1
|
Jalakani R, Khodadadi A, Rezaei E, Motamed N, Bavarsad AH, Mohammadi SA, Mohammadi Z, Khamisipour G. Predictive value of miR-24-3p and miR-1277-5p in hemodialysis patients with vascular access thrombosis. Blood Coagul Fibrinolysis 2025:00001721-990000000-00194. [PMID: 40289845 DOI: 10.1097/mbc.0000000000001357] [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: 11/08/2024] [Accepted: 02/23/2025] [Indexed: 04/30/2025]
Abstract
One of the complications of chronic kidney disease (CKD) is venous thromboembolism (VTE). Currently, the D-dimer test is used for the diagnosis of VTE. This test has low diagnostic value and specificity. Circulating microRNAs are present in plasma, serum, and other body fluids and have recently been shown to be valuable biomarkers in numerous illnesses. Therefore, this study aimed to evaluate the diagnostic potential of serum microRNAs as noninvasive biomarkers for VTE diagnosis in hemodialysis patients. Serum samples were collected from 42 hemodialysis patients with thrombosis, 42 hemodialysis patients without thrombosis, and 42 healthy individuals. After the synthesis of cDNA from serum, the expression of miR-24-3P and miR-1277-5P was measured by qRT-PCR. The data were analyzed using SPSS 20 and GraphPad Prism7 software. The expression level of miR-24-3P in the thrombotic and nonthrombotic hemodialysis groups was significantly greater than that in the healthy groups after adjustment for hyperglycemia (P = 0.003, P = 0.04). Receiver-operating characteristic (ROC) analysis revealed that the area under the curve (AUC) values were 0.769 and 0.649, respectively. However, in the thrombotic group compared with the nonthrombotic group after adjustment for hyperglycemia, no significant difference was detected (P = 0.063), and the AUC was 0.62. After adjustment for age, sex, and BMI, there was a significant difference between the thrombotic group and the nonthrombotic group (P = 0.002), and the AUC was 0.71. Compared with that in the control group, the odds ratio (OR) of increased miR-1277-5P expression was greater in the thrombotic group (P = 0.05, OR = 1.618). There was no significant difference between the nonthrombotic group and the control group (P = 0.73, OR = 0.914). Our results indicated that miR-24-3P is not a reliable marker for examining thrombosis in the studied samples, while miR-1277-5P has a positive association with VAT and could be considered a diagnostic and therapeutic marker.
Collapse
Affiliation(s)
- Raziyeh Jalakani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr
| | | | - Eisa Rezaei
- Department of Nephrology, Ahvaz Jundishapur University of Medical Science, Ahvaz
| | - Niloofar Motamed
- Department of Community Medicine, School of Medicine, Bushehr University of Medical Science, Bushehr
| | - Amir Hooshang Bavarsad
- Department of Internal Medicine, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Amin Mohammadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr
| | - Zahra Mohammadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr
| | - Gholamreza Khamisipour
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr
| |
Collapse
|
2
|
Imiela AM, Kucharska J, Kukliński F, Fernandez Moreno T, Dzik K, Pruszczyk P. Advanced Research in the Pathophysiology of Venous Thromboembolism-Acute Pulmonary Embolism. Biomedicines 2025; 13:906. [PMID: 40299499 PMCID: PMC12025274 DOI: 10.3390/biomedicines13040906] [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: 03/03/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
According to the literature, cardiovascular diseases (CVDs)-including myocardial infarction, stroke, and venous thromboembolism (VTE)-are among the leading causes of mortality and morbidity worldwide. Evidence suggests that CVDs share common risk factors and pathophysiological mechanisms. Similar to the Mosaic Theory of Hypertension proposed by Irvine Page in 1949, the pathophysiology of VTE is multifactorial, involving multiple interacting processes. The concept of immunothrombosis, introduced by Engelmann and Massberg in 2009, describes the interplay between the immune system and thrombosis. Both thrombosis and hemostasis share core mechanisms, including platelet activation and fibrin formation. Additionally, immune mediators-such as monocytes, neutrophil extracellular traps (NETs), lymphocytes, selectins, and various molecular factors-play a critical role in thrombus formation. This review highlights inflammation as a key risk factor for pulmonary embolism (APE). Immunity is central to the complex interactions among the coagulation cascade, platelets, endothelium, reactive oxygen species (ROS), and genetic factors. Specifically, we examine the roles of the endothelium, immune cells, and microRNAs (miRNAs) in the pathophysiology of APE and explore potential therapeutic targets. This review aims to elucidate the roles of the endothelium, immune cells, and miRNAs in the pathophysiology of APE and explore potential future perspective.
Collapse
Affiliation(s)
- Anna M. Imiela
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Joanna Kucharska
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Franciszek Kukliński
- Department of Intensive Cardiac Care, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Teresa Fernandez Moreno
- Department of Intensive Cardiac Care, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Konrad Dzik
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Piotr Pruszczyk
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| |
Collapse
|
3
|
Tang L, Niu S, Xu J, Lu W, Zhou L. miR-221-3p is upregulated in acute pulmonary embolism complicated with pulmonary hypertension and promotes pulmonary arterial smooth muscle cells proliferation and migration by inhibiting PTEN. Cytotechnology 2024; 76:453-463. [PMID: 38933873 PMCID: PMC11196540 DOI: 10.1007/s10616-024-00628-z] [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: 07/27/2023] [Accepted: 03/18/2024] [Indexed: 06/28/2024] Open
Abstract
Pulmonary arterial smooth muscle cells (PASMCs) functions are associated with the pathogenesis of pulmonary hypertension (PH) which is a life-threatening complication of acute pulmonary embolism (APE). This study sought to explore the expression pattern of microRNA (miR)-221-3p in APE-PH patients and its role in PASMCs proliferation and migration. The clinical data and venous blood of APE-PH patients were collected. The expression levels of miR-221-3p and phosphatase and tensin homolog (PTEN) in serum were determined, followed by receiver operator characteristic curve analysis of miR-221-3p diagnostic efficacy. PASMCs were transfected with miR-221-3p mimics and PTEN-overexpressed vector, followed by assessment of cell viability, proliferation, and migration through cell counting kit-8, 5-ethynyl-2'-deoxyuridine, Transwell, and wound healing assays. The binding between miR-221-3p and PTEN 3'UTR region was testified by the dual-luciferase assay. miR-221 was upregulated in the serum of APE-PH patients and presented with good diagnostic efficacy with 1.155 cutoff value, 66.25% sensitivity, and 67.50% specificity. miR-221 was negatively correlated with PTEN in APE-PH patients. miR-221 overexpression facilitated PASMCs proliferation and migration in vitro. miR-221-3p bound to PTEN 3'UTR region to decrease PTEN protein levels. PTEN overexpression abolished the promotive role of miR-221-3p in PASMCs. Overall, miR-221-3p targeted PTEN to facilitate PASMC proliferation and migration.
Collapse
Affiliation(s)
- Lei Tang
- Vascular Surgery Department, Hebei General Hospital, Shijiazhuang City, 050000 China
| | - Shuai Niu
- Vascular Surgery Department, Hebei General Hospital, Shijiazhuang City, 050000 China
| | - Jinwei Xu
- Respiratory Medicine Department, Hebei General Hospital, Shijiazhuang City, 050000 China
| | - Wei Lu
- Respiratory Medicine Department, Hebei Medical University Third Hospital, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang City, 050000 Hebei Province China
| | - Li Zhou
- Respiratory Medicine Department, Hebei Medical University Third Hospital, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang City, 050000 Hebei Province China
| |
Collapse
|
4
|
Kenneweg F, Hobohm L, Bang C, Gupta SK, Xiao K, Thum S, Ten Cate V, Rapp S, Hasenfuß G, Wild P, Konstantinides S, Wachter R, Lankeit M, Thum T. Circulating miR-let7a levels predict future diagnosis of chronic thromboembolic pulmonary hypertension. Sci Rep 2024; 14:4514. [PMID: 38402278 PMCID: PMC10894210 DOI: 10.1038/s41598-024-55223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
Distinct patterns of circulating microRNAs (miRNAs) were found to be involved in misguided thrombus resolution. Thus, we aimed to investigate dysregulated miRNA signatures during the acute phase of pulmonary embolism (PE) and test their diagnostic and predictive value for future diagnosis of chronic thromboembolic pulmonary hypertension (CTEPH). Microarray screening and subsequent validation in a large patient cohort (n = 177) identified three dysregulated miRNAs as potential biomarkers: circulating miR-29a and miR-720 were significantly upregulated and miR-let7a was significantly downregulated in plasma of patients with PE. In a second validation study equal expression patterns for miR-29a and miR-let7a regarding an acute event of recurrent venous thromboembolism (VTE) or deaths were found. MiR-let7a concentrations significantly correlated with echocardiographic and laboratory parameters indicating right ventricular (RV) dysfunction. Additionally, circulating miR-let7a levels were associated with diagnosis of CTEPH during follow-up. Regarding CTEPH diagnosis, ROC analysis illustrated an AUC of 0.767 (95% CI 0.54-0.99) for miR-let7a. Using logistic regression analysis, a calculated patient-cohort optimized miR-let7a cut-off value derived from ROC analysis of ≥ 11.92 was associated with a 12.8-fold increased risk for CTEPH. Therefore, miR-let7a might serve as a novel biomarker to identify patients with haemodynamic impairment and as a novel predictor for patients at risk for CTEPH.
Collapse
Affiliation(s)
- Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Lukas Hobohm
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Claudia Bang
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Shashi K Gupta
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Sabrina Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Vincent Ten Cate
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Steffen Rapp
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Cardiovascular Research Centre (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Gerd Hasenfuß
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Philipp Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
- German Cardiovascular Research Centre (DZHK), Partner Site Rhine Main, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Rolf Wachter
- Clinic of Cardiology and Pneumology, Heart Center, University Medical Center, Goettingen, Germany
- Clinic and Policlinic for Cardiology, University Hospital Leipzig, Leipzig, Germany
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum (CVK), Charité-University Medicine Berlin, Berlin, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.
- REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
5
|
Tavares V, Neto BV, Marques IS, Assis J, Pereira D, Medeiros R. Cancer-associated thrombosis: What about microRNAs targeting the tissue factor coagulation pathway? Biochim Biophys Acta Rev Cancer 2024; 1879:189053. [PMID: 38092078 DOI: 10.1016/j.bbcan.2023.189053] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023]
Abstract
Cancer patients are often diagnosed with venous thromboembolism (VTE), a cardiovascular disease that substantially decreases their quality of life and survival rate. Haemostasis in these patients is deregulated, which is reflected in the common presentation of a blood hypercoagulation state. Despite the inconsistent results, existing evidence suggests that the expression of microRNAs (miRNAs) is deregulated in the context of venous thrombogenesis in the general population. However, few miRNAs are known to be linked to cancer-associated VTE due to the lack of studies with oncological patients. Parallelly, coagulation factor III, also known as tissue factor (TF), tissue factor pathway inhibitor 1 (TFPI1) and tissue factor pathway inhibitor 2 (TFPI2) have been proposed to have a central role in cancer-associated VTE and tumour progression. Yet, contrary to what was expected, the role of miRNAs targeting the TF coagulation pathway (or extrinsic coagulation pathway) is poorly explored in cancer-induced thrombogenesis. In this review, in addition to miRNAs implicated in VTE, TF and TFPI1/2-targeting miRNAs were revised. Future studies should clarify the implications of these non-coding RNAs in tumour coagulome.
Collapse
Affiliation(s)
- Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/ Pathology and Laboratory Medicine Dep., Clinical Pathology SV/ RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal; Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Beatriz Vieira Neto
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/ Pathology and Laboratory Medicine Dep., Clinical Pathology SV/ RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| | - Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/ Pathology and Laboratory Medicine Dep., Clinical Pathology SV/ RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Sciences of University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/ Pathology and Laboratory Medicine Dep., Clinical Pathology SV/ RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal; Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal; Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal; Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal.
| |
Collapse
|
6
|
Gareev I, Pavlov V, Du W, Yang B. MiRNAs and Their Role in Venous Thromboembolic Complications. Diagnostics (Basel) 2023; 13:3383. [PMID: 37958279 PMCID: PMC10650162 DOI: 10.3390/diagnostics13213383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Venous thromboembolic complications (VTCs), which include deep vein thrombosis (DVT) and pulmonary embolism (PE), have remained a pressing problem in modern clinical medicine for a long time. Despite the already wide arsenal of modern methods for diagnosing and treating this disease, VTCs rank third in the structure of causes of death among all cardiovascular diseases, behind myocardial infarction (MI) and ischemic stroke (IS). Numerous studies have confirmed the importance of understanding the molecular processes of VTCs for effective therapy and diagnosis. Significant progress has been made in VTC research in recent years, where the relative contribution of microRNAs (miRNAs) in the mechanism of thrombus formation and their consideration as therapeutic targets have been well studied. In this case, accurate, timely, and as early as possible diagnosis of VTCs is of particular importance, which will help improve both short-term and long-term prognoses of patients. This case accounts for the already well-studied circulating miRNAs as non-invasive biomarkers. This study presents currently available literature data on the role of miRNAs in VTCs, revealing their potential as therapeutic targets and diagnostic and prognostic tools for this terrible disease.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, 3 Lenin Street, 450008 Ufa, Russia;
| | - Valentin Pavlov
- Department of Urology, Bashkir State Medical University, 3 Lenin Street, 450008 Ufa, Russia;
| | - Weijie Du
- Department of Pharmacology, The Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Baofeng Yang
- Department of Pharmacology, The Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| |
Collapse
|
7
|
Danckwardt S, Trégouët DA, Castoldi E. Post-transcriptional control of haemostatic genes: mechanisms and emerging therapeutic concepts in thrombo-inflammatory disorders. Cardiovasc Res 2023; 119:1624-1640. [PMID: 36943786 PMCID: PMC10325701 DOI: 10.1093/cvr/cvad046] [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: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/23/2023] Open
Abstract
The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be 'hijacked' in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.
Collapse
Affiliation(s)
- Sven Danckwardt
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK),
Berlin, Germany
- Posttranscriptional Gene Regulation, University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University
Medical Centre Mainz, Langenbeckstr. 1, 55131
Mainz, Germany
- Center for Healthy Aging (CHA), Mainz,
Germany
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Department of
Molecular Epidemiology of Vascular and Brain Disorders (ELEANOR), University of
Bordeaux, Bordeaux, France
| | - Elisabetta Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht
(CARIM), Maastricht University, Universiteitsingel 50, 6229
ER Maastricht, The Netherlands
| |
Collapse
|
8
|
Kotlyar MJ, Krebs M, Solimando AG, Marquardt A, Burger M, Kübler H, Bargou R, Kneitz S, Otto W, Breyer J, Vergho DC, Kneitz B, Kalogirou C. Critical Evaluation of a microRNA-Based Risk Classifier Predicting Cancer-Specific Survival in Renal Cell Carcinoma with Tumor Thrombus of the Inferior Vena Cava. Cancers (Basel) 2023; 15:cancers15071981. [PMID: 37046643 PMCID: PMC10093292 DOI: 10.3390/cancers15071981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
(1) Background: Clear cell renal cell carcinoma extending into the inferior vena cava (ccRCCIVC) represents a clinical high-risk setting. However, there is substantial heterogeneity within this patient subgroup regarding survival outcomes. Previously, members of our group developed a microRNA(miR)-based risk classifier—containing miR-21-5p, miR-126-3p and miR-221-3p expression—which significantly predicted the cancer-specific survival (CSS) of ccRCCIVC patients. (2) Methods: Examining a single-center cohort of tumor tissue from n = 56 patients with ccRCCIVC, we measured the expression levels of miR-21, miR-126, and miR-221 using qRT-PCR. The prognostic impact of clinicopathological parameters and miR expression were investigated via single-variable and multivariable Cox regression. Referring to the previously established risk classifier, we performed Kaplan–Meier analyses for single miR expression levels and the combined risk classifier. Cut-off values and weights within the risk classifier were taken from the previous study. (3) Results: miR-21 and miR-126 expression were significantly associated with lymphonodal status at the time of surgery, the development of metastasis during follow-up, and cancer-related death. In Kaplan–Meier analyses, miR-21 and miR-126 significantly impacted CSS in our cohort. Moreover, applying the miR-based risk classifier significantly stratified ccRCCIVC according to CSS. (4) Conclusions: In our retrospective analysis, we successfully validated the miR-based risk classifier within an independent ccRCCIVC cohort.
Collapse
Affiliation(s)
- Mischa J. Kotlyar
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Markus Krebs
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Antonio Giovanni Solimando
- Guido Baccelli Unit of Internal Medicine, Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
| | - André Marquardt
- Department of Pathology, Klinikum Stuttgart, 70174 Stuttgart, Germany
| | - Maximilian Burger
- Department of Urology, Caritas St. Josef, University of Regensburg Medical Center, 93053 Regensburg, Germany
| | - Hubert Kübler
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Susanne Kneitz
- Physiological Chemistry I, Theodor-Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Wolfgang Otto
- Department of Urology, Caritas St. Josef, University of Regensburg Medical Center, 93053 Regensburg, Germany
| | - Johannes Breyer
- Department of Urology, Caritas St. Josef, University of Regensburg Medical Center, 93053 Regensburg, Germany
| | - Daniel C. Vergho
- Department of Urology, Caritas St. Josef, University of Regensburg Medical Center, 93053 Regensburg, Germany
| | - Burkhard Kneitz
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
- Correspondence: ; Tel.: +49-931-201-32001
| |
Collapse
|
9
|
Contemporary Biomarkers in Pulmonary Embolism Diagnosis: Moving beyond D-Dimers. J Pers Med 2022; 12:jpm12101604. [PMID: 36294744 PMCID: PMC9604705 DOI: 10.3390/jpm12101604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 12/05/2022] Open
Abstract
Pulmonary embolism (PE) is a rather common cardiovascular disorder constituting one of the major manifestations of venous thromboembolism (VTE). It is associated with high mortality and substantial recurrence rates, and its diagnosis may be challenging, especially in patients with respiratory comorbidities. Therefore, providing a prompt and accurate diagnosis for PE through developing highly sensitive and specific diagnostic algorithms would be of paramount importance. There is sound evidence supporting the use of biomarkers to enhance the diagnosis and predict the recurrence risk in patients with PE. Therefore, several novel biomarkers, such as factor VIII, Ischemia Modified Albumin, and fibrinogen, as well as several MicroRNAs and microparticles, have been investigated for the diagnosis of this clinical entity. The present review targets to comprehensively present the literature regarding the novel diagnostic biomarkers for PE, as well as to discuss the evidence for their use in daily routine.
Collapse
|
10
|
Li Y, Shao J, Song J, Yu S, Wang J, Sun K. MiR-34a-3p suppresses pulmonary vascular proliferation in acute pulmonary embolism rat by targeting DUSP1. Biosci Rep 2022; 42:BSR20210116. [PMID: 34778900 PMCID: PMC8703022 DOI: 10.1042/bsr20210116] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Acute pulmonary embolism (APE) is a prevalent reason of cardiovascular morbidity and mortality. Recent studies have underscored the positive effects of microRNAs (miRNAs) on many diseases. The present study aimed to identify the critical miRNA with differential expressions and explore its role in APE. METHODS The critical miRNA with its target gene was screened by bioinformatics analysis. Their binding relationship was analyzed by TargetScan, Dual-luciferase reporter and RNA pull-down assays. A rat model of APE was established by self-blood coagulum. Human pulmonary artery smooth muscle cells (PASMCs) were exposed to platelet-derived growth factor (PDGF-BB) for excessive proliferation, and transfected with miR-34a-3p mimic. Mean pulmonary arterial pressure (mPAP) of rat was measured, and the pulmonary tissues were used for the pathological observation by Hematoxylin-Eosin (H&E) staining. Cell viability and proliferation were detected by Cell Counting Kit-8 (CCK-8) and EdU assays. The expressions of miR-34a-3p with its target genes (including dual-specificity phosphatase-1 (DUSP1)), neuron-derived orphan receptor-1 (NOR-1) and proliferating cell nuclear antigen (PCNA) were determined by quantitative reverse transcription polymerase chain reaction (RT-qPCR) or/and Western blot. RESULTS MiR-34a-3p expression was down-regulated in APE patients, which attenuated the increment of mPAP and thickening of the pulmonary arterial walls in APE rats, accompanied with regulation of NOR-1 and PCNA levels. MiR-34a-3p suppressed DUSP1 expression by directly binding to its 3'-untranslated region (UTR), and attenuated cell viability, proliferation, and the expressions of NOR-1 and PCNA in PDGF-BB-induced PASMCs by inhibiting DUSP1 expression. CONCLUSION Up-regulated miR-34a-3p negatively regulates DUSP1 expression to inhibit PASMC proliferation, which, thus, may act on APE treatment by negatively regulating pulmonary vascular proliferation.
Collapse
MESH Headings
- Animals
- Case-Control Studies
- Cell Proliferation
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Dual Specificity Phosphatase 1/genetics
- Dual Specificity Phosphatase 1/metabolism
- Gene Expression Regulation, Enzymologic
- Male
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Proliferating Cell Nuclear Antigen/genetics
- Proliferating Cell Nuclear Antigen/metabolism
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Pulmonary Embolism/enzymology
- Pulmonary Embolism/genetics
- Pulmonary Embolism/pathology
- Rats, Sprague-Dawley
- Signal Transduction
- Vascular Remodeling
- Rats
Collapse
Affiliation(s)
- Yang Li
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| | - Jinyan Shao
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| | - Jianfeng Song
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| | - Shuili Yu
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiqin Wang
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| | - Keyu Sun
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Sobrero M, Montecucco F, Carbone F. Circulating MicroRNAs for Diagnosis of Acute Pulmonary Embolism: Still a Long Way to Go. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4180215. [PMID: 35047634 PMCID: PMC8763471 DOI: 10.1155/2022/4180215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/14/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022]
Abstract
Venous thromboembolism (VTE) represents the third most frequent cause of acute cardiovascular syndrome. Among VTE, acute pulmonary embolism (APE) is the most life-threatening complication. Due to the low specificity of symptoms clinical diagnosis of APE may be sometimes very difficult. Accordingly, the latest European guidelines only suggest clinical prediction tests for diagnosis of APE, eventually associated with D-dimer, a biomarker burdened by a very low specificity. A growing body of evidence is highlighting the role of miRNAs in hemostasis and thrombosis. Due to their partial inheritance and susceptibility to the environmental factors, miRNAs are increasingly described as active modifiers of the classical Virchow's triad. Clinical evidence on deep venous thrombosis reported specific miRNA signatures associated to thrombosis development, organization, recanalization, and resolution. Conversely, data of miRNA profiling as a predictor/diagnostic marker of APE are still preliminary. Here, we have summarized clinical evidence on the potential role of miRNA in diagnosis of APE. Despite some intriguing insight, miRNA assay is still far from any potential clinical application. Especially, the small sample size of cohorts likely represents the major limitation of published studies, so that extensive analysis of miRNA profiles with a machine learning approach are warranted in the next future. In addition, the cost-benefit ratio of miRNA assay still has a negative impact on their clinical application and routinely test.
Collapse
Affiliation(s)
- Matteo Sobrero
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| |
Collapse
|
12
|
Zhang Y, Tang S, Yang W, Du F. let-7b-5p suppresses the proliferation and migration of pulmonary artery smooth muscle cells via down-regulating IGF1. Clinics (Sao Paulo) 2022; 77:100051. [PMID: 35636162 PMCID: PMC9156868 DOI: 10.1016/j.clinsp.2022.100051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Some previous studies indicated that the excessive proliferation and migration of Pulmonary Artery Smooth Muscle Cells (PASMCs) could be observed in pulmonary artery intima after Pulmonary Embolism (PE) occurred. In addition, recent studies identified some miRNAs that are differentially expressed in the blood of PE patients, which might be used as a diagnostic biomarker for PE, including let-7a-5p, let-7b-5p, and miR-150-5p. Hence, the authors sought to explore the effects of let-7b-5p in PASMC proliferation and migration and the corresponding regulatory mechanism. METHODS Platelet-Derived Growth Factor (PDGF) was utilized to induce the hyper-proliferation model in PASMCs. The mRNA and protein expression levels were detected by RT-qPCR and western blot, respectively. The proliferation of PASMCs was evaluated by the detection of PCNA expression, as well as CCK-8 and Edu assays. Wound healing and Transwell assays were exploited to assess the migration ability of PASMCs. The targets of let-7b-5p were predicted based on two bioinformatics online tools. Dual-luciferase and Ago2 pull-down assays were applied to confirm the interaction between let-7b-5p and IGF1. RESULTS 40 ng/mL PDGF was selected as the optimal concentration to induce PASMCs. let-7b-5p mimics suppressed the proliferation and migration of PDGF-induced PASMCs, while let-7b-5p inhibitor led to the opposite result. In further mechanism exploration, IGF1 was predicted and confirmed as the direct target gene of let-7b-5p. The promotion role of IGF1 overexpression on the proliferation and migration of PDGF-induced PASMCs was dramatically countered by let-7b-5p mimics. CONCLUSION let-7b-5p prohibits the proliferation and migration of PDGF-induced PASMCs by modulating IGF1.
Collapse
Affiliation(s)
- Yadi Zhang
- Department of Respiratory Medicine, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Sihui Tang
- Department of Respiratory Medicine, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Wanchun Yang
- Department of Respiratory Medicine, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Fangbing Du
- Department of Respiratory Medicine, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China.
| |
Collapse
|
13
|
Luo M, Du M, Shu C, Liu S, Li J, Zhang L, Li X. The Function of microRNAs in Pulmonary Embolism: Review and Research Outlook. Front Pharmacol 2021; 12:743945. [PMID: 34737702 PMCID: PMC8560694 DOI: 10.3389/fphar.2021.743945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023] Open
Abstract
Pulmonary embolism (PE) is a common pathologic condition that frequently occurs in patients with deep venous thrombosis. Severe PE may critically suppress cardiopulmonary function, thereby threatening the life of patients. Chronic pulmonary hypertension caused by PE may lead to deterioration of respiratory dysfunction, resulting in complete disability. MicroRNAs (miRNAs) are a group of abundantly expressed non-coding RNAs that exert multiple functions in regulating the transcriptome via post-transcriptional targeting of mRNAs. Specifically, miRNAs bind to target mRNAs in a matching mechanism between the miRNA seed sequence and mRNA 3ʹ UTR, thus modulating the transcript stability or subsequent translation activity by RNA-induced silencing complex. Current studies have reported the function of miRNAs as biomarkers of PE, revealing their mechanism, function, and targetome in venous thrombophilia. This review summarizes the literature on miRNA functions and downstream mechanisms in PE. We conclude that various related miRNAs play important roles in PE and have great potential as treatment targets. For clinical application, we propose that miRNA biomarkers combined with traditional biomarkers or miRNA signatures generated from microchips may serve as a great predictive tool for PE occurrence and prognosis. Further, therapies targeting miRNAs or their upstream/downstream molecules need to be developed more quickly to keep up with the progress of routine treatments, such as anticoagulation, thrombolysis, or surgery.
Collapse
Affiliation(s)
- Mingyao Luo
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Mingyuan Du
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Chang Shu
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Sheng Liu
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jiehua Li
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Lei Zhang
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,The Institute of Vascular Diseases, Central South University, Changsha, China
| | - Xin Li
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,The Institute of Vascular Diseases, Central South University, Changsha, China
| |
Collapse
|
14
|
Yang R, Liu G, Deng C. Pulmonary embolism with chronic obstructive pulmonary disease. Chronic Dis Transl Med 2021; 7:149-156. [PMID: 34505015 PMCID: PMC8413125 DOI: 10.1016/j.cdtm.2021.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 11/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, preventable, and treatable disease. The incidence of COPD is growing annually in China, and it is a significant and growing public health burden. Multivariate analysis showed that COPD was one of the independent risk factors for the occurrence of pulmonary embolism (PE), and the incidence of PE was significantly higher in COPD patients than in normal subjects. However, PE is often overlooked in patients with acute exacerbation of COPD (AECOPD) because there are many similarities in clinical symptoms between PE and AECOPD, which are difficult to distinguish, resulting in the failure of timely treatment and poor prognosis. Therefore, it is of great significance to understand the clinical manifestations, diagnosis, and treatment of COPD combined with PE for making a more accurate diagnosis, providing timely and effective treatment, and improving the prognosis of such patients.
Collapse
Affiliation(s)
- Ruohan Yang
- Institute of Respiratory Disease, Fujian Medical University, Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Guiqing Liu
- The Hammersmith Hospital, London W12 0NN, United Kingdom
| | - Chaosheng Deng
- Institute of Respiratory Disease, Fujian Medical University, Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| |
Collapse
|
15
|
Rossetti P, Goldoni M, Pengo V, Vescovini R, Mozzoni P, Tassoni MI, Lombardi M, Rubino P, Bernuzzi G, Verzicco I, Manotti C, Quintavalla R. MiRNA 126 as a New Predictor Biomarker in Venous Thromboembolism of Persistent Residual Vein Obstruction: A Review of the Literature Plus a Pilot Study. Semin Thromb Hemost 2021; 47:982-991. [PMID: 34243207 DOI: 10.1055/s-0041-1726341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Venous thromboembolism (VTE) is the third most common cardiovascular disease. Interleukins (ILs) and micro-ribonucleic acids (miRNAs) have been proposed as molecules able to modulate endothelial inflammation and platelet hyperactivity. At present, no early biomarkers are available to predict the outcome of VTE. We investigated in a pilot study a selected number of miRNAs and ILs as prognostic VTE biomarkers and reviewed literature in this setting. Twenty-three patients (aged 18-65) with a new diagnosis of non-oncological VTE and free from chronic inflammatory diseases were enrolled. Twenty-three age- and sex-matched healthy blood donors were evaluated as control subjects. Serum miRNAs (MiRNA 126, 155, 17.92, 195), inflammatory cytokines (IL-6, tumor necrosis factor-α, IL-8), and lymphocyte subsets were evaluated in patients at enrolment (T0) and in controls. In VTE patients, clinical and instrumental follow-up were performed assessing residual vein obstruction, miRNA and ILs evaluation at 3 months' follow-up (T1). At T0, IL-8, activated T lymphocytes, Treg lymphocytes, and monocytes were higher in patients compared with healthy controls, as were miRNA 126 levels. Moreover, miRNA 126 and IL-6 were significantly increased at T0 compared with T1 evaluation in VTE patients. Higher levels of MiR126 at T0 correlated with a significant overall thrombotic residual at follow-up. In recent years an increasing number of studies (case-control studies, in vivo studies in animal models, in vitro studies) have suggested the potential role of miRNAs in modulating the cellular and biohumoral responses involved in VTE. In the frame of epidemiological evidence, this pilot study with a novel observational approach supports the notion that miRNA can be diagnostic biomarkers of VTE and first identifies miRNA 126 as a predictor of outcome, being associated with poor early recanalization.
Collapse
Affiliation(s)
- Pietro Rossetti
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| | - Matteo Goldoni
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Vittorio Pengo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Rosanna Vescovini
- Department of Clinical and Experimental Medicine, University Hospital of Parma, Parma, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Maria Ilaria Tassoni
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| | - Maria Lombardi
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| | - Pasquale Rubino
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| | - Gino Bernuzzi
- Immunohematology and Transfusion Center, University Hospital of Parma, Parma, Italy
| | - Ignazio Verzicco
- Department of Clinical and Experimental Medicine, University Hospital of Parma, Parma, Italy
| | - Cesare Manotti
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| | - Roberto Quintavalla
- Department of Internal Medicine, Angiology and Coagulation Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
16
|
A novel rationale for targeting FXI: Insights from the hemostatic microRNA targetome for emerging anticoagulant strategies. Pharmacol Ther 2021; 218:107676. [DOI: 10.1016/j.pharmthera.2020.107676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
|
17
|
Peng L, Han L, Li XN, Miao YF, Xue F, Zhou C. The Predictive Value of microRNA-134 and microRNA-1233 for the Early Diagnosis of Acute Exacerbation of Chronic Obstructive Pulmonary Disease with Acute Pulmonary Embolism. Int J Chron Obstruct Pulmon Dis 2020; 15:2495-2503. [PMID: 33116466 PMCID: PMC7575827 DOI: 10.2147/copd.s266021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The differential diagnosis of acute exacerbation of chronic obstructive pulmonary disease (AECOPD) with acute pulmonary embolism (APE) complications are difficult because of the variability of clinical presentations and the shortage of an unfailing screening biomarkers or instruments. OBJECTIVE Aimed to detect and compare the expression of serum microRNAs (miR-1233, miR-134) in AECOPD patients complicated with APE. PATIENTS/METHODS Blood samples were collected from 52 AECOPD patients (13 patients with APE complications, 39 patients without APE) and 10 patients with stable COPD. Serum miRNAs expression was detected with real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). The levels of plasma D-dimers were determined by detection with an enzyme-linked immunosorbent assay (ELISA). The receiver-operator characteristic (ROC) curve was used for evaluating the diagnostic accuracy of the studied miRNAs. RESULTS According to the Wells score, 42 of the 52 AECOPD patients were unlikely to have APE (≤4 points), whereas the remaining 10 (>4 points) were likely to have APE. There were 4 cases (4/13 30.8%) in the AECOPD combined with APE group with a Wells score of >4 points. The expression levels of miR-1233 and miR-134 in the serum were considerably upregulated in the AECOPD+APE group compared with the AECOPD group and the stable COPD group (P<0.05). The areas under the curve (AUCs) for miR-134 and miR-1233 were, respectively, 0.931 (95% CI 0.863-0.999) (P<0.05) and 0.884 (95% CI 0.79-0.978) (P<0.05) and were higher compared with the AUC for D-dimer of 0.628 (95% CI 0.447-0.809), the AUC for age-adjusted D-dimer of 0.705 (95% CI 0.525-0.885) and the AUC for Wells score of 0.577 (95% CI 0.389-0.765). CONCLUSION Our study indicated that serum miR-1233 and miR-134 have high clinical value in the early diagnosis of AECOPD patients combined with APE, or could be used as potential biomarkers for clinical identification of AECOPD with or without APE complication.
Collapse
Affiliation(s)
- Ling Peng
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Li Han
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Xiao-Ning Li
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Ya-Fang Miao
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Fei Xue
- Department of Respiratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Chao Zhou
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Department of Respiratory Medicine, Guangming Traditional Chinese Medicine Hospital of Pudong New Area, Shanghai, People’s Republic of China
| |
Collapse
|
18
|
Jankowska KI, Sauna ZE, Atreya CD. Role of microRNAs in Hemophilia and Thrombosis in Humans. Int J Mol Sci 2020; 21:ijms21103598. [PMID: 32443696 PMCID: PMC7279366 DOI: 10.3390/ijms21103598] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNA) play an important role in gene expression at the posttranscriptional level by targeting the untranslated regions of messenger RNA (mRNAs). These small RNAs have been shown to control cellular physiological processes including cell differentiation and proliferation. Dysregulation of miRNAs have been associated with numerous diseases. In the past few years miRNAs have emerged as potential biopharmaceuticals and the first miRNA-based therapies have entered clinical trials. Our recent studies suggest that miRNAs may also play an important role in the pathology of genetic diseases that are currently considered to be solely due to mutations in the coding sequence. For instance, among hemophilia A patients there exist a small subset, with normal wildtype genes; i.e., lacking in mutations in the coding and non-coding regions of the F8 gene. Similarly, in many patients with missense mutations in the F8 gene, the genetic defect does not fully explain the severity of the disease. Dysregulation of miRNAs that target mRNAs encoding coagulation factors have been shown to disturb gene expression. Alterations in protein levels involved in the coagulation cascade mediated by miRNAs could lead to bleeding disorders or thrombosis. This review summarizes current knowledge on the role of miRNAs in hemophilia and thrombosis. Recognizing and understanding the functions of miRNAs by identifying their targets is important in identifying their roles in health and diseases. Successful basic research may result in the development and improvement of tools for diagnosis, risk evaluation or even new treatment strategies.
Collapse
Affiliation(s)
- Katarzyna I. Jankowska
- OBRR/DBCD/LCH in the Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Zuben E. Sauna
- OTAT/DPPT/HB in the Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Chintamani D. Atreya
- OBRR/DBCD/LCH in the Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA;
- Correspondence:
| |
Collapse
|
19
|
Chen H, Ma Q, Zhang J, Meng Y, Pan L, Tian H. miR‑106b‑5p modulates acute pulmonary embolism via NOR1 in pulmonary artery smooth muscle cells. Int J Mol Med 2020; 45:1525-1533. [PMID: 32323756 PMCID: PMC7138273 DOI: 10.3892/ijmm.2020.4532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/17/2020] [Indexed: 02/05/2023] Open
Abstract
Acute pulmonary embolism (APE) is a common cause of acute cardiovascular failure and has a high morbidity and mortality rate. Inhibiting the excessive proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) is a potential treatment strategy following an APE. Various microRNAs (miRNAs/miRs) have been shown to regulate cell proliferation, apoptosis and other physiological processes. However, the specific mechanisms underlying the action of multiple miRNAs are still not understood in APE. In the present study, the role of miR‑106b‑5p on APE was demonstrated in platelet‑derived growth factor (PDGF)‑induced PASMCs in vitro and in an APE‑mouse model in vivo. The results showed that miR‑106b‑5p expression was downregulated in PDGF‑induced PASMCs and APE mice, and NOR1 levels were upregulated. Proliferating cell nuclear antigen (PCNA) expression levels in cells and proliferation of PASMCs proliferation and migration were reduced following treatment with miR‑106b‑5p agomiR, and increased following treatment with miR‑106b‑5p antagomiR. miR‑106b‑5p targeted the 3' untranslated region of NOR‑1 mRNA and reduced NOR1 expression. NOR1 overexpression reversed the effects of miR‑106‑5p on PDGF‑induced PASMCs. The functional roles of miR‑106b‑5p in PDGF‑induced PASMCs and an APE mouse‑model, and the underlying molecular mechanisms were evaluated. AgomiR‑106b‑5p improved APE‑induced mortality and pulmonary vascular proliferation in mice. These data suggest that miR‑106‑5p is a novel regulator of proliferation of PASMCs and of pulmonary vascular remodeling through PDGF‑induced PASMCs in an APE mouse model via targeting NOR1. These results expand the understanding of the pathogenesis underlying APE and highlight potential novel therapeutic targets.
Collapse
Affiliation(s)
- Heming Chen
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
- Department of Endocrinology, Ankang Central Hospital, Ankang, Shaanxi 725000, P.R. China
| | - Qiang Ma
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
| | - Junbo Zhang
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
| | - Yan Meng
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
| | - Longfei Pan
- Department of Emergency Medicine, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
| | - Hongyan Tian
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, P.R. China
| |
Collapse
|
20
|
Morelli VM, Brækkan SK, Hansen JB. Role of microRNAs in Venous Thromboembolism. Int J Mol Sci 2020; 21:ijms21072602. [PMID: 32283653 PMCID: PMC7177540 DOI: 10.3390/ijms21072602] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that execute their function by targeted downregulation of gene expressions. There is growing evidence from epidemiological studies and animal models suggesting that the expression level of miRNAs is dysregulated in venous thromboembolism (VTE). In this review, we summarize the current knowledge on the role of miRNAs as biomarkers for VTE and provide general insight into research exploring the modulation of miRNA activity in animal models of venous thrombosis. Up to now, published studies have yielded inconsistent results on the role of miRNAs as biomarkers for VTE with most of the reports focused on diagnostic research. The limited statistical power of the individual studies, due to the small sample sizes, may substantially contribute to the poor reproducibility among studies. In animal models, over-expression or inhibition of some miRNAs appear to influence venous thrombus formation and resolution. However, there is an important gap in knowledge on the potential role of miRNAs as therapeutic targets in VTE. Future research involving large cohorts should be designed to clarify the clinical usefulness of miRNAs as biomarkers for VTE, and animal model studies should be pursued to unravel the role of miRNAs in the pathogenesis of VTE and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Vânia M. Morelli
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT—The Arctic University of Norway, N-9037 Tromsø, Norway; (S.K.B.); (J.-B.H.)
- Correspondence: ; Tel.: +47-77625105
| | - Sigrid K. Brækkan
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT—The Arctic University of Norway, N-9037 Tromsø, Norway; (S.K.B.); (J.-B.H.)
- Division of Internal Medicine, University Hospital of North Norway, N-9037 Tromsø, Norway
| | - John-Bjarne Hansen
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT—The Arctic University of Norway, N-9037 Tromsø, Norway; (S.K.B.); (J.-B.H.)
- Division of Internal Medicine, University Hospital of North Norway, N-9037 Tromsø, Norway
| |
Collapse
|
21
|
Jafarzadeh-Esfehani R, Mostafa Parizadeh S, Sabeti Aghabozorgi A, Yavari N, Sadr-Nabavi A, Alireza Parizadeh S, Ghandehari M, Javanbakht A, Rezaei-Kalat A, Mahdi Hassanian S, Vojdanparast M, Ferns GA, Khazaei M, Avan A. Circulating and tissue microRNAs as a potential diagnostic biomarker in patients with thrombotic events. J Cell Physiol 2020; 235:6393-6403. [PMID: 32198752 DOI: 10.1002/jcp.29639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022]
Abstract
Venous and arterial thrombosis are conditions that have a considerable burden if left untreated. The hypoxia-induced by the occluded vessel can disrupt the circulation of any organ, the cornerstone of treating thrombosis is rapid diagnosis and appropriate treatment. Diagnosis of thrombosis may be made by using laboratory tests or imaging techniques in individuals who have clinical manifestations of a thrombotic event. The use of serum micro ribonucleic acids (RNAs) has recently been applied to the diagnosis of thrombosis. These small RNA molecules are emerging as new diagnostic markers but have had very limited applications in vascular disease. Most of the articles provided various microRNAs with different levels of accuracy. However, there remains a lack of an appropriate panel of the most specific microRNA in the literature. The purpose of the present review was to summarize the existing data on the use of microRNAs as a diagnostic biomarker for venous thrombosis.
Collapse
Affiliation(s)
- Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mostafa Parizadeh
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Negar Yavari
- Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ariane Sadr-Nabavi
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Parizadeh
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Ghandehari
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Javanbakht
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsaneh Rezaei-Kalat
- Department of Psychiatry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Vojdanparast
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, UK
| | - Majid Khazaei
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Department of Physiology, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
Sun Y, Zhang X, Gao H, Liu M, Cao Q, Kang X, Wang Y, Zhu L. Expression of microRNA-514a-5p and its biological function in experimental pulmonary thromboembolism. Am J Transl Res 2019; 11:5514-5530. [PMID: 31632526 PMCID: PMC6789257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/12/2019] [Indexed: 06/10/2023]
Abstract
It is difficult to diagnose pulmonary thromboembolism (PTE) in clinical practice. While microRNAs (miRNAs) have been widely investigated as biomarkers for various diseases, their value as biomarkers for PTE remains largely unknown. In the present study, 83 miRNAs showed altered expression in an intermediate-risk PTE group when compared with their expression in a low-risk PTE group as detected by miRNA microarray analysis. After reviewing those data, hsa-miR-514a-5p was selected as a potential biomarker for PTE progression. Disordered myocardial fibroblast arrangements, broadened intercellular spaces, diapedesis of erythrocytes, and lower numbers of nuclei in the right ventricular wall were observed in rats in a PTE model group when compared to rats in a normal saline (NS) group. Furthermore, hyperexpression of miR-514a-5p exacerbated the morphological characteristics of lung and right ventricular tissues, and caused increased RVHI and lung index values, as well as increased BNP and NT-pro-BNP levels in the PTE model rats, possibly by downregulating Chordin-like 1 (CHRDL1) expression. These results suggest that MiR-514a-5p helps to exasperate PTE development by promoting several aspects of PTE pathology, including inflammation, lung injury, and right ventricular hypertrophy by targeting CHRDL1.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| | - Xingguo Zhang
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| | - Hua Gao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| | - Mingjie Liu
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| | - Qi Cao
- Department of Respiratory Medicine, Shandong Provincial Third HospitalJinan 250031, Shandong, China
| | - Xinyang Kang
- Department of General Surgery, The Hospital of China National Heavy Duty Truck Group CompanyJinan 250031, Shandong, China
| | - Yusheng Wang
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| | - Ling Zhu
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, Shandong, China
| |
Collapse
|