patients, there were down-regulated miR-146a levels with increased TRAF6 expression and PMA/LPS-induced NETosis in PBNs, and down-regulated miR-146a levels with increased TRAF6, high-mobility group box 1 (HMGB1), IL-8, NETs and apoptosis expression in lung tissues. HMGB1-stimulated mouse neutrophils had down-regulated miR-146a levels with increased TRAF6, IL-8 and NETs expression. PMA-stimulated HL-60 cells had down-regulated miR-146a levels with enhanced NETosis. MiR-146a-overexpressed or SNHG16-silenced HL-60 cells showed reduced NETosis. Apoptotic MLE-12 cells had down-regulated miR-146a expression and increased HMGB1 release, while miR-146a-overexpressed MLE-12 cells showed reduced apoptosis and HMGB1 production. There were down-regulated miR-146a levels with increased TRAF6, HMGB1, IL-8, NETs and apoptosis expression in mouse DAH lung tissues. Intra-pulmonary miR-146a delivery could suppress DAH by reducing TRAF6, IL-8, NETs and apoptosis expression. CONCLUSIONS Our results demonstrate firstly down-regulated pulmonary miR-146a levels with increased TRAF6 and IL-8 expression and NETs and apoptosis formation in autoimmune-mediated DAH, and implicate a therapeutic potential of intra-pulmonary miR-146a delivery.
Collapse
Affiliation(s)
- Yu-Tung Hsieh
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Pin-Yu Kuo
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Wen Tsai
- Departments of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Yen
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chrong-Reen Wang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
29
|
Han T, Tang H, Lin C, Shen Y, Yan D, Tang X, Guo D. Extracellular traps and the role in thrombosis. Front Cardiovasc Med 2022; 9:951670. [PMID: 36093130 PMCID: PMC9452724 DOI: 10.3389/fcvm.2022.951670] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Thrombotic complications pose serious health risks worldwide. A significant change in our understanding of the pathophysiology of thrombosis has occurred since the discovery of extracellular traps (ETs) and their prothrombotic properties. As a result of immune cells decondensing chromatin into extracellular fibers, ETs promote thrombus formation by acting as a scaffold that activates platelets and coagulates them. The involvement of ETs in thrombosis has been reported in various thrombotic conditions including deep vein thrombosis (DVT), pulmonary emboli, acute myocardial infarction, aucte ischemic stroke, and abdominal aortic aneurysms. This review summarizes the existing evidence of ETs in human and animal model thrombi. The authors described studies showing the existence of ETs in venous or arterial thrombi. In addition, we studied potential novel therapeutic opportunities related to the resolution or prevention of thrombosis by targeting ETs.
Collapse
|
30
|
de los Reyes-García AM, Rivera-Caravaca JM, Zapata-Martínez L, Águila S, Véliz-Martínez A, García-Barberá N, Gil-Perez P, Guijarro-Carrillo PJ, Orenes-Piñero E, López-García C, Lozano ML, Marín F, Martínez C, González-Conejero R. MiR-146a Contributes to Thromboinflammation and Recurrence in Young Patients with Acute Myocardial Infarction. J Pers Med 2022; 12:jpm12071185. [PMID: 35887682 PMCID: PMC9318357 DOI: 10.3390/jpm12071185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/06/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Studies on older patients have established notable conceptual changes in the etiopathogenesis of acute coronary syndrome (ACS), but little is known about this disease in young patients (<45 years). Of special interest is thromboinflammation, key at onset, evolution and therapy of cardiovascular pathology. Therefore, we explored whether ACS at an early age is a thromboinflammatory disease by analyzing NETs and rs2431697 of miR-146a (a miRNA considered as a brake of TLR/NF-kB pathway), elements previously related to higher rates of recurrence in atrial fibrillation and sepsis. We included 359 ACS patients (<45 years) and classified them for specific analysis into G1 (collected during the hospitalization of the first event), G2 and G3 (retrospectively collected from patients with or without ACS recurrence, respectively). cfDNA and citH3−DNA were quantified, and rs2431697 was genotyped. Analysis in the overall cohort showed a moderate but significant correlation between cfDNA and citH3−DNA and Killip−Kimball score. In addition, patients with citH3−DNA > Q4 more frequently had a history of previous stroke (6.1% vs. 1.6%). In turn, rs2431697 did not confer increased risk for the onset of ACS, but T carriers had significantly higher levels of NET markers. By groups, we found that cfDNA levels were similarly higher in all patients, but citH3−DNA was especially higher in G1, suggesting that in plasma, this marker may be attenuated over time. Finally, patients from G2 with the worst markers (cfDNA and citH3−DNA > Q2 and T allele) had a two-fold increased risk of a new ischemic event at 2-year follow-up. In conclusion, our data confirm that ACS is younger onset with thromboinflammatory disease. In addition, these data consolidate rs2431697 as a silent proinflammatory factor predisposing to NETosis, and to a higher rate of adverse events in different cardiovascular diseases.
Collapse
Affiliation(s)
- Ascensión M. de los Reyes-García
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
| | - José Miguel Rivera-Caravaca
- Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain; (J.M.R.-C.); (A.V.-M.); (P.G.-P.); (C.L.-G.); (F.M.)
| | - Laura Zapata-Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
| | - Sonia Águila
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
| | - Andrea Véliz-Martínez
- Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain; (J.M.R.-C.); (A.V.-M.); (P.G.-P.); (C.L.-G.); (F.M.)
| | - Nuria García-Barberá
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
| | - Pablo Gil-Perez
- Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain; (J.M.R.-C.); (A.V.-M.); (P.G.-P.); (C.L.-G.); (F.M.)
| | - Pedro J. Guijarro-Carrillo
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology, University of Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain;
| | - Cecilia López-García
- Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain; (J.M.R.-C.); (A.V.-M.); (P.G.-P.); (C.L.-G.); (F.M.)
| | - María L. Lozano
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, 30003 Murcia, Spain;
| | - Francisco Marín
- Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia, IMIB-Arrixaca, CIBERCV, 30003 Murcia, Spain; (J.M.R.-C.); (A.V.-M.); (P.G.-P.); (C.L.-G.); (F.M.)
| | - Constantino Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), 30003 Murcia, Spain; (A.M.d.l.R.-G.); (L.Z.-M.); (S.Á.); (N.G.-B.); (P.J.G.-C.)
- Correspondence: (C.M.); (R.G.-C.); Tel.: +34-968341990 (ext. 911948) (C.M.); +34-968341990 (ext. 911915) (R.G.-C.)
| | - Rocío González-Conejero
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, 30003 Murcia, Spain;
- Correspondence: (C.M.); (R.G.-C.); Tel.: +34-968341990 (ext. 911948) (C.M.); +34-968341990 (ext. 911915) (R.G.-C.)
| |
Collapse
|
31
|
Xiao S, Liu L, Sun Z, Liu X, Xu J, Guo Z, Yin X, Liao F, Xu J, You Y, Zhang T. Network Pharmacology and Experimental Validation to Explore the Mechanism of Qing-Jin-Hua-Tan-Decoction Against Acute Lung Injury. Front Pharmacol 2022; 13:891889. [PMID: 35873580 PMCID: PMC9304690 DOI: 10.3389/fphar.2022.891889] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
Qing-Jin-Hua-Tan-Decoction (QJHTD), a classic famous Chinese ancient prescription, has been used for treatment of pulmonary diseases since Ming Dynasty. A total of 22 prototype compounds of QJHTD absorbed into rat blood were chosen as candidates for the pharmacological network analysis and molecular docking. The targets from the intersection of compound target and ALI disease targets were used for GO and KEGG enrichment analyses. Molecular docking was adopted to further verify the interactions between 22 components and the top 20 targets with higher degree values in the component-target-pathway network. In vitro experiments were performed to verify the results of network pharmacology using SPR experiments, Western blot experiments, and the PMA-induced neutrophils to produce neutrophil extracellular trap (NET) model. The compound-target-pathway network includes 176 targets and 20 signaling pathways in which the degree of MAPK14, CDK2, EGFR, F2, SRC, and AKT1 is higher than that of other targets and which may be potential disease targets. The biological processes in QJHTD for ALI mainly included protein phosphorylation, response to wounding, response to bacterium, regulation of inflammatory response, and so on. KEGG enrichment analyses revealed multiple signaling pathways, including lipid and atherosclerosis, HIF-1 signaling pathway, renin-angiotensin system, and neutrophil extracellular trap formation. The molecular docking results showed that baicalin, oroxylin A-7-glucuronide, hispidulin-7-O-β-D-glucuronide, wogonoside, baicalein, wogonin, tianshic acid, and mangiferin can be combined with most of the targets, which might be the core components of QJHTD in treatment of ALI. Direct binding ability of baicalein, wogonin, and baicalin to thrombin protein was all micromolar, and their KD values were 11.92 μM, 1.303 μM, and 1.146 μM, respectively, revealed by SPR experiments, and QJHTD could inhibit Src phosphorylation in LPS-activated neutrophils by Western blot experiments. The experimental results of PMA-induced neutrophils to produce NETs indicated that QJHTD could inhibit the production of NETs. This study revealed the active compounds, effective targets, and potential pharmacological mechanisms of QJHTD acting on ALI.
Collapse
Affiliation(s)
- Shunli Xiao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengxiao Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoqian Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyuan Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaojie Yin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fulong Liao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Xu
- National and Local United Engineering Laboratory of Modern Preparation and Quality Control Technology of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Yun You
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiejun Zhang
- National and Local United Engineering Laboratory of Modern Preparation and Quality Control Technology of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| |
Collapse
|
32
|
Sabbatinelli J, Matacchione G, Giuliani A, Ramini D, Rippo MR, Procopio AD, Bonafè M, Olivieri F. Circulating biomarkers of inflammaging as potential predictors of COVID-19 severe outcomes. Mech Ageing Dev 2022; 204:111667. [PMID: 35341896 PMCID: PMC8949647 DOI: 10.1016/j.mad.2022.111667] [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] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 infection has been of unprecedented clinical and socio-economic worldwide relevance. The case fatality rate for COVID-19 grows exponentially with age and the presence of comorbidities. In the older patients, COVID-19 manifests predominantly as a systemic disease associated with immunological, inflammatory, and procoagulant responses. Timely diagnosis and risk stratification are crucial steps to define appropriate therapies and reduce mortality, especially in the older patients. Chronically and systemically activated innate immune responses and impaired antiviral responses have been recognized as the results of a progressive remodeling of the immune system during aging, which can be described by the words 'immunosenescence' and 'inflammaging'. These age-related features of the immune system were highlighted in patients affected by COVID-19 with the poorest clinical outcomes, suggesting that the mechanisms underpinning immunosenescence and inflammaging could be relevant for COVID-19 pathogenesis and progression. Increasing evidence suggests that senescent myeloid and endothelial cells are characterized by the acquisition of a senescence-associated pro-inflammatory phenotype (SASP), which is considered as the main culprit of both immunosenescence and inflammaging. Here, we reviewed this evidence and highlighted several circulating biomarkers of inflammaging that could provide additional prognostic information to stratify COVID-19 patients based on the risk of severe outcomes.
Collapse
Affiliation(s)
- Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Laboratory Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Deborah Ramini
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, Università di Bologna, Bologna, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
33
|
Wang Y, Liu X, Xia P, Li Z, FuChen X, Shen Y, Yu P, Zhang J. The Regulatory Role of MicroRNAs on Phagocytes: A Potential Therapeutic Target for Chronic Diseases. Front Immunol 2022; 13:901166. [PMID: 35634335 PMCID: PMC9130478 DOI: 10.3389/fimmu.2022.901166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/19/2022] [Indexed: 11/27/2022] Open
Abstract
An effective acute inflammatory response results in the elimination of infectious microorganisms, followed by a smooth transition to resolution and repair. During the inflammatory response, neutrophils play a crucial role in antimicrobial defense as the first cells to reach the site of infection damage. However, if the neutrophils that have performed the bactericidal effect are not removed in time, the inflammatory response will not be able to subside. Anti-inflammatory macrophages are the main scavengers of neutrophils and can promote inflammation towards resolution. MicroRNAs (miRNAs) have great potential as clinical targeted therapy and have attracted much attention in recent years. This paper summarizes the involvement of miRNAs in the process of chronic diseases such as atherosclerosis, rheumatoid arthritis and systemic lupus erythematosus by regulating lipid metabolism, cytokine secretion, inflammatory factor synthesis and tissue repair in two types of cells. This will provide a certain reference for miRNA-targeted treatment of chronic diseases.
Collapse
Affiliation(s)
- Yongbo Wang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xingyu Liu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xinxi FuChen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yunfeng Shen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
34
|
Martínez-Fleta P, Vera-Tomé P, Jiménez-Fernández M, Requena S, Roy-Vallejo E, Sanz-García A, Lozano-Prieto M, López-Sanz C, Vara A, Lancho-Sánchez Á, Martín-Gayo E, Muñoz-Calleja C, Alfranca A, González-Álvaro I, Galván-Román JM, Aspa J, de la Fuente H, Sánchez-Madrid F. A Differential Signature of Circulating miRNAs and Cytokines Between COVID-19 and Community-Acquired Pneumonia Uncovers Novel Physiopathological Mechanisms of COVID-19. Front Immunol 2022; 12:815651. [PMID: 35087533 PMCID: PMC8787267 DOI: 10.3389/fimmu.2021.815651] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) pneumonia is a life-threatening infectious disease, especially for elderly patients with multiple comorbidities. Despite enormous efforts to understand its underlying etiopathogenic mechanisms, most of them remain elusive. In this study, we compared differential plasma miRNAs and cytokines profiles between COVID-19 and other community-acquired pneumonias (CAP). A first screening and subsequent validation assays in an independent cohort of patients revealed a signature of 15 dysregulated miRNAs between COVID-19 and CAP patients. Additionally, multivariate analysis displayed a combination of 4 miRNAs (miR-106b-5p, miR-221-3p, miR-25-3p and miR-30a-5p) that significantly discriminated between both pathologies. Search for targets of these miRNAs, combined with plasma protein measurements, identified a differential cytokine signature between COVID-19 and CAP that included EGFR, CXCL12 and IL-10. Significant differences were also detected in plasma levels of CXCL12, IL-17, TIMP-2 and IL-21R between mild and severe COVID-19 patients. These findings provide new insights into the etiopathological mechanisms underlying COVID-19.
Collapse
Affiliation(s)
- Pedro Martínez-Fleta
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Paula Vera-Tomé
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - María Jiménez-Fernández
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Silvia Requena
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Emilia Roy-Vallejo
- Department of Internal Medicine, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Ancor Sanz-García
- Data Analysis Unit, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Marta Lozano-Prieto
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Alicia Vara
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Ángel Lancho-Sánchez
- Biobank, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Enrique Martín-Gayo
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Department of Medicine, Universidad Autónoma de Madrid (IIS-IP), Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Department of Medicine, Universidad Autónoma de Madrid (IIS-IP), Madrid, Spain
| | - Arantzazu Alfranca
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Isidoro González-Álvaro
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Department of Rheumatology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - José María Galván-Román
- Department of Internal Medicine, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Javier Aspa
- Department of Pneumology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain
| | - Hortensia de la Fuente
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Department of Immunology, Hospital Universitario de La Princesa IIS-IP (Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa), Madrid, Spain.,Department of Medicine, Universidad Autónoma de Madrid (IIS-IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
35
|
Herrero-Cervera A, Soehnlein O, Kenne E. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol 2022; 19:177-191. [PMID: 35039631 PMCID: PMC8803838 DOI: 10.1038/s41423-021-00832-3] [Citation(s) in RCA: 309] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is a component of many disease conditions that affect a large group of individuals worldwide. Chronic inflammation is characterized by persistent, low-grade inflammation and is increased in the aging population. Neutrophils are normally the first responders to acute inflammation and contribute to the resolution of inflammation. However, in chronic inflammation, the role of neutrophils is less well understood and has been described as either beneficial or detrimental, causing tissue damage and enhancing the immune response. Emerging evidence suggests that neutrophils are important players in several chronic diseases, such as atherosclerosis, diabetes mellitus, nonalcoholic fatty liver disease and autoimmune disorders. This review will highlight the interaction of neutrophils with other cells in the context of chronic inflammation, the contribution of neutrophils to selected chronic inflammatory diseases, and possible future therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Herrero-Cervera
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Mao JY, Zhang JH, Cheng W, Chen JW, Cui N. Effects of Neutrophil Extracellular Traps in Patients With Septic Coagulopathy and Their Interaction With Autophagy. Front Immunol 2021; 12:757041. [PMID: 34707618 PMCID: PMC8542927 DOI: 10.3389/fimmu.2021.757041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/21/2021] [Indexed: 12/01/2022] Open
Abstract
Introduction Neutrophil extracellular traps (NETs) act as a critical trigger of inflammation and coagulation. We hypothesized that NETs are associated with septic hypercoagulability. Materials and Methods In total, 82 patients admitted with sepsis in the Department of Critical Care Medicine of Peking Union Medical College Hospital were enrolled between February 2017 and April 2018. Clinical and hematological parameters and thrombotic or hemorrhagic events were recorded. Blood samples were obtained to assess biomarkers of NET formation, including neutrophil elastase 2 (ELA2) and citrullinated histone H3, and endothelial-derived biomarker syndecan-1. Autophagy levels and their regulation pathway were also examined to explore their interaction with NETs. Result Sepsis patients with disseminated intravascular coagulation (DIC) showed significantly higher levels of NET formation [ELA2, 1,247 (86–625) vs. 2,039 (1,544–2,534), p < 0.0001; H3, 140 (47–233) vs. 307 (199–415), p < 0.0001]. NET formation was independently associated with DIC risk [ELA2, OR 1.0028, 95% CI, 1.0010–1.0045; H3, OR 1.0104, 95% CI, 1.0032–1.0176] and mortality [ELA2, HR 1.0014, 95% CI, 1.0004–1.0024; H3, HR 1.0056, 95% CI, 1.0008–1.0115]. The area under the curve value for ELA2 in predicting DIC occurrence was 0.902 (95% CI, 0.816–0.957), and that of H3 was 0.870 (95% CI, 0.778–0.934). Furthermore, biomarkers of NET formation, endothelial cells, and autophagy exhibited a significant correlation [ELA2 and Syn (r = 0.5985, p < 0.0001), LC3B (r = −0.4224, p < 0.0001); H3 and Syn (r = 0.6383, p < 0.0001), LC3B (r = −0.3005, p = 0.0061)]. Conclusion Increased NET formation is significantly associated with sepsis-induced DIC incidence and mortality in sepsis patients, revealing a significant relationship with the autophagy pathway. Clinical Trial Registration chictr.org.cn, identifier ChiCTR-ROC-17010750.
Collapse
Affiliation(s)
- Jia-Yu Mao
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jia-Hui Zhang
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Cheng
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jian-Wei Chen
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Allegra A, Tonacci A, Musolino C, Pioggia G, Gangemi S. Secondary Immunodeficiency in Hematological Malignancies: Focus on Multiple Myeloma and Chronic Lymphocytic Leukemia. Front Immunol 2021; 12:738915. [PMID: 34759921 PMCID: PMC8573331 DOI: 10.3389/fimmu.2021.738915] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Secondary immunodeficiency is reported in most patients with hematological malignancies such as chronic lymphocytic leukemia and multiple myeloma. The aim of our review was to evaluate the existing literature data on patients with hematological malignancies, with regard to the effect of immunodeficiency on the outcome, the clinical and therapeutic approach, and on the onset of noninfectious complications, including thrombosis, pleural effusion, and orofacial complications. Immunodeficiency in these patients has an intense impact on their risk of infection, in turn increasing morbidity and mortality even years after treatment completion. However, these patients with increased risk of severe infectious diseases could be treated with adequate vaccination coverage, but the vaccines' administration can be associated with a decreased immune response and an augmented risk of adverse reactions. Probably, immunogenicity of the inactivated is analogous to that of healthy subjects at the moment of vaccination, but it undertakes a gradual weakening over time. However, the dispensation of live attenuated viral vaccines is controversial because of the risk of the activation of vaccine viruses. A particular immunization schedule should be employed according to the clinical and immunological condition of each of these patients to guarantee a constant immune response without any risks to the patients' health.
Collapse
MESH Headings
- Animals
- Humans
- Immunocompromised Host
- Immunogenicity, Vaccine
- Immunologic Deficiency Syndromes/epidemiology
- Immunologic Deficiency Syndromes/immunology
- Immunologic Deficiency Syndromes/therapy
- Incidence
- Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Multiple Myeloma/epidemiology
- Multiple Myeloma/immunology
- Multiple Myeloma/therapy
- Opportunistic Infections/epidemiology
- Opportunistic Infections/immunology
- Opportunistic Infections/prevention & control
- Risk Factors
- Vaccination
- Vaccine Efficacy
- Vaccines/administration & dosage
- Vaccines/adverse effects
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), Pisa, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy
| | - Sebastiano Gangemi
- School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
38
|
Yaykasli KO, Schauer C, Muñoz LE, Mahajan A, Knopf J, Schett G, Herrmann M. Neutrophil Extracellular Trap-Driven Occlusive Diseases. Cells 2021; 10:2208. [PMID: 34571857 PMCID: PMC8466545 DOI: 10.3390/cells10092208] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
The enlightenment of the formation of neutrophil extracellular traps (NETs) as a part of the innate immune system shed new insights into the pathologies of various diseases. The initial idea that NETs are a pivotal defense structure was gradually amended due to several deleterious effects in consecutive investigations. NETs formation is now considered a double-edged sword. The harmful effects are not limited to the induction of inflammation by NETs remnants but also include occlusions caused by aggregated NETs (aggNETs). The latter carries the risk of occluding tubular structures like vessels or ducts and appear to be associated with the pathologies of various diseases. In addition to life-threatening vascular clogging, other occlusions include painful stone formation in the biliary system, the kidneys, the prostate, and the appendix. AggNETs are also prone to occlude the ductal system of exocrine glands, as seen in ocular glands, salivary glands, and others. Last, but not least, they also clog the pancreatic ducts in a murine model of neutrophilia. In this regard, elucidating the mechanism of NETs-dependent occlusions is of crucial importance for the development of new therapeutic approaches. Therefore, the purpose of this review is to address the putative mechanisms of NETs-associated occlusions in the pathogenesis of disease, as well as prospective treatment modalities.
Collapse
Affiliation(s)
- Kursat Oguz Yaykasli
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christine Schauer
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Luis E. Muñoz
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Aparna Mahajan
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jasmin Knopf
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (K.O.Y.); (L.E.M.); (A.M.); (J.K.); (G.S.); (M.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
39
|
Wan W, Liu H, Long Y, Wan W, Li Q, Zhu W, Wu Y. The association between circulating neutrophil extracellular trap related biomarkers and retinal vein occlusion incidence: A case-control pilot study. Exp Eye Res 2021; 210:108702. [PMID: 34270977 DOI: 10.1016/j.exer.2021.108702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 06/22/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022]
Abstract
Retinal vein occlusion (RVO) is the second most common retinal vascular disorders and causes visual damage in a large population. Neutrophil extracellular traps (NETs) formation (NETosis) is an important cause of vascular diseases, however, the association between NETs related biomarkers and RVO development remained unclear. In this pilot study, a total of 77 RVO cases and 48 controls were included between Jan 2020 and July 2020. Besides, the circulating levels of three NETs related markers, cell-free DNA (cfDNA), myeloperoxidase (MPO)-DNA and citrullinated histone H3 (H3Cit), were detected in all the participants and thus the association between NETosis and RVO incidence was analyzed. Advanced assays were conducted to investigate the inflammation and thrombosis related biomarkers in RVO cases with higher or lower NETs biomarkers. When the results were considered, it was found that NETs biomarkers, including cfDNA, MPO-DNA and H3Cit, were increased in the RVO cases comparing with the controls (P < 0.05). Through the receiver operating characteristic (ROC) analyses, we found that circulating NETs related biomarkers demonstrated potential diagnostic effects for RVO and the AUCs of plasma cfDNA, MPO-DNA and H3Cit were 0.859, 0.871 and 0.928, respectively (P < 0.001). Through analyzing the correlations between circulating NETs markers and RVO stages and durations, inflammatory markers as well as thrombotic indexes, it was found that NETs were related with the RVO subtypes, inflammatory status and thrombus formation. In conclusion, the plasma NETs remnants are significantly increased in RVO cases. Besides, advanced studies demonstrate that inflammation as well as thrombus formation might be involved in this association.
Collapse
Affiliation(s)
- Wencui Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongzhuo Liu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Long
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiuming Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei Zhu
- Department of Ophthalmology, Changshu No. 2 People's Hospital, Changshu, China.
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China; Mois Biotech Company, Shanghai, China.
| |
Collapse
|
40
|
Mitchell MI, Ben‐Dov IZ, Liu C, Ye K, Chow K, Kramer Y, Gangadharan A, Park S, Fitzgerald S, Ramnauth A, Perlin DS, Donato M, Bhoy E, Manouchehri Doulabi E, Poulos M, Kamali‐Moghaddam M, Loudig O. Extracellular Vesicle Capture by AnTibody of CHoice and Enzymatic Release (EV-CATCHER): A customizable purification assay designed for small-RNA biomarker identification and evaluation of circulating small-EVs. J Extracell Vesicles 2021; 10:e12110. [PMID: 34122779 PMCID: PMC8173589 DOI: 10.1002/jev2.12110] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Circulating nucleic acids, encapsulated within small extracellular vesicles (EVs), provide a remote cellular snapshot of biomarkers derived from diseased tissues, however selective isolation is critical. Current laboratory-based purification techniques rely on the physical properties of small-EVs rather than their inherited cellular fingerprints. We established a highly-selective purification assay, termed EV-CATCHER, initially designed for high-throughput analysis of low-abundance small-RNA cargos by next-generation sequencing. We demonstrated its selectivity by specifically isolating and sequencing small-RNAs from mouse small-EVs spiked into human plasma. Western blotting, nanoparticle tracking, and transmission electron microscopy were used to validate and quantify the capture and release of intact small-EVs. As proof-of-principle for sensitive detection of circulating miRNAs, we compared small-RNA sequencing data from a subset of small-EVs serum-purified with EV-CATCHER to data from whole serum, using samples from a small cohort of recently hospitalized Covid-19 patients. We identified and validated, only in small-EVs, hsa-miR-146a and hsa-miR-126-3p to be significantly downregulated with disease severity. Separately, using convalescent sera from recovered Covid-19 patients with high anti-spike IgG titers, we confirmed the neutralizing properties, against SARS-CoV-2 in vitro, of a subset of small-EVs serum-purified by EV-CATCHER, as initially observed with ultracentrifuged small-EVs. Altogether our data highlight the sensitivity and versatility of EV-CATCHER.
Collapse
Affiliation(s)
- Megan I. Mitchell
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Iddo Z. Ben‐Dov
- Laboratory of Medical TranscriptomicsHadassah‐Hebrew University Medical CenterJerusalemIsrael
| | - Christina Liu
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Kenny Ye
- Department of Epidemiology and Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Kar Chow
- BiorepositoryHackensack University Medical CenterHackensackNew JerseyUSA
| | - Yael Kramer
- BiorepositoryHackensack University Medical CenterHackensackNew JerseyUSA
| | - Anju Gangadharan
- BiorepositoryHackensack University Medical CenterHackensackNew JerseyUSA
| | - Steven Park
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Sean Fitzgerald
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Andrew Ramnauth
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkUSA
| | - David S. Perlin
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Michele Donato
- BiorepositoryHackensack University Medical CenterHackensackNew JerseyUSA
| | - Emily Bhoy
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Ehsan Manouchehri Doulabi
- Department of Immunology, Genetics and PathologyScience for Life LaboratoryUppsala UniversityUppsalaSweden
| | - Michael Poulos
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| | - Masood Kamali‐Moghaddam
- Department of Immunology, Genetics and PathologyScience for Life LaboratoryUppsala UniversityUppsalaSweden
| | - Olivier Loudig
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew JerseyUSA
| |
Collapse
|
41
|
Blanch-Ruiz MA, Ortega-Luna R, Martínez-Cuesta MÁ, Álvarez Á. The Neutrophil Secretome as a Crucial Link between Inflammation and Thrombosis. Int J Mol Sci 2021; 22:4170. [PMID: 33920656 PMCID: PMC8073391 DOI: 10.3390/ijms22084170] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases are a leading cause of death. Blood-cell interactions and endothelial dysfunction are fundamental in thrombus formation, and so further knowledge of the pathways involved in such cellular crosstalk could lead to new therapeutical approaches. Neutrophils are secretory cells that release well-known soluble inflammatory signaling mediators and other complex cellular structures whose role is not fully understood. Studies have reported that neutrophil extracellular vesicles (EVs) and neutrophil extracellular traps (NETs) contribute to thrombosis. The objective of this review is to study the role of EVs and NETs as key factors in the transition from inflammation to thrombosis. The neutrophil secretome can promote thrombosis due to the presence of different factors in the EVs bilayer that can trigger blood clotting, and to the release of soluble mediators that induce platelet activation or aggregation. On the other hand, one of the main pathways by which NETs induce thrombosis is through the creation of a scaffold to which platelets and other blood cells adhere. In this context, platelet activation has been associated with the induction of NETs release. Hence, the structure and composition of EVs and NETs, as well as the feedback mechanism between the two processes that causes pathological thrombus formation, require exhaustive analysis to clarify their role in thrombosis.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
42
|
Chen Z, Zhang H, Qu M, Nan K, Cao H, Cata JP, Chen W, Miao C. Review: The Emerging Role of Neutrophil Extracellular Traps in Sepsis and Sepsis-Associated Thrombosis. Front Cell Infect Microbiol 2021; 11:653228. [PMID: 33816356 PMCID: PMC8010653 DOI: 10.3389/fcimb.2021.653228] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with sepsis commonly suffer from coagulation dysfunction and lead to the formation of thrombus. During the development of sepsis, neutrophils migrate from the circulating blood to infected tissues and mediate the formation of neutrophil extracellular traps (NETs) that kill pathogens. However, the overactivation of neutrophils can promote the formation of immunothrombosis and even cause disseminated intravascular coagulation (DIC), which damages microcirculation. The outcome of sepsis depends on early recognition and intervention, so clinical evaluation of NETs function may be a valuable biomarker for early diagnosis of sepsis. The interaction of NETs with platelets, complement, and endothelium mediates the formation of immunothrombosis in sepsis. Inhibiting the formation of NETs is also considered to be one of the potential treatments for sepsis. In this review, we will discuss the key role of neutrophils and NETs in sepsis and septic thrombosis, in order to reveal new mechanisms for thrombosis treatment of sepsis.
Collapse
Affiliation(s)
- Zhaoyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanzhong Cao
- Anesthesiology and Surgical Oncology Research Group, Department of Anesthesiology and Perioperative Medicine, Nantong, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, Anesthesiology and Surgical Oncology Research Group, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.,Zhangjiang Institute, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Ling S, Xu JW. NETosis as a Pathogenic Factor for Heart Failure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687096. [PMID: 33680285 PMCID: PMC7929675 DOI: 10.1155/2021/6687096] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
Heart failure threatens the lives of patients and reduces their quality of life. Heart failure, especially heart failure with preserved ejection fraction, is closely related to systemic and local cardiac persistent chronic low-grade aseptic inflammation, microvascular damage characterized by endothelial dysfunction, oxidative stress, myocardial remodeling, and fibrosis. However, the initiation and development of persistent chronic low-grade aseptic inflammation is unexplored. Oxidative stress-mediated neutrophil extracellular traps (NETs) are the main immune defense mechanism against external bacterial infections. Furthermore, NETs play important roles in noninfectious diseases. After the onset of myocardial infarction, atrial fibrillation, or myocarditis, neutrophils infiltrate the damaged tissue and aggravate inflammation. In tissue injury, damage-related molecular patterns (DAMPs) may induce pattern recognition receptors (PRRs) to cause NETs, but whether NETs are directly involved in the pathogenesis and development of heart failure and the mechanism is still unclear. In this review, we analyzed the markers of heart failure and heart failure-related diseases and comorbidities, such as mitochondrial DNA, high mobility box group box 1, fibronectin extra domain A, and galectin-3, to explore their role in inducing NETs and to investigate the mechanism of PRRs, such as Toll-like receptors, receptor for advanced glycation end products, cGAS-STING, and C-X-C motif chemokine receptor 2, in activating NETosis. Furthermore, we discussed oxidative stress, especially the possibility that imbalance of thiol redox and MPO-derived HOCl promotes the production of 2-chlorofatty acid and induces NETosis, and analyzed the possibility of NETs triggering coronary microvascular thrombosis. In some heart diseases, the deletion or blocking of neutrophil-specific myeloperoxidase and peptidylarginine deiminase 4 has shown effectiveness. According to the results of current pharmacological studies, MPO and PAD4 inhibitors are effective at least for myocardial infarction, atherosclerosis, and certain autoimmune diseases, whose deterioration can lead to heart failure. This is essential for understanding NETosis as a therapeutic factor of heart failure and the related new pathophysiology and therapeutics of heart failure.
Collapse
Affiliation(s)
- Shuang Ling
- Institute of Interdisciplinary Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jin-Wen Xu
- Institute of Interdisciplinary Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
44
|
Águila S, de los Reyes-García AM, Fernández-Pérez MP, Reguilón-Gallego L, Zapata-Martínez L, Ruiz-Lorente I, Vicente V, González-Conejero R, Martínez C. MicroRNAs as New Regulators of Neutrophil Extracellular Trap Formation. Int J Mol Sci 2021; 22:ijms22042116. [PMID: 33672737 PMCID: PMC7924615 DOI: 10.3390/ijms22042116] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are formed after neutrophils expelled their chromatin content in order to primarily capture and eliminate pathogens. However, given their characteristics due in part to DNA and different granular proteins, NETs may induce a procoagulant response linking inflammation and thrombosis. Unraveling NET formation molecular mechanisms as well as the intracellular elements that regulate them is relevant not only for basic knowledge but also to design diagnostic and therapeutic tools that may prevent their deleterious effects observed in several inflammatory pathologies (e.g., cardiovascular and autoimmune diseases, cancer). Among the potential elements involved in NET formation, several studies have investigated the role of microRNAs (miRNAs) as important regulators of this process. miRNAs are small non-coding RNAs that have been involved in the control of almost all physiological processes in animals and plants and that are associated with the development of several pathologies. In this review, we give an overview of the actual knowledge on NETs and their implication in pathology with a special focus in cardiovascular diseases. We also give a brief overview on miRNA biology to later focus on the different miRNAs implicated in NET formation and the perspectives opened by the presented data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rocío González-Conejero
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| | - Constantino Martínez
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| |
Collapse
|