1
|
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
|
2
|
Kasho AKA, Nahand JS, Salmaninejad A, Mirzaei H, Moghoofei M, Bazmani A, Aghbash PS, Rasizadeh R, Farsad-Akhtar N, Baghi HB. PBMC MicroRNAs: Promising Biomarkers for the Differential Diagnosis of COVID-19 Patients with Abnormal Coagulation Indices. Curr Microbiol 2023; 80:248. [PMID: 37341794 DOI: 10.1007/s00284-023-03365-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
MicroRNAs, or miRNAs, may involve in coagulation and inflammation pathways caused by severe Coronavirus disease (COVID-19). Accordingly, this attempt was made to explore the behavior of peripheral blood mononuclear cells (PBMCs) miRNAs as effective biomarkers to diagnose COVID-19 patients with normal and abnormal coagulation indices. We selected the targeted miRNAs (miR-19a-3p, miR-223-3p, miR-143-5p, miR-494-3p and miR-301a-5p) according to previous reports, whose PBMC levels were then determined by real-time PCR. Receiver operating characteristic (ROC) curve was obtained to clarify the diagnostic potency of studied miRNAs. The differentially expressed miRNA profiles and corresponding biological activities were predicted in accordance with bioinformatics data. Targeted miRNAs' expression profiles displayed a significant difference between COVID-19 subjects with normal and abnormal coagulation indices. Moreover, the average miR-223-3p level expressed in COVID-19 cases with normal coagulation indices was significantly lower than that in healthy controls. Based on data from ROC analysis, miR-223-3p and miR-494-3p are promising biomarkers to distinguish the COVID-19 cases with normal or abnormal coagulation indices. Bioinformatics data highlighted the prominent role of selected miRNAs in the inflammation and TGF-beta signaling pathway. The differences existed in the expression profiles of selected miRNAs between the groups introduced miR-494-3p and miR-223-3p as potent biomarkers to prognosis the incidence of COVID-19.
Collapse
Affiliation(s)
- Ammar Khalo Abass Kasho
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Iraqi Ministry of Higher Education and Scientific Research, Tal Afar University, Tal Afar, Iraq
- Department of Plant Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Arash Salmaninejad
- Regenerative Medicine, Organ Procurement and Transplantation Multi-Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Medical Genetics, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Moghoofei
- Infectious Diseases Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahad Bazmani
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Farsad-Akhtar
- Department of Plant Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Goncalves BDS, Meadows A, Pereira DG, Puri R, Pillai SS. Insight into the Inter-Organ Crosstalk and Prognostic Role of Liver-Derived MicroRNAs in Metabolic Disease Progression. Biomedicines 2023; 11:1597. [PMID: 37371692 PMCID: PMC10295788 DOI: 10.3390/biomedicines11061597] [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: 04/07/2023] [Revised: 05/19/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Dysfunctional hepatic metabolism has been linked to numerous diseases, including non-alcoholic fatty liver disease, the most common chronic liver disorder worldwide, which can progress to hepatic fibrosis, and is closely associated with insulin resistance and cardiovascular diseases. In addition, the liver secretes a wide array of metabolites, biomolecules, and microRNAs (miRNAs) and many of these secreted factors exert significant effects on metabolic processes both in the liver and in peripheral tissues. In this review, we summarize the involvement of liver-derived miRNAs in biological processes with an emphasis on delineating the communication between the liver and other tissues associated with metabolic disease progression. Furthermore, the review identifies the primary molecular targets by which miRNAs act. These consolidated findings from numerous studies provide insight into the underlying mechanism of various metabolic disease progression and suggest the possibility of using circulatory miRNAs as prognostic predictors and therapeutic targets for improving clinical intervention strategies.
Collapse
Affiliation(s)
- Bruno de Souza Goncalves
- Department of Surgery and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Avery Meadows
- Department of Surgery and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Duane G Pereira
- Department of Surgery and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Raghav Puri
- Department of Surgery and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Sneha S Pillai
- Department of Surgery and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
4
|
Palakurti R, Biswas N, Roy S, Gnyawali SC, Sinha M, Singh K, Ghatak S, Sen CK, Khanna S. Inducible miR-1224 silences cerebrovascular Serpine1 and restores blood flow to the stroke-affected site of the brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:276-292. [PMID: 36726407 PMCID: PMC9868883 DOI: 10.1016/j.omtn.2022.12.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
The α-tocotrienol (TCT) form of natural vitamin E is more potent than the better known α-tocopherol against stroke. Angiographic studies of canine stroke have revealed beneficial cerebrovascular effects of TCT. This work seeks to understand the molecular basis of such effect. In mice, TCT supplementation improved perfusion at the stroke-affected site by inducing miR-1224. miRNA profiling of a laser-capture-microdissected stroke-affected brain site identified miR-1224 as the only vascular miR induced. Lentiviral knockdown of miR-1224 significantly blunted the otherwise beneficial effects of TCT on stroke outcomes. Studies on primary brain microvascular endothelial cells revealed direct angiogenic properties of miR-1224. In mice not treated with TCT, advance stereotaxic delivery of an miR-1224 mimic to the stroke site markedly improved stroke outcomes. Mechanistic studies identified Serpine1 as a target of miR-1224. Downregulation of Serpine1 augmented the angiogenic response of the miR-1224 mimic in the brain endothelial cells. The inhibition of Serpine1, by dietary TCT and pharmacologically, increased cerebrovascular blood flow at the stroke-affected site and protected against stroke. This work assigns Serpine1, otherwise known to be of critical significance in stroke, a cerebrovascular function that worsens stroke outcomes. miR-1224-dependent inhibition of Serpine1 can be achieved by dietary TCT as well as by the small-molecule inhibitor TM5441.
Collapse
Affiliation(s)
- Ravichand Palakurti
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nirupam Biswas
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sashwati Roy
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Surya C. Gnyawali
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mithun Sinha
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kanhaiya Singh
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Subhadip Ghatak
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandan K. Sen
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Savita Khanna
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Corresponding author: Savita Khanna, PhD, Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
5
|
Anijs RJS, Nguyen YN, Cannegieter SC, Versteeg HH, Buijs JT. MicroRNAs as prognostic biomarkers for (cancer-associated) venous thromboembolism. J Thromb Haemost 2023; 21:7-17. [PMID: 36695398 DOI: 10.1016/j.jtha.2022.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 01/11/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs with gene regulatory functions and are commonly dysregulated in disease states. As miRNAs are relatively stable, easily measured, and accessible from plasma or other body fluids, they are promising biomarkers for the diagnosis and prediction of cancer and cardiovascular diseases. Venous thromboembolism (VTE) is the third most common cardiovascular disease worldwide with high morbidity and mortality. The suggested roles of miRNAs in regulating the pathophysiology of VTE and as VTE biomarkers are nowadays more evidenced. Patients with cancer are at increased risk of developing VTE compared to the general population. However, current risk prediction models for cancer-associated thrombosis (CAT) perform suboptimally, and novel biomarkers are therefore urgently needed to identify which patients may benefit the most from thromboprophylaxis. This review will first discuss how miRNAs mechanistically contribute to the pathophysiology of VTE. Next, the potential use of miRNAs as predictive biomarkers for VTE in subjects without cancer is reviewed, followed by an in-depth focus on CAT. Several of the identified miRNAs in CAT were found to be differentially regulated in VTE as well, giving clues on the pathophysiology of CAT. We propose that subsequent studies should be adequately sized to determine which panel of miRNAs best predicts VTE and CAT. Thereafter, validation studies using comparable patient populations are required to ultimately unveil whether miRNAs-as standalone or incorporated into existing risk models-are promising valuable VTE and CAT biomarkers.
Collapse
Affiliation(s)
- Rayna J S Anijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yen Nhi Nguyen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne C Cannegieter
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
6
|
Emerging Role of MicroRNA-30c in Neurological Disorders. Int J Mol Sci 2022; 24:ijms24010037. [PMID: 36613480 PMCID: PMC9819962 DOI: 10.3390/ijms24010037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) are a class of small non-coding RNAs that negatively regulate the expression of target genes by interacting with 3' untranslated regions of target mRNAs to induce mRNA degradation and translational repression. The miR-30 family members are involved in the development of many tissues and organs and participate in the pathogenesis of human diseases. As a key member of the miR-30 family, miR-30c has been implicated in neurological disorders such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and stroke. Mechanistically, miR-30c may act as a multi-functional regulator of different pathogenic processes such as autophagy, apoptosis, endoplasmic reticulum stress, inflammation, oxidative stress, thrombosis, and neurovascular function, thereby contributing to different disease states. Here, we review and discuss the biogenesis, gene regulation, and the role and mechanisms of action of miR-30c in several neurological disorders and therapeutic potential in clinics.
Collapse
|
7
|
Onuoha CP, Ipe J, Simpson E, Liu Y, Skaar T, Kreutz RP. MicroRNA
sequencing in patients with coronary artery disease – considerations for use as biomarker for thrombotic risk. Clin Transl Sci 2022; 15:1946-1958. [PMID: 35643946 PMCID: PMC9372418 DOI: 10.1111/cts.13307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/24/2022] [Accepted: 04/30/2022] [Indexed: 12/05/2022] Open
Abstract
MicroRNAs (miRNAs) are small RNAs integral in the regulation of gene expression. Analysis of circulating miRNA levels may identify patients with coronary artery disease (CAD) at risk for recurrent myocardial infarction (MI) after percutaneous coronary interventions (PCIs). Subjects with CAD were selected from the GENCATH cardiac catheterization biobank. Subjects with recurrent MI after PCI were compared with those without recurrent MI during follow‐up in the initial (n = 48) and replication cohort (n = 67). Next generation MiRNA sequencing was performed on plasma samples and whole blood samples fixed with PAXGENE tubes upon collection. Overall, 164 miRNAs derived from whole blood were differentially expressed in the replication cohort between subjects with and without recurrent MI events (p < 0.05), with 69 remaining significant after false‐discovery rate (FDR) correction. None of the miRNAs in plasma was significantly different by FDR among subjects with and without MI. Overall, correlation between direction of effects between plasma and whole blood assays was variable, and only two miRNAs were concordant and significant in both. Associations of miRNA with vascular disease, MI, and thrombosis were further explored. MiRNA profiling has potential as the future biomarker for disease prognosis and treatment response marker in secondary treatment of patients with CAD after PCI. Whole blood may be the preferred sample source as compared to plasma.
Collapse
Affiliation(s)
- Chimnonso P. Onuoha
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Joseph Ipe
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Edward Simpson
- Center for Medical Genomics Indiana University School of Medicine Indianapolis Indiana USA
| | - Yunlong Liu
- Center for Medical Genomics Indiana University School of Medicine Indianapolis Indiana USA
| | - Todd C. Skaar
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Rolf P. Kreutz
- Department of Medicine/Cardiology Indiana University School of Medicine Indianapolis Indiana USA
| |
Collapse
|
8
|
Selective inhibition of miRNA processing by a herpesvirus-encoded miRNA. Nature 2022; 605:539-544. [PMID: 35508655 DOI: 10.1038/s41586-022-04667-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Herpesviruses have mastered host cell modulation and immune evasion to augment productive infection, life-long latency and reactivation1,2. A long appreciated, yet undefined relationship exists between the lytic-latent switch and viral non-coding RNAs3,4. Here we identify viral microRNA (miRNA)-mediated inhibition of host miRNA processing as a cellular mechanism that human herpesvirus 6A (HHV-6A) exploits to disrupt mitochondrial architecture, evade intrinsic host defences and drive the switch from latent to lytic virus infection. We demonstrate that virus-encoded miR-aU14 selectively inhibits the processing of multiple miR-30 family members by direct interaction with the respective primary (pri)-miRNA hairpin loops. Subsequent loss of miR-30 and activation of the miR-30-p53-DRP1 axis triggers a profound disruption of mitochondrial architecture. This impairs induction of type I interferons and is necessary for both productive infection and virus reactivation. Ectopic expression of miR-aU14 triggered virus reactivation from latency, identifying viral miR-aU14 as a readily druggable master regulator of the herpesvirus lytic-latent switch. Our results show that miRNA-mediated inhibition of miRNA processing represents a generalized cellular mechanism that can be exploited to selectively target individual members of miRNA families. We anticipate that targeting miR-aU14 will provide new therapeutic options for preventing herpesvirus reactivations in HHV-6-associated disorders.
Collapse
|
9
|
Van Guilder GP, Preston CC, Munce TA, Faustino RS. Impacts of circulating microRNAs in exercise-induced vascular remodeling. Am J Physiol Heart Circ Physiol 2021; 320:H2401-H2415. [PMID: 33989080 DOI: 10.1152/ajpheart.00894.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cardiovascular adaptation underlies all athletic training modalities, with a variety of factors contributing to overall response during exercise-induced stimulation. In this regard the role of circulating biomarkers is a well-established and invaluable tool for monitoring cardiovascular function. Specifically, novel biomarkers such as circulating cell free DNA and RNA are now becoming attractive tools for monitoring cardiovascular function with the advent of next generation technologies that can provide unprecedented precision and resolution of these molecular signatures, paving the way for novel diagnostic and prognostic avenues to better understand physiological remodeling that occurs in trained versus untrained states. In particular, microRNAs are a species of regulatory RNAs with pleiotropic effects on multiple pathways in tissue-specific manners. Furthermore, the identification of cell free microRNAs within peripheral circulation represents a distal signaling mechanism that is just beginning to be explored via a diversity of molecular and bioinformatic approaches. This article provides an overview of the emerging field of sports/performance genomics with a focus on the role of microRNAs as novel functional diagnostic and prognostic tools, and discusses present knowledge in the context of athletic vascular remodeling. This review concludes with current advantages and limitations, touching upon future directions and implications for applying contemporary systems biology knowledge of exercise-induced physiology to better understand how disruption can lead to pathology.
Collapse
Affiliation(s)
- Gary P Van Guilder
- Vascular Protection Research Laboratory, Exercise & Sport Science Department, Western Colorado University, Gunnison, Colorado
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, Sioux Falls, South Dakota
| | - Thayne A Munce
- Environmental Influences on Health & Disease Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| |
Collapse
|
10
|
Elevation of Plasminogen Activator Inhibitor-1 promotes differentiation of Cancer Stem-like Cell state by Hepatitis C Virus infection. J Virol 2021; 95:JVI.02057-20. [PMID: 33627392 PMCID: PMC8139667 DOI: 10.1128/jvi.02057-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a critical factor that regulates protein synthesis and degradation. The increased PAI-1 levels are detectable in the serum of patients with chronic hepatitis C virus (HCV) liver disease. The differentiation state and motility of HCV-induced cancer stem-like cells (CSC) play a major role in severe liver disease progression. However, the role of PAI-1 in the pathological process of chronic liver diseases remains unknown. In this study, we determined how PAI-1 affects the differentiation of CSC state in hepatocytes upon HCV infection. We found that HCV infection induced the expression of PAI-1 while decreasing miR-30c expression in Huh7.5.1 cells. Similar results were obtained from isolated hepatocytes from humanized liver mice after HCV infection. Moreover, decreased miR-30c expression in HCV-infected hepatocytes was associated with the increased levels of PAI-1 mRNA and protein. Notably, the increased PAI-1 levels resulted in the activation of Protein Kinase B/AKT, a major mediator of cell proliferation, in HCV-infected hepatocytes along with the increased expression of CSC markers such as Human Differentiated Protein (CD) 133, Epithelial cell adhesion molecule (EpCAM), Octamer 4 (Oct4), Nanog, Cyclin D1, and MYC. Moreover, blockade of PAI-1 activity by miR-30c mimic and anti-PAI-1 mAb abrogated the AKT activation with decreased expression of CSC markers. Our findings suggest that HCV infection induces the CSC state via PAI-1-mediated AKT activation in hepatocytes. It implicates that the manipulation of PAI-1 activity could provide potential therapeutics to prevent the development of HCV-associated chronic liver diseases.IMPORTANCEThe progression of chronic liver disease by HCV infection is considered a major risk factor for hepatocellular carcinoma (HCC), one of the major causes of death from cancer. Recent studies have demonstrated that increased CSC properties in HCV-infected hepatocytes are associated with the progression of HCC. Since proteins and miRNAs production by HCV-infected hepatocytes can play various roles in physiological processes, investigating these factors can potentially lead to new therapeutic targets. However, the mechanism of HCV associated progression of hepatocytes to CSC remains unclear. Here we identify the roles of PAI-1 and miR-30c in the progression of CSC during HCV infection in hepatocytes. Our data shows that increased secretion of PAI-1 following HCV infection promotes this CSC state and activation of AKT. We report that the inhibition of PAI-1 by miR-30c mimic reduces HCV associated CSC properties in hepatocytes. Taken together, targeting this interaction of secreted PAI-1 and miR-30c in HCV-infected hepatocytes may provide a potential therapeutic intervention against the progression to chronic liver diseases and HCC.
Collapse
|
11
|
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]
|
12
|
Gbotosho OT, Kapetanaki MG, Kato GJ. The Worst Things in Life are Free: The Role of Free Heme in Sickle Cell Disease. Front Immunol 2021; 11:561917. [PMID: 33584641 PMCID: PMC7873693 DOI: 10.3389/fimmu.2020.561917] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hemolysis is a pathological feature of several diseases of diverse etiology such as hereditary anemias, malaria, and sepsis. A major complication of hemolysis involves the release of large quantities of hemoglobin into the blood circulation and the subsequent generation of harmful metabolites like labile heme. Protective mechanisms like haptoglobin-hemoglobin and hemopexin-heme binding, and heme oxygenase-1 enzymatic degradation of heme limit the toxicity of the hemolysis-related molecules. The capacity of these protective systems is exceeded in hemolytic diseases, resulting in high residual levels of hemolysis products in the circulation, which pose a great oxidative and proinflammatory risk. Sickle cell disease (SCD) features a prominent hemolytic anemia which impacts the phenotypic variability and disease severity. Not only is circulating heme a potent oxidative molecule, but it can act as an erythrocytic danger-associated molecular pattern (eDAMP) molecule which contributes to a proinflammatory state, promoting sickle complications such as vaso-occlusion and acute lung injury. Exposure to extracellular heme in SCD can also augment the expression of placental growth factor (PlGF) and interleukin-6 (IL-6), with important consequences to enthothelin-1 (ET-1) secretion and pulmonary hypertension, and potentially the development of renal and cardiac dysfunction. This review focuses on heme-induced mechanisms that are implicated in disease pathways, mainly in SCD. A special emphasis is given to heme-induced PlGF and IL-6 related mechanisms and their role in SCD disease progression.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maria G. Kapetanaki
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gregory J. Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
13
|
Nam DE, Seong HC, Hahn YS. Plasminogen Activator Inhibitor-1 and Oncogenesis in the Liver Disease. JOURNAL OF CELLULAR SIGNALING 2021; 2:221-227. [PMID: 34671766 PMCID: PMC8525887 DOI: 10.33696/signaling.2.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of cancer mortality worldwide. Chronic hepatic inflammation and fibrosis play a critical role in the development of HCC. Liver fibrosis develops as a result of response to injury such that a persistent and excessive wound healing response induces extracellular matrix (ECM) deposition leading to HCC. PAI-1 is a fibrinolysis inhibitor involved in regulating protein degradation and homeostasis while assisting wound healing. PAI-1 presents increased levels in various diseases such as fibrosis, cancer, obesity and metabolic syndrome. Moreover, PAI-1 has been extensively studied for developing potential therapies against fibrosis. In the present review, we summarize how PAI-1 affects oncogenesis during liver disease progression based on the recently published literatures. Although there are controversies regarding the role of PAI-1 and approaches to treatment, this review suggests that proper manipulation of PAI-1 activity could provide a novel therapeutic option on the development of chronic liver disease via modulation of cancer stem-like cells (CSCs) differentiation.
Collapse
Affiliation(s)
- Da-eun Nam
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
| | - Hae Chang Seong
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, USA
- Correspondence should be addressed to Young S. Hahn;
| |
Collapse
|
14
|
Danese E, Montagnana M, Gelati M, Lippi G. The Role of Epigenetics in the Regulation of Hemostatic Balance. Semin Thromb Hemost 2020; 47:53-62. [PMID: 33368118 DOI: 10.1055/s-0040-1718400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigenetics, a term conventionally used to explain the intricate interplay between genes and the environment, is now regarded as the fundament of developmental biology. Several lines of evidence garnered over the past decades suggest that epigenetic alterations, mostly encompassing DNA methylation, histone tail modifications, and generation of microRNAs, play an important, though still incompletely explored, role in both primary and secondary hemostasis. Epigenetic variations may interplay with platelet functions and their responsiveness to antiplatelet drugs, and they may also exert a substantial contribution in modulating the production and release into the bloodstream of proteins involved in blood coagulation and fibrinolysis. This emerging evidence may have substantial biological and clinical implications. An enhanced understanding of posttranscriptional mechanisms would help to clarify some remaining enigmatic issues in primary and secondary hemostasis, which cannot be thoughtfully explained by genetics or biochemistry alone. Increased understanding would also pave the way to developing innovative tests for better assessment of individual risk of bleeding or thrombosis. The accurate recognition of key epigenetic mechanisms in hemostasis would then contribute to identify new putative therapeutic targets, and develop innovative agents that could be helpful for preventing or managing a vast array of hemostasis disturbances.
Collapse
Affiliation(s)
- Elisa Danese
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Martina Montagnana
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Matteo Gelati
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Pourteymour Fard Tabrizi Z, Miraj S, Tahmasebian S, Ghasemi S. Plasma Levels of miR-27a, miR-130b, and miR-301a in Polycystic Ovary Syndrome. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:198-206. [PMID: 33274182 PMCID: PMC7703662 DOI: 10.22088/ijmcm.bums.9.3.198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 09/06/2020] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a gynecological endocrine disorder in women of reproductive age. There is adequate evidence that suggests several microRNAs (miRNAs) are of great importance for PCOS. It seems that dysregulated expression of miR-27a, miR-130b, and miR-301a are associated with PCOS. The aim of this study was to investigate whether plasma levels of these miRNAs are different between patients with PCOS and healthy controls. Fifty-three women with a definite diagnosis of PCOS, and 53 healthy controls were enrolled. MiRNAs expression levels in plasma were evaluated by real-time PCR. The diagnostic values of each miRNA were calculated by the receiver operating characteristic (ROC) curve and areas under the curves (AUC). The main clinical characteristics were not significantly different between the two groups. The circulating plasma expression levels of miR-27a and miR-301a had a significant increase (P = 0.0008 and P <0.0001, respectively) but miR-130b expression level decreased in the patient group (P <0.0001). The AUC for miR-27a, miR-130b, and miR-301a were 0.71, 0.77, and 0.66, respectively. A positive exponential was observed for miR-27a and miR-301a in multiple logistic regression. Changes in the plasma expressions of the studied miRNAs are likely to be associated with PCOS phenotypes. MiR-27a has a potential to serve as a diagnostic biomarker of PCOS.
Collapse
Affiliation(s)
- Zahra Pourteymour Fard Tabrizi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sepideh Miraj
- Department of Obstetrics and Gynecology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Tahmasebian
- School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
16
|
Xu Y, Liu Y, Cai R, He S, Dai R, Yang X, Kong B, Qin Z, Su Q. Long non-coding RNA CASC7 is associated with the pathogenesis of heart failure via modulating the expression of miR-30c. J Cell Mol Med 2020; 24:11500-11511. [PMID: 32860492 PMCID: PMC7576250 DOI: 10.1111/jcmm.15764] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022] Open
Abstract
MiRNAs can be used as promising diagnostic biomarkers of heart failure, while lncRNAs act as competing endogenous RNAs of miRNAs. In this study, we collected peripheral blood monocytes from subjects with or without HF to explore the association between certain lncRNAs, miRNAs and HF. Heart failure patients with preserved or reduced ejection fraction were recruited for investigation. ROC analysis was carried out to evaluate the diagnostic values of certain miRNAs and lncRNAs in HF. Luciferase assays were used to study the regulatory relationship between above miRNAs and lncRNAs. LncRNA overexpression was used to explore the effect of certain miRNAs in H9C2 cells. Expression of miR-30c was significantly decreased in the plasma and peripheral blood monocytes of patients suffering from heart failure, especially in these with reduced ejection fraction. On the contrary, the expression of lncRNA-CASC7 was remarkably increased in the plasma and peripheral blood monocytes of patients suffering from heart failure. Both miR-30c and lncRNA-CASC7 expression showed a promising efficiency as diagnostic biomarkers of heart failure. Luciferase assays indicated that miR-30c played an inhibitory role in lncRNA-CASC7 and IL-11 mRNA expression. Moreover, the overexpression of lncRNA-CASC7 suppressed the expression of miR-30c while evidently increasing the expression of IL-11 mRNA and protein in H9C2 cells. This study clarified the relationship among miR-30c, lncRNA-CASC7 and IL-11 expression and the risk of heart failure and showed that lncRNA-CASC7 is potentially involved in the pathogenesis of HF via modulating the expression of miR-30c.
Collapse
Affiliation(s)
- Yu‐li Xu
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Yang Liu
- Department of CardiologyThe Second People’s Hospital of Nanning CityThe Third Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ru‐ping Cai
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Shi‐rong He
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Ri‐xin Dai
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Xi‐heng Yang
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Bing‐hui Kong
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Zhen‐bai Qin
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qiang Su
- Department of CardiologyAffiliated Hospital of Guilin Medical UniversityGuilinChina
| |
Collapse
|
17
|
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
|
18
|
Chen Y, Yang C, Li Y, Chen L, Yang Y, Belguise K, Wang X, Lu K, Yi B. MiR145-5p inhibits proliferation of PMVECs via PAI-1 in experimental hepatopulmonary syndrome rat pulmonary microvascular hyperplasia. Biol Open 2019; 8:bio.044800. [PMID: 31649116 PMCID: PMC6899039 DOI: 10.1242/bio.044800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hepatopulmonary syndrome (HPS) is a triad of advanced liver disease, intrapulmonary vasodilatation and arterial hypoxemia. Increasing evidence shows that HPS is associated with pulmonary microvascular hyperplasia. The aim of this work was to investigate the underlying mechanism of miR-145 in regulating the proliferation of pulmonary microvascular endothelial cells (PMVECs) and angiogenesis in HPS via plasminogen activator inhibitor-1 (PAI-1). To test this, morphology score and number of pulmonary microvascular were assessed in lung tissues from rats with HPS by Hematoxylin and Eosin (H&E) staining. Expression levels of PAI-1 were assessed in lung tissues from HPS rats, as well as in PMVECs treated with HPS rat serum. We also selected the putative microRNA binding site on PAI-1 by bioinformatics analysis. Then, miR145-3p and miR145-5p expression levels in the lungs and PMVECs of rats were detected by qRT-PCR because miR145-5p is a microRNA binding site on PAI-1. In addition, the effects of miR-145-5p regulation on PAI-1 were examined by upregulation and downregulation of miR-145-5p and specific lentivirus transfection was used to overexpress and knockdown PAI-1 to assess PAI-1 function on PMVECs proliferation. Our data showed that levels of PAI-1 expression in lung tissue of rats increased significantly when rats were treated with common bile duct ligation. We found that levels of miR-145-5p were frequently downregulated in HPS tissues and cell lines, and overexpression of miR-145-5p dramatically inhibited PMVECs proliferation. We further verified PAI-1 as a novel and direct target of miR-145-5p in HPS. MiR-145-5p inhibits PAI-1 synthesis and the expression changes of PAI-1 directly affect the proliferation of PMVECs. We concluded that miR-145-5p negatively regulates PMVEC proliferation through PAI-1 expression. In addition, overexpression of miR-145-5p may prove beneficial as a therapeutic strategy for HPS treatment. Summary: Our findings provide proof of principle that microRNAs may be useful for the future development of novel therapeutic strategies in HPS.
Collapse
Affiliation(s)
- Yang Chen
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Congwen Yang
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yujie Li
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Chen
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yong Yang
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Karine Belguise
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, Université Paul Sabatier (UPS), 31062 Toulouse, France
| | - Xiaobo Wang
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, Université Paul Sabatier (UPS), 31062 Toulouse, France
| | - Kaizhi Lu
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yi
- Department of Anesthesia, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
19
|
McCann JV, Xiao L, Kim DJ, Khan OF, Kowalski PS, Anderson DG, Pecot CV, Azam SH, Parker JS, Tsai YS, Wolberg AS, Turner SD, Tatsumi K, Mackman N, Dudley AC. Endothelial miR-30c suppresses tumor growth via inhibition of TGF-β-induced Serpine1. J Clin Invest 2019; 129:1654-1670. [PMID: 30855280 DOI: 10.1172/jci123106] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/01/2019] [Indexed: 12/15/2022] Open
Abstract
In tumors, extravascular fibrin forms provisional scaffolds for endothelial cell (EC) growth and motility during angiogenesis. We report that fibrin-mediated angiogenesis was inhibited and tumor growth delayed following postnatal deletion of Tgfbr2 in the endothelium of Cdh5-CreERT2 Tgfbr2fl/fl mice (Tgfbr2iECKO mice). ECs from Tgfbr2iECKO mice failed to upregulate the fibrinolysis inhibitor plasminogen activator inhibitor 1 (Serpine1, also known as PAI-1), due in part to uncoupled TGF-β-mediated suppression of miR-30c. Bypassing TGF-β signaling with vascular tropic nanoparticles that deliver miR-30c antagomiRs promoted PAI-1-dependent tumor growth and increased fibrin abundance, whereas miR-30c mimics inhibited tumor growth and promoted vascular-directed fibrinolysis in vivo. Using single-cell RNA-Seq and a NanoString miRNA array, we also found that subtypes of ECs in tumors showed spectrums of Serpine1 and miR-30c expression levels, suggesting functional diversity in ECs at the level of individual cells; indeed, fresh EC isolates from lung and mammary tumor models had differential abilities to degrade fibrin and launch new vessel sprouts, a finding that was linked to their inverse expression patterns of miR-30c and Serpine1 (i.e., miR-30chi Serpine1lo ECs were poorly angiogenic and miR-30clo Serpine1hi ECs were highly angiogenic). Thus, by balancing Serpine1 expression in ECs downstream of TGF-β, miR-30c functions as a tumor suppressor in the tumor microenvironment through its ability to promote fibrin degradation and inhibit blood vessel formation.
Collapse
Affiliation(s)
- James V McCann
- Department of Cell Biology and Physiology, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lin Xiao
- Children's Cancer Institute, Kensington, New South Wales, Australia
| | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, Virginia, USA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT).,Department of Chemical Engineering
| | - Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT)
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT).,Department of Chemical Engineering.,Harvard-MIT Division of Health Sciences and Technology, and.,Institute for Medical Engineering and Science, MIT, Cambridge, Massachusetts, USA
| | - Chad V Pecot
- Lineberger Comprehensive Cancer Center.,School of Medicine
| | | | - Joel S Parker
- Lineberger Comprehensive Cancer Center.,School of Medicine.,Department of Genetics, and
| | | | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, UNC McAllister Heart Institute, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephen D Turner
- Department of Public Health Sciences, and.,Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kohei Tatsumi
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, Virginia, USA.,Emily Couric Cancer Center, The University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
20
|
Wu M, Liang G, Duan H, Yang X, Qin G, Sang N. Synergistic effects of sulfur dioxide and polycyclic aromatic hydrocarbons on pulmonary pro-fibrosis via mir-30c-1-3p/ transforming growth factor β type II receptor axis. CHEMOSPHERE 2019; 219:268-276. [PMID: 30543962 DOI: 10.1016/j.chemosphere.2018.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/23/2018] [Accepted: 12/02/2018] [Indexed: 06/09/2023]
Abstract
SO2 and PAHs are well-known pollutants of coal burning and significant contributors to haze episodes. The purpose of the study is to determine whether the combined effects of SO2 and BaP are synergetic and to investigate the pro-fibrotic influences and possible mechanism from the aspect of microRNAs. In the present study cellular metabolic activity of BEAS-2B was assessed using MTT probe. C57BL/6 mice were exposed to BaP (40 mg/kg b.w.) for 5 days or SO2 (7 mg/m3) inhalation for 4 weeks alone or together. Lung tissues were processed for histology to assess pulmonary fibrosis. The protein level of pulmonary pro-fibrotic genes (Col1a1, Col3a1, alpha-SMA, fibronectin) and TGFβR2 were analyzed by Western blot and immunofluorescence in vivo and in vitro. Furthermore, we clarified that the microRNA expression of mir-30c-1-3p by real-time RT-PCR. The luciferase reporter assay was used to determine the binding sites of mir-30c-1-3p in the 3'-UTR of TGFβR2. It was confirmed that SO2 and BaP acted together to produce synergistic effects in cellular metabolic activity. Coexisting of SO2 and BaP increased the protein expression of pro-fibrotic genes and TGFβR2 and decreased mir-30c-1-3p in vivo and in vitro. Dual-luciferase reporter gene assays showed that TGFβR2 was a validated target of mir-30c-1-3p. All above results demonstrated that mir-30c-1-3p was involved in the synergistic pro-fibrotic effects of SO2 and BaP in lung via targeting TGFβR2. This work implies the potential risk of pulmonary fibrosis from the co-existence of SO2 and PAHs and provides new insights into the molecular markers for relevant diseases.
Collapse
Affiliation(s)
- Meiqiong Wu
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Gang Liang
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Huiling Duan
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Xiaofeng Yang
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Guohua Qin
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China.
| | - Nan Sang
- College of Environmental Science and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China.
| |
Collapse
|
21
|
Feng C, Song C, Ning Z, Ai B, Wang Q, Xu Y, Li M, Bai X, Zhao J, Liu Y, Li X, Zhang J, Li C. ce-Subpathway: Identification of ceRNA-mediated subpathways via joint power of ceRNAs and pathway topologies. J Cell Mol Med 2018; 23:967-984. [PMID: 30421585 PMCID: PMC6349186 DOI: 10.1111/jcmm.13997] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/28/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022] Open
Abstract
Competing endogenous RNAs (ceRNAs) represent a novel mechanism of gene regulation that may mediate key subpathway regions and contribute to the altered activities of pathways. However, the classical methods used to identify pathways fail to specifically consider ceRNAs within the pathways and key regions impacted by them. We proposed a powerful strategy named ce-Subpathway for the identification of ceRNA-mediated functional subpathways. It provided an effective level of pathway analysis via integrating ceRNAs, differentially expressed (DE) genes and their key regions within the given pathways. We respectively analysed one pulmonary arterial hypertension (PAH) and one myocardial infarction (MI) data sets and demonstrated that ce-Subpathway could identify many subpathways whose corresponding entire pathways were ignored by those non-ceRNA-mediated pathway identification methods. And these pathways have been well reported to be associated with PAH/MI-related cardiovascular diseases. Further evidence showed reliability of ceRNA interactions and robustness/reproducibility of the ce-Subpathway strategy by several data sets of different cancers, including breast cancer, oesophageal cancer and colon cancer. Survival analysis was finally applied to illustrate the clinical application value of the ceRNA-mediated functional subpathways using another data sets of pancreatic cancer. Comprehensive analyses have shown the power of a joint ceRNAs/DE genes and subpathway strategy based on their topologies.
Collapse
Affiliation(s)
- Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Chao Song
- Department of Pharmacology, Daqing Campus, Harbin Medical University, Daqing, China
| | - Ziyu Ning
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Bo Ai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qiuyu Wang
- School of Nursing, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yong Xu
- The fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Meng Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuefeng Bai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianmei Zhao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yuejuan Liu
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuecang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| |
Collapse
|
22
|
Parchem JG, Kanasaki K, Kanasaki M, Sugimoto H, Xie L, Hamano Y, Lee SB, Gattone VH, Parry S, Strauss JF, Garovic VD, McElrath TF, Lu KH, Sibai BM, LeBleu VS, Carmeliet P, Kalluri R. Loss of placental growth factor ameliorates maternal hypertension and preeclampsia in mice. J Clin Invest 2018; 128:5008-5017. [PMID: 30179860 PMCID: PMC6205389 DOI: 10.1172/jci99026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/28/2018] [Indexed: 12/28/2022] Open
Abstract
Preeclampsia remains a clinical challenge due to its poorly understood pathogenesis. A prevailing notion is that increased placental production of soluble fms-like tyrosine kinase-1 (sFlt-1) causes the maternal syndrome by inhibiting proangiogenic placental growth factor (PlGF) and VEGF. However, the significance of PlGF suppression in preeclampsia is uncertain. To test whether preeclampsia results from the imbalance of angiogenic factors reflected by an abnormal sFlt-1/PlGF ratio, we studied PlGF KO (Pgf-/-) mice and noted that the mice did not develop signs or sequelae of preeclampsia despite a marked elevation in circulating sFLT-1. Notably, PlGF KO mice had morphologically distinct placentas, showing an accumulation of junctional zone glycogen. We next considered the role of placental PlGF in an established model of preeclampsia (pregnant catechol-O-methyltransferase-deficient [COMT-deficient] mice) by generating mice with deletions in both the Pgf and Comt genes. Deletion of placental PlGF in the context of COMT loss resulted in a reduction in maternal blood pressure and increased placental glycogen, indicating that loss of PlGF might be protective against the development of preeclampsia. These results identify a role for PlGF in placental development and support a complex model for the pathogenesis of preeclampsia beyond an angiogenic factor imbalance.
Collapse
Affiliation(s)
- Jacqueline G Parchem
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Megumi Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Liang Xie
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuki Hamano
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Soo Bong Lee
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Vincent H Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samuel Parry
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Vesna D Garovic
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas F McElrath
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Karen H Lu
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Liu B, Shyr Y, Cai J, Liu Q. Interplay between miRNAs and host genes and their role in cancer. Brief Funct Genomics 2018; 18:255-266. [PMID: 30785618 PMCID: PMC6609535 DOI: 10.1093/bfgp/elz002] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/21/2018] [Accepted: 01/23/2019] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding functional RNAs that post-transcriptionally regulate gene expression. They play essential roles in nearly all biological processes including cell development and differentiation, DNA damage repair, cell death as well as intercellular communication. They are highly involved in cancer, acting as tumor suppressors and/or promoters to modulate cell proliferation, epithelial-mesenchymal transition and tumor invasion and metastasis. Recent studies have shown that more than half of miRNAs are located within protein-coding or non-coding genes. Intragenic miRNAs and their host genes either share the promoter or have independent transcription. Meanwhile, miRNAs work as partners or antagonists of their host genes by fine-tuning their target genes functionally associated with host genes. This review outlined the complicated relationship between intragenic miRNAs and host genes. Focusing on miRNAs known as oncogenes or tumor suppressors in specific cancer types, it studied co-expression relationships between these miRNAs and host genes in the cancer types using TCGA data sets, which validated previous findings and revealed common, tumor-specific and even subtype-specific patterns. These observations will help understand the function of intragenic miRNAs and further develop miRNA therapeutics in cancer.
Collapse
Affiliation(s)
- Baohong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jianping Cai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
24
|
Arroyo AB, de Los Reyes-García AM, Teruel-Montoya R, Vicente V, González-Conejero R, Martínez C. microRNAs in the haemostatic system: More than witnesses of thromboembolic diseases? Thromb Res 2018; 166:1-9. [PMID: 29649766 DOI: 10.1016/j.thromres.2018.03.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) are small endogenous RNAs that post-transcriptionally regulate gene expression. In the last few years, these molecules have been implicated in the regulation of haemostasis, and an increasing number of studies have investigated their relationship with the development of thrombosis. In this review, we discuss the latest developments regarding the role of miRNAs in the regulation of platelet function and secondary haemostasis. We also discuss the genetic and environmental factors that regulate miRNAs. Finally, we address the potential use of miRNAs as prognostic and diagnostic tools in thrombosis.
Collapse
Affiliation(s)
- Ana B Arroyo
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Ascensión M de Los Reyes-García
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Raúl Teruel-Montoya
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain; Red CIBERER CB15/00055, Murcia, Spain
| | - Vicente Vicente
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain; Red CIBERER CB15/00055, Murcia, Spain
| | - Rocío González-Conejero
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain.
| | - Constantino Martínez
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, Murcia, Spain.
| |
Collapse
|
25
|
Gao J, Ma X, Zhang Y, Guo M, Shi D. The role of microRNAs in prethrombotic status associated with coronary artery disease. Thromb Haemost 2017; 117:429-436. [DOI: 10.1160/th16-07-0503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022]
Abstract
SummaryThe acute cardiovascular events following thrombus formation is a primary cause of morbidity and mortality of patients with coronary artery disease (CAD). Numerous studies have shown that a prethrombotic status, which can be defined as an imbalance between the procoagulant and anticoagulant conditions, would exist for a period of time before thrombogenesis. Therefore, early diagnosis and intervention of prethrombotic status are important for reducing acute cardiovascular events. However, none of prethrombotic indicators have been identified as golden standard for diagnosis of prethrombotic status to date. MicroRNAs (miRNAs), a class of short non-coding RNAs, have been shown to be involved in pathophysiologic processes related to prethrombotic status, such as endothelial dysfunction, platelet activation, impaired fibrinolysis and elevated procoagulant factors, etc. Owing to their multiple and fine-tuning impacts on gene expression, miRNAs raise a novel understanding in the underlying mechanism of prethrombotic status. This review aims to discuss the role of miRNAs in prethrombotic status, especially the differently expressed miRNAs in CAD, which may be meaningful for developing promising diagnostic biomarkers and therapeutic strategies for CAD patients in future.
Collapse
|
26
|
de Mendonça L, Felix NS, Blanco NG, Da Silva JS, Ferreira TP, Abreu SC, Cruz FF, Rocha N, Silva PM, Martins V, Capelozzi VL, Zapata-Sudo G, Rocco PRM, Silva PL. Mesenchymal stromal cell therapy reduces lung inflammation and vascular remodeling and improves hemodynamics in experimental pulmonary arterial hypertension. Stem Cell Res Ther 2017; 8:220. [PMID: 28974252 PMCID: PMC5627397 DOI: 10.1186/s13287-017-0669-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
Background Experimental research has reported beneficial effects of mesenchymal stromal cell (MSC) therapy in pulmonary arterial hypertension (PAH). However, these studies either were based on prophylactic protocols or assessed basic remodeling features without evaluating possible mechanisms. We analyzed the effects of MSC therapy on lung vascular remodeling and hemodynamics and its possible mechanisms of action in monocrotaline (MCT)-induced PAH. Methods Twenty-eight Wistar rats were randomly divided into two groups. In the PAH group, animals received MCT 60 mg/kg intraperitoneally, while a control group received saline (SAL) instead. On day 14, both groups were further randomized to receive 105 adipose-derived MSCs or SAL intravenously (n = 7/group). On day 28, right ventricular systolic pressure (RVSP) and the gene expression of mediators associated with apoptosis, inflammation, fibrosis, Smad-1 levels, cell proliferation, and endothelial–mesenchymal transition were determined. In addition, lung histology (smooth muscle cell proliferation and plexiform-like injuries), CD68+ and CD163+ macrophages, and plasma levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) were evaluated. Results In the PAH group, adipose-derived MSCs, compared to SAL, reduced mean RVSP (29 ± 1 vs 39 ± 2 mmHg, p < 0.001), lung tissue collagen fiber content, smooth muscle cell proliferation, CD68+ macrophages, interleukin-6 expression, and the antiapoptotic mediators Bcl-2 and survivin. Conversely, expression of the proapoptotic mediator procaspase-3 and plasma VEGF increased, with no changes in PDGF. In the pulmonary artery, MSCs dampened the endothelial–mesenchymal transition. Conclusion In MCT-induced PAH, MSC therapy reduced lung vascular remodeling, thus improving hemodynamics. These beneficial effects were associated with increased levels of proapoptotic markers, mesenchymal-to-endothelial transition, reduced cell proliferation markers, and inflammation due to a shift away from the M1 phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0669-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lucas de Mendonça
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nathane S Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Natália G Blanco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Jaqueline S Da Silva
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tatiana P Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nazareth Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Physiology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Patrícia M Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Vanessa Martins
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Vera L Capelozzi
- Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Gizele Zapata-Sudo
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
27
|
Chen X, Jiang Y, Pan D. miR-30c may serve a role in endometriosis by targeting plasminogen activator inhibitor-1. Exp Ther Med 2017; 14:4846-4852. [PMID: 29201189 PMCID: PMC5704271 DOI: 10.3892/etm.2017.5145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/28/2016] [Indexed: 01/03/2023] Open
Abstract
The present study aimed to investigate the role of miR-30c in endometriosis (EMs) and the underlying mechanism. The expression of miR-30c and plasminogen activator inhibitor type 1 (PAI-1) mRNA in EMs tissues was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and the expression of PAI-1 protein was detected by western blot analysis. The proliferation, migration, invasion and adhesion of endometrial stromal cells (ESCs) in different groups transfected with miR-30c mimic or inhibitor were compared. It was demonstrated that miR-30c expression in ectopic and eutopic endometriosis tissues were significantly lower than in normal endometrial tissue. However, PAI-1 mRNA expression in ectopic and eutopic endometrial tissues was higher than in normal endometrial tissues. Furthermore, the expression of PAI-1 protein was higher in ectopic and eutopic endometrosis tissues than in normal tissues. RT-qPCR results indicated that miR-30c expression was significantly increased or decreased in ESCs following transfection of mimic or inhibitor of miR-30c, respectively. Overexpression of miR-30c repressed the expression of PAI-1 mRNA and protein, while inhibition of miR-30c upregulated the expression of PAI-1 in ESCs. In addition, the invasion, migration, proliferation and adhesion of ESCs was repressed following the overexpression of miR-30c, whereas they were promoted when miR-30c expression was downregulated. The results of the present study indicated that miR-30c serves an important role in the development and progression of EMs by regulating the expression of PAI-1.
Collapse
Affiliation(s)
- Xiaoli Chen
- Department of Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yan Jiang
- Department of Gynecology and Obstetrics, Tengzhou Maternity and Child Care Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Dianling Pan
- Department of Gynecology and Obstetrics, Jinan Maternity and Child Care Hospital, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
28
|
Egan SM, Karasik E, Ellis L, Gollnick SO. miR-30e* is overexpressed in prostate cancer and promotes NF-κB-mediated proliferation and tumor growth. Oncotarget 2017; 8:67626-67638. [PMID: 28978058 PMCID: PMC5620198 DOI: 10.18632/oncotarget.18795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/02/2017] [Indexed: 12/30/2022] Open
Abstract
According to the CDC prostate cancer (CaP) has the highest incidence and second highest mortality rate amongst cancers in American men. Constitutive NF-κB activation is a hallmark of CaP and this pathway drives many pro-tumorigenic characteristics of CaP cells, including cell proliferation and survival. An activated NF-κB gene signature is predictive of CaP progression and biochemical recurrence following therapeutic intervention. However, the mechanisms that perpetuate NF-κB activation are incompletely understood. Genes that control NF-κB activity are rarely mutated in CaP suggesting that epigenetic mechanisms may contribute to constitutive NF-κB activation. microRNAs (miRs) epigenetically regulate many genes involved with NF-κB activation. IκBα is a direct inhibitor of NF-κB; it binds to and sequesters NF-κB in the cytoplasm resulting in functional inhibition. IκBα is a target gene of miR-30e* yet the expression and oncological impact of miR-30e* in CaP is unknown. We report that miR-30e* expression is elevated in multiple murine models of CaP and is most pronounced in late stage disease. miR-30e* drives CaP proliferation and tumor growth through inhibition of IκBα, which results in chronic activation of NF-κB. Additionally, we show that inhibition of miR-30e* improves chemotherapeutic control of CaP. Thus, miR-30e* may prove to be a novel clinical target whose inhibition leads to decreased CaP cell proliferation and sensitization of CaP cells to chemotherapeutics.
Collapse
Affiliation(s)
- Shawn M. Egan
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Leigh Ellis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sandra O. Gollnick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| |
Collapse
|
29
|
Placenta growth factor mediated gene regulation in sickle cell disease. Blood Rev 2017; 32:61-70. [PMID: 28823762 DOI: 10.1016/j.blre.2017.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 01/07/2023]
Abstract
Sickle cell anemia (SCA) is an autosomal recessive disorder caused by mutation in the β-globin gene. Pulmonary hypertension (PH), a complication of SCA, results in severe morbidity and mortality. PH is a multifactorial disease: systemic vasculopathy, pulmonary vasoconstriction, and endothelial dysfunction and remodeling. Placenta growth factor (PlGF), an angiogenic growth factor, elaborated from erythroid cells, has been shown to contribute to inflammation, pulmonary vasoconstriction and airway hyper-responsiveness (AH) in mouse models of sickle cell disease. In this review, we summarize the cell-signaling mechanism(s) by which PlGF regulates the expression of genes involved in inflammation, PH and AH in cell culture and corroborate these findings in mouse models of SCA and in individuals with SCA. The role of microRNAs (miRNAs) in the post-transcriptional regulation of these genes is presented and how these miRNAs located in their host genes are transcriptionally regulated. An understanding of the transcriptional regulation of these miRNAs provides a new therapeutic approach to ameliorate the clinical manifestations of SCA.
Collapse
|
30
|
Katta S, Karnewar S, Panuganti D, Jerald MK, Sastry BKS, Kotamraju S. Mitochondria-targeted esculetin inhibits PAI-1 levels by modulating STAT3 activation and miR-19b via SIRT3: Role in acute coronary artery syndrome. J Cell Physiol 2017; 233:214-225. [PMID: 28213977 DOI: 10.1002/jcp.25865] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 12/13/2022]
Abstract
In this study we explored the microRNAs responsible for the regulation of PAI-1 during LPS-stimulated inflammation in human aortic endothelial cells and subsequently studied the effect of a newly synthesized mitochondria-targeted esculetin (Mito-Esc) that was shown for its anti-atherosclerotic potential, in modulating PAI-1 levels and its targeted miRs during angiotensin-II-induced atherosclerosis in ApoE-/- mice. LPS-stimulated PAI-1 was accompanied with an upregulation of miR-19b and down-regulation of miR-30c. These effects of LPS on PAI-1 were reversed in the presence of both parent esculetin and Mito-Esc. However, the effect of Mito-Esc was more pronounced in the regulation of PAI-1. In addition, LPS-stimulated PAI-1 expression was significantly decreased in cells treated with Anti-miR-19b, thereby suggesting that miR-19b co-expression plays a key role in PAI-1 regulation. The results also show that incubation of cells with Stattic, an inhibitor of STAT-3, inhibited LPS-stimulated PAI-1 expression. Interestingly, knockdown of SIRT3, a mitochondrial biogenetic marker, enhanced PAI-1 levels via modulation of miR-19b and -30c. Mito-Esc treatment significantly inhibited Ang-II-induced PAI-1, possibly via altering miR-19b and 30c in ApoE-/- mice. The association between PAI-1, miR-19b and -30c were further confirmed in plasma and microparticles isolated from patients suffering from acute coronary syndrome of various degrees. Taken together, LPS-induced PAI-1 involves co-expression of miR-19b and down regulation of miR-30c, and Mito-Esc treatment by modulating miR-19b and miR-30c through SIRT3 activation, inhibits PAI-1 levels that, in part, contribute to its anti-atherosclerotic effects. Moreover, there exists a strong positive correlation between miR-19b and PAI-1 in patients suffering from ST-elevated myocardial infarction.
Collapse
Affiliation(s)
- Sujana Katta
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Santosh Karnewar
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research, Training and Development Complex, CSIR Campus, Taramani, Chennai, Tamilnadu, India
| | - Devayani Panuganti
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | | | - B K S Sastry
- Department of Cardiology, CARE Hospitals and CARE Foundation, Hyderabad, India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research, Training and Development Complex, CSIR Campus, Taramani, Chennai, Tamilnadu, India
| |
Collapse
|
31
|
Hyperglycaemia-induced reciprocal changes in miR-30c and PAI-1 expression in platelets. Sci Rep 2016; 6:36687. [PMID: 27819307 PMCID: PMC5098184 DOI: 10.1038/srep36687] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/18/2016] [Indexed: 02/01/2023] Open
Abstract
Type 2 diabetic mellitus (DM2) is associated with accelerated thrombotic complications and is characterized by high levels of plasminogen activator inhibitor-1 (PAI-1). Recent studies show that human platelets have high levels of miR-30c and synthesize considerable active PAI-1. The underlying mechanism of how PAI-1 expression is upregulated in DM2 is poorly understood. We now report that hyperglycaemia-induced repression of miR-30c increases PAI-1 expression and thrombus formation in DM2. Bioinformatic analysis and identification of miRNA targets were assessed using luciferase assays, quantitative real-time PCR and western blots invitro and in vivo. The changes in miR-30c and PAI-1 levels were identified in platelets from healthy and diabetic individuals. We found that miR-30c directly targeted the 3′ UTR of PAI-1 and negatively regulated its expression. miR-30c was negatively correlated with glucose and HbA1c levels in DM2. In HFD-fed diabetic mice, increasing miR-30c expression by lenti-miR-30c significantly decreased the PAI-1 expression and prolonged the time to occlusion in an arterial thrombosis model. Platelet depletion/reinfusion experiments generating mice with selective ablation of PAI-1 demonstrate a major contribution by platelet-derived PAI-1 in the treatment of lenti-miR-30c to thrombus formation. These results provide important implications regarding the regulation of fibrinolysis by platelet miRNA under diabetic mellitus.
Collapse
|
32
|
Lee CY, Shin S, Lee J, Seo HH, Lim KH, Kim H, Choi JW, Kim SW, Lee S, Lim S, Hwang KC. MicroRNA-Mediated Down-Regulation of Apoptosis Signal-Regulating Kinase 1 (ASK1) Attenuates the Apoptosis of Human Mesenchymal Stem Cells (MSCs) Transplanted into Infarcted Heart. Int J Mol Sci 2016; 17:E1752. [PMID: 27775615 PMCID: PMC5085777 DOI: 10.3390/ijms17101752] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 09/19/2016] [Accepted: 10/14/2016] [Indexed: 11/17/2022] Open
Abstract
Stem cell therapy using adult stem cells, such as mesenchymal stem cells (MSCs) has produced some promising results in treating the damaged heart. However, the low survival rate of MSCs after transplantation is still one of the crucial factors that limit the therapeutic effect of stem cells. In the damaged heart, oxidative stress due to reactive oxygen species (ROS) production can cause the death of transplanted MSCs. Apoptosis signal-regulating kinase 1 (ASK1) has been implicated in the development of oxidative stress-related pathologic conditions. Thus, we hypothesized that down-regulation of ASK1 in human MSCs (hMSCs) might attenuate the post-transplantation death of MSCs. To test this hypothesis, we screened microRNAs (miRNAs) based on a miRNA-target prediction database and empirical data and investigated the anti-apoptotic effect of selected miRNAs on human adipose-derived stem cells (hASCs) and on rat myocardial infarction (MI) models. Our data indicated that miRNA-301a most significantly suppressed ASK1 expression in hASCs. Apoptosis-related genes were significantly down-regulated in miRNA-301a-enriched hASCs exposed to hypoxic conditions. Taken together, these data show that miRNA-mediated down-regulation of ASK1 protects MSCs during post-transplantation, leading to an increase in the efficacy of MSC-based cell therapy.
Collapse
Affiliation(s)
- Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, 03722 Seoul, Korea.
| | - Sunhye Shin
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, 03722 Seoul, Korea.
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 03722 Seoul, Korea.
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 03722 Seoul, Korea.
| | - Kyu Hee Lim
- Department of Veterinary Medicine, Chonbuk National University, 54896 Jeonju, Korea.
| | - Hyemin Kim
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, 03722 Seoul, Korea.
| | - Jung-Won Choi
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, 25601 Gangwon-do, Korea.
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, 25601 Gangwon-do, Korea.
| | - Seahyung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, 25601 Gangwon-do, Korea.
- Catholic Kwandong University, International St. Mary's Hospital, 22711 Incheon, Korea.
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, 25601 Gangwon-do, Korea.
- Catholic Kwandong University, International St. Mary's Hospital, 22711 Incheon, Korea.
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, 25601 Gangwon-do, Korea.
- Catholic Kwandong University, International St. Mary's Hospital, 22711 Incheon, Korea.
| |
Collapse
|
33
|
miR-101 sensitizes K562 cell line to imatinib through Jak2 downregulation and inhibition of NF-κB target genes. Tumour Biol 2016; 37:14117-14128. [DOI: 10.1007/s13277-016-5205-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022] Open
|
34
|
Wen M, Men R, Liu X, Yang L. Involvement of miR-30c in hepatic stellate cell activation through the repression of plasminogen activator inhibitor-1. Life Sci 2016; 155:21-8. [DOI: 10.1016/j.lfs.2016.04.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 02/06/2023]
|
35
|
Tay J, Tiao J, Hughes Q, Gilmore G, Baker R. Therapeutic Potential of miR-494 in Thrombosis and Other Diseases: A Review. Aust J Chem 2016. [DOI: 10.1071/ch16020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Functional nucleic acids, such as microRNAs (miRNAs), have been implicated in the pathophysiology of many diseases. The miRNA expression profiles of various cancers including haematological malignancies are well defined, but the role of miRNAs in haemostasis and the regulation of coagulation is poorly understood. We identified that miR-494 is oestrogen responsive and directly targets the anticoagulant protein, Protein S, as a mechanism for acquiring Protein S deficiency under high oestrogenic conditions such as during pregnancy and oral contraceptive use. Furthermore, previous studies have also characterised miR-494 to be involved in many biological processes. This paper reviews the current knowledge in the role of miRNAs in regulating haemostatic proteins and the known biological functions of miR-494, highlighting miR-494 as an emerging therapeutic target, with an overview of the strategy we have employed in identifying functional nucleic acids such as miRNAs that target haemostatic factors and the therapeutic potential of miR-494-directed therapy for the treatment of thrombotic disorders.
Collapse
|
36
|
Lu Y, Gao W, Zhang C, Wen S, Huangfu H, Kang J, Wang B. Hsa-miR-301a-3p Acts as an Oncogene in Laryngeal Squamous Cell Carcinoma via Target Regulation of Smad4. J Cancer 2015; 6:1260-75. [PMID: 26640587 PMCID: PMC4643083 DOI: 10.7150/jca.12659] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/28/2015] [Indexed: 01/07/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the second most common malignant head and neck squamous cell carcinoma. Exploring the molecular indicators of malignant behavior will enhance our knowledge of this type cancer and provide novel options for its prevention, diagnosis, and treatment. MicroRNA might exert regulatory roles as oncogenes or anti-oncogenes. We studied the expression of miR-301a-3p in LSCC tissues and cell lines and conducted a functional analysis of miR-301a-3p to confirm if miR-301a-3p functions as an oncogene in LSCC. We found Smad4 to be one of the potential target genes of miR-301a-3p, and it functioned as a tumor suppressor in LSCC. Hsa-miR-301a-3p participated in the epithelial-mesenchymal transition (EMT) process, which is considered to be linked to the process of LSCC development. Our present findings indicate that miR-301a-3p acts as an oncogene by directly regulating the anti-oncogene Smad4, thereby playing a role in the occurrence and development of LSCC. The present findings are expected to help in the development of novel targets for the prevention and treatment of LSCC.
Collapse
Affiliation(s)
- Yan Lu
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 3. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Wei Gao
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 2. Shanxi Key Laboratory of Otolaryngology Head & Neck Cancer, Taiyuan, Shanxi, 030001, China
| | - Chunming Zhang
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 2. Shanxi Key Laboratory of Otolaryngology Head & Neck Cancer, Taiyuan, Shanxi, 030001, China
| | - Shuxin Wen
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 2. Shanxi Key Laboratory of Otolaryngology Head & Neck Cancer, Taiyuan, Shanxi, 030001, China
| | - Hui Huangfu
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 2. Shanxi Key Laboratory of Otolaryngology Head & Neck Cancer, Taiyuan, Shanxi, 030001, China
| | - Jian Kang
- 3. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Binquan Wang
- 1. Department of Otolaryngology, Head & Neck Surgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, 030001, China ; 2. Shanxi Key Laboratory of Otolaryngology Head & Neck Cancer, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
37
|
Gonsalves CS, Li C, Malik P, Tahara SM, Kalra VK. Peroxisome proliferator-activated receptor-α-mediated transcription of miR-301a and miR-454 and their host gene SKA2 regulates endothelin-1 and PAI-1 expression in sickle cell disease. Biosci Rep 2015; 35:e00275. [PMID: 26460070 PMCID: PMC4672349 DOI: 10.1042/bsr20150190] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/01/2015] [Accepted: 10/09/2015] [Indexed: 01/24/2023] Open
Abstract
Endothelin-1 (ET-1) and plasminogen activator inhibitor-1 (PAI-1) play important roles in pulmonary hypertension (PH) in sickle cell disease (SCD). Our previous studies show higher levels of placenta growth factor (PlGF) in SCD correlate with increased plasma levels of ET-1, PAI-1, and other physiological markers of PH. PlGF-mediated ET-1 and PAI-1 expression occurs via activation of hypoxia-inducible factor-1α (HIF-1α). However, relatively little is understood regarding post-transcriptional regulation of PlGF-mediated expression of ET-1 and PAI-1. Herein, we show PlGF treatment of endothelial cells reduced levels of miR-301a and miR-454 from basal levels. In addition, both miRNAs targeted the 3'-UTRs of ET-1 and PAI-1 mRNAs. These results were corroborated in the mouse model of SCD [Berkeley sickle mice (BK-SS)] and in SCD subjects. Plasma levels of miR-454 in SCD subjects were significantly lower compared with unaffected controls, which correlated with higher plasma levels of both ET-1 and PAI-1. Moreover, lung tissues from BK-SS mice showed significantly reduced levels of pre-miR-301a and concomitantly higher levels of ET-1 and PAI-1. Furthermore, we show that miR-301a/miR-454 located in the spindle and kinetochore-associated protein-2 (SKA2) transcription unit was co-transcriptionally regulated by both HIF-1α and peroxisome proliferator-activated receptor-α (PPAR-α) as demonstrated by SKA2 promoter mutational analysis and ChIP. Finally we show that fenofibrate, a PPAR-α agonist, increased the expression of miR-301a/miR-454 and SKA2 in human microvascular endothelial cell line (HMEC) cells; the former were responsible for reduced expression of ET-1 and PAI-1. Our studies provide a potential therapeutic approach whereby fenofibrate-induced miR-301a/miR-454 expression can ameliorate PH and lung fibrosis by reduction in ET-1 and PAI-1 levels in SCD.
Collapse
MESH Headings
- Anemia, Sickle Cell/complications
- Anemia, Sickle Cell/drug therapy
- Anemia, Sickle Cell/genetics
- Anemia, Sickle Cell/pathology
- Animals
- Cell Line
- Chromosomal Proteins, Non-Histone/biosynthesis
- Chromosomal Proteins, Non-Histone/genetics
- Endothelin-1/biosynthesis
- Endothelin-1/genetics
- Fenofibrate/administration & dosage
- Gene Expression Regulation/drug effects
- Humans
- Hypertension, Pulmonary/complications
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Mice
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- PPAR alpha/antagonists & inhibitors
- PPAR alpha/genetics
- PPAR alpha/metabolism
- Placenta Growth Factor
- Plasminogen Activator Inhibitor 1/biosynthesis
- Plasminogen Activator Inhibitor 1/genetics
- Pregnancy Proteins/genetics
- Pregnancy Proteins/metabolism
- Promoter Regions, Genetic
Collapse
Affiliation(s)
- Caryn S Gonsalves
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Chen Li
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, U.S.A
| | - Stanley M Tahara
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Vijay K Kalra
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, U.S.A.
| |
Collapse
|
38
|
Demolli S, Doebele C, Doddaballapur A, Lang V, Fisslthaler B, Chavakis E, Vinciguerra M, Sciacca S, Henschler R, Hecker M, Savant S, Augustin HG, Kaluza D, Dimmeler S, Boon RA. MicroRNA-30 mediates anti-inflammatory effects of shear stress and KLF2 via repression of angiopoietin 2. J Mol Cell Cardiol 2015; 88:111-9. [PMID: 26456066 DOI: 10.1016/j.yjmcc.2015.10.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 01/13/2023]
Abstract
MicroRNAs are endogenously expressed small noncoding RNAs that regulate gene expression. Laminar blood flow induces atheroprotective gene expression in endothelial cells (ECs) in part by upregulating the transcription factor KLF2. Here, we identified KLF2- and flow-responsive miRs that affect gene expression in ECs. Bioinformatic assessment of mRNA expression patterns identified the miR-30-5p seed sequence to be highly enriched in mRNAs that are downregulated by KLF2. Indeed, KLF2 overexpression and shear stress stimulation in vitro and in vivo increased the expression of miR-30-5p family members. Furthermore, we identified angiopoietin 2 (Ang2) as a target of miR-30. MiR-30 overexpression reduces Ang2 levels, whereas miR-30 inhibition by LNA-antimiRs induces Ang2 expression. Consistently, miR-30 reduced basal and TNF-α-induced expression of the inflammatory cell–cell adhesion molecules E-selectin, ICAM1 and VCAM1, which was rescued by stimulation with exogenous Ang2. In summary, KLF2 and shear stress increase the expression of the miR-30-5p family which acts in an anti-inflammatory manner in ECs by impairing the expression of Ang2 and inflammatory cell–cell adhesion molecules. The upregulation of miR-30-5p family members may contribute to the atheroprotective effects of shear stress.
Collapse
Affiliation(s)
- Shemsi Demolli
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Carmen Doebele
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Anuradha Doddaballapur
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Victoria Lang
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen GmbH, Frankfurt, Germany
| | - Beate Fisslthaler
- Institute for Vascular Signaling, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Emmanouil Chavakis
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; Division of Internal Medicine III, Cardiology, Hospital of the Goethe University, Frankfurt, Germany
| | | | - Sergio Sciacca
- Cardiac Surgery and Heart Transplantation Unit, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Reinhard Henschler
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen GmbH, Frankfurt, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Soniya Savant
- Medical Faculty Mannheim (CBTM), Heidelberg University, Germany
| | - Hellmut G Augustin
- Medical Faculty Mannheim (CBTM), Heidelberg University, Germany; German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - David Kaluza
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany..
| |
Collapse
|
39
|
Erythropoietin-mediated expression of placenta growth factor is regulated via activation of hypoxia-inducible factor-1α and post-transcriptionally by miR-214 in sickle cell disease. Biochem J 2015; 468:409-23. [PMID: 25876995 DOI: 10.1042/bj20141138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 04/16/2015] [Indexed: 12/31/2022]
Abstract
Placental growth factor (PlGF) plays an important role in various pathological conditions and diseases such as inflammation, cancer, atherosclerosis and sickle cell disease (SCD). Abnormally high PlGF levels in SCD patients are associated with increased inflammation and pulmonary hypertension (PHT) and reactive airway disease; however, the transcriptional and post-transcriptional mechanisms regulating PlGF expression are not well defined. Herein, we show that treatment of human erythroid cells and colony forming units with erythropoietin (EPO) increased PlGF expression. Our studies showed EPO-mediated activation of HIF-1α led to subsequent binding of HIF-1α to hypoxia response elements (HREs) within the PlGF promoter, as demonstrated by luciferase transcription reporter assays and ChIP analysis of the endogenous gene. Additionally, we showed miR-214 post-transcriptionally regulated the expression of PlGF as demonstrated by luciferase reporter assays using wild-type (wt) and mutant PlGF-3'-UTR constructs. Furthermore, synthesis of miR-214, located in an intron of DNM3 (dynamin 3), was transcriptionally regulated by transcription factors, peroxisome proliferator-activated receptor-α (PPARα) and hypoxia-inducible factor-1α (HIF-1α). These results were corroborated in vivo wherein plasma from SCD patients and lung tissues from sickle mice showed an inverse correlation between PlGF and miR-214 levels. Finally, we observed that miR-214 expression could be induced by fenofibrate, a Food and Drug Administration (FDA) approved PPARα agonist, thus revealing a potential therapeutic approach for reduction in PlGF levels by increasing miR-214 transcription. This strategy has potential clinical implications for several pathological conditions including SCD.
Collapse
|
40
|
Abstract
Epidemiologic studies have revealed that modification of the levels of individual components of the hemostatic system may have effects on the development of thrombosis or hemorrhage. To maintain the necessary equilibrium, the hemostatic system is finely regulated. It is known that acquired factors and/or alterations in genes (single-nucleotide polymorphisms or mutations) may be the cause of interindividual differences or exacerbated levels of hemostatic proteins in plasma, but there are still many non-characterized factors that provoke such variations. The search for new elements, such as microRNAs (miRNAs), a family of small non-coding RNAs that are novel regulators of protein expression, may reveal an additional layer at which to investigate the causes of hemostatic diseases. In this review, we discuss the latest developments in research into the role of miRNAs in the regulation of several hemostatic factors, and the potential use of miRNAs as prognostic or diagnostic tools in hemostasis and thrombosis.
Collapse
Affiliation(s)
- R Teruel-Montoya
- Centro Regional de Hemodonación, IMIB-Arrixaca, University of Murcia, Murcia, Spain
| | | | | |
Collapse
|
41
|
Gao F, Chen S, Sun M, Mitchel REJ, Li B, Chu Z, Cai J, Liu C. MiR-467a is upregulated in radiation-induced mouse thymic lymphomas and regulates apoptosis by targeting Fas and Bax. Int J Biol Sci 2015; 11:109-21. [PMID: 25552935 PMCID: PMC4278260 DOI: 10.7150/ijbs.10276] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/18/2014] [Indexed: 01/23/2023] Open
Abstract
It has been reported dysregulation of certain microRNAs (miRNAs / miRs) is involved in tumorigenesis. However, the miRNAs associated with radiocarcinogenesis remain undefined. In this study, we validated the upregulation of miR-467a in radiation-induced mouse thymic lymphoma tissues. Then, we investigated whether miR-467a functions as an oncogenic miRNA in thymic lymphoma cells. For this purpose, we assessed the biological effect of miR-467a on thymic lymphoma cells. Using miRNA microarray, we found four miRNAs (miR-467a, miR-762, miR-455 and miR-714) were among the most upregulated (>4-fold) miRNAs in tumor tissues. Bioinformatics prediction suggests miR-467a may potentially regulate apoptosis pathway via targeting Fas and Bax. Consistently, in miR-467a-transfected cells, both proliferation and colony formation ability were significantly increased with decrease of apoptosis rate, while, in miR-467a-knockdown cells, proliferation was suppressed with increase of apoptosis rate, indicating that miR-467a may be involved in the regulation of apoptosis. Furthermore, miR-467a-knockdown resulted in smaller tumors and better prognosis in an in vivo tumor-transplanted model. To explain the mechanism of apoptosis suppression by miR-467a, we explore the expression of candidate target genes (Fas and Bax) in miR-467a-transfected relative to negative control transfected cells using flow cytometry and immunoblotting. Fas and Bax were commonly downregulated in miR-467a-transfected EL4 and NIH3T3 cells, and all of the genes harbored miR-467a target sequences in the 3'UTR of their mRNA. Fas and Bax were actually downregulated in radiation-induced thymic lymphoma tissues, and therefore both were identified as possible targets of miR-467a in thymic lymphoma. To ascertain whether downregulation of Fas and / or Bax is involved in apoptosis suppression by miR-467a, we transfected vectors expressing Fas and Bax into miR-467a-upregulated EL4 cells. Then we found that both Fas- and Bax-overexpression decreased cell viability with increase of apoptosis rate, indicating that downregulation of Fas and Bax may be at least partly responsible for apoptosis suppression by miR-467a. These data suggest that miR-467a may have oncogenic functions in radiation-induced thymic lymphoma cells and that its increased expression may confer a growth advantage on tumor cells via aberrant expression of Fas and Bax.
Collapse
Affiliation(s)
- Fu Gao
- 1. Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Song Chen
- 2. Department of Radiological Protection, Naval Medical Research Institute, Shanghai 200433, PR China
| | - Mingjuan Sun
- 3. Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, PR China
| | - Ronald E J Mitchel
- 4. Radiological Protection Research and Instrumentation Branch, Atomic Energy of Canada Limited, Chalk River Laboratories, Chalk River, ON, K0J1J0, Canada
| | - Bailong Li
- 1. Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Zhiyong Chu
- 2. Department of Radiological Protection, Naval Medical Research Institute, Shanghai 200433, PR China
| | - Jianming Cai
- 1. Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Cong Liu
- 1. Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
42
|
MicroRNA 648 Targets ET-1 mRNA and is cotranscriptionally regulated with MICAL3 by PAX5. Mol Cell Biol 2014; 35:514-28. [PMID: 25403488 DOI: 10.1128/mcb.01199-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pulmonary hypertension (PHT) is associated with high mortality in sickle cell anemia (SCA). Previously, we showed that elevated levels of placenta growth factor (PlGF) in SCA patients correlate with increased levels of the potent vasoconstrictor endothelin-1 (ET-1) and PHT. Moreover, PlGF induced the expression of ET-1 via hypoxia-inducible factor 1α. Here, we show a novel example of ET-1 posttranscriptional regulation by PlGF via action of microRNA 648 (miR-648), which is subject to transcriptional coregulation with its host gene, MICAL3 (microtubule-associated monooxygenase, calponin, and LIM domain containing 3gene). PlGF repressed expression of miR-648 in endothelial cells. Luciferase reporter assays using wild-type and mutant ET-1 3' untranslated region (UTR) constructs, and transfection of miR-648 mimics showed that miR-648 targets the 3' UTR of ET-1 mRNA. Since miR-648 is located in a 5'-proximal intron of MICAL3, we examined which of three potential promoters was responsible for its expression. The MICAL3 distal promoter (P1) was the predominant promoter used for transcription of pre-miR-648, and it was under positive control by PAX5 (paired box protein 5) transcription factor, as demonstrated by the loss and gain of function of PAX5 activity, and chromatin immunoprecipitation analysis. These studies provide a novel link wherein PlGF-mediated downregulation of PAX5 attenuates miR-648 expression leading to increased ET-1 levels that are known to induce PHT in SCA.
Collapse
|
43
|
Li C, Mpollo MSEM, Gonsalves CS, Tahara SM, Malik P, Kalra VK. Peroxisome proliferator-activated receptor-α-mediated transcription of miR-199a2 attenuates endothelin-1 expression via hypoxia-inducible factor-1α. J Biol Chem 2014; 289:36031-47. [PMID: 25389292 DOI: 10.1074/jbc.m114.600775] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Endothelin-1, a potent vasoconstrictor, plays an important role in pulmonary hypertension (PH) in sickle cell disease (SCD). Our previous studies show that higher levels of placenta growth factor (PlGF), secreted by erythroid precursor cells, correlate with increased plasma levels of endothelin-1 (ET-1) and other functional markers of PH in SCD. PlGF-mediated ET-1 expression occurs via activation of hypoxia-inducible factor-1α (HIF-1α). However, relatively less is understood regarding how PlGF-mediated expression of HIF-1α and its downstream effector ET-1 are post-transcriptionally regulated. Herein, we show that PlGF treatment of endothelial cells resulted in reduced levels of miR-199a2, which targeted the 3'-UTR of HIF-1α mRNA and concomitantly led to augmented ET-1 expression. Plasma levels of miR-199a2 in SCD subjects were significantly lower with reciprocally high levels of plasma ET-1, unlike unaffected controls. This observation provided a molecular link between miR-199a2 and high levels of ET-1 in SCD. Furthermore, we show that miR-199a2 located in the DNM3os transcription unit was co-transcriptionally regulated by peroxisome proliferator-activated receptor α (PPARα). Binding of the latter to PPARα cis-elements in the promoter of DNM3os was demonstrated by promoter mutational analysis and ChIP. Additionally, we show that fenofibrate, a PPARα agonist, increased the expression of miR-199a2 and DNM3os; the former was responsible for reduced expression of HIF-1α and ET-1. In vivo studies of fenofibrate-fed Berkeley sickle mice resulted in increased levels of miR-199a2 and reduced levels of ET-1 in lung tissues. Our studies provide a potential therapeutic approach whereby fenofibrate-induced miR-199a2 expression can ameliorate PH by reduction of ET-1 levels.
Collapse
Affiliation(s)
- Chen Li
- From the Departments of Biochemistry and Molecular Biology and
| | - Marthe-Sandrine Eiymo Mwa Mpollo
- the Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | | | - Stanley M Tahara
- Molecular Microbiology and Immunology, Keck School of Medicine of University of Southern California, Los Angeles, California 90089 and
| | - Punam Malik
- the Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Vijay K Kalra
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
44
|
Li S, Chen H, Ren J, Geng Q, Song J, Lee C, Cao C, Zhang J, Xu N. MicroRNA-223 inhibits tissue factor expression in vascular endothelial cells. Atherosclerosis 2014; 237:514-20. [PMID: 25463083 DOI: 10.1016/j.atherosclerosis.2014.09.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/09/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory process, in which vascular endothelial cells (ECs) become dysfunctional owing to the effects of chemical substances, such as inflammatory factor and growth factors. Tissue factor (TF) expression is induced by the above chemical substances in activated ECs. TF initiates thrombosis on disrupted atherosclerotic plaques which plays an essential role during the onset of acute coronary syndromes (ACS). Increasing evidences suggest the important role of microRNAs as epigenetic regulators of atherosclerotic disease. The aim of our study is to identify if microRNA-223 (miR-223) targets TF in ECs. METHODS AND RESULTS Bioinformatic analysis showed that TF is a target candidate of miR-223. Western blotting analysis revealed that tumor necrosis factor α (TNF-α) increased TF expression in aorta of C57BL/6J mice and cultured ECs (EA.hy926 cells and HUVEC) after 4 h treatment. In TNF-α treated ECs, TF mRNA was also increased measured by real-time PCR. Real-time PCR results showed that miR-223 levels were downregulated in TNF-α-treated aorta of C57BL/6J mice and cultured ECs. Transfection of ECs with miR-223 mimic or miR-223 inhibitor modified TF expression both in mRNA and protein levels. Luciferase assays confirmed that miR-223 suppressed TF expression by binding to the sequence of TF 3'-untranslated regions (3'UTR). TF procoagulant activity was inhibited by overexpressing miR-223 with or without TNF-α stimulation. CONCLUSIONS MiR-223-mediated suppression of TF expression provides a novel molecular mechanism for the regulation of coagulation cascade, and suggests a clue against thrombogenesis during the process of atherosclerotic plaque rupture.
Collapse
Affiliation(s)
- Sufang Li
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Jingyi Ren
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Qiang Geng
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Junxian Song
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Chongyou Lee
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Chengfu Cao
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jing Zhang
- Department of Cardiology, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Ning Xu
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
The malignancy suppression role of miR-23a by targeting the BCR/ABL oncogene in chromic myeloid leukemia. Cancer Gene Ther 2014; 21:397-404. [PMID: 25213664 DOI: 10.1038/cgt.2014.44] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 07/25/2014] [Indexed: 12/22/2022]
Abstract
The aim of this study was to investigate the role and mechanism of miR-23a in the regulation of BCR/ABL and to provide a new prognostic biomarker for chronic myeloid leukemia (CML). The expression levels of miR-23a and BCR/ABL were assessed in 42 newly diagnosed CML patients, 37 CML patients in first complete remission and 25 healthy controls. Quantitative real-time PCR, western blot analysis and colony formation assay were used to evaluate changes induced by overexpression or inhibition of miR-23a or BCR/ABL. MiR-23a mimic or negative control mimic was transfected into a CML cell line (K562) and two lung cancer cell lines (H157 and SKMES1) using Lipofectamine 2000, and the cells were used for real-time reverse transcription-PCR (RT-PCR) and western blot analysis. We found that the downregulation of miR-23a expression was a frequent event in both leukemia cell lines and primary leukemic cells from patients with de novo CML. The microarray results showed that most of the CML patients expressed high levels of BCR/ABL and low levels of miR-23a. Real-time RT-PCR and western blot analysis showed that the BCR/ABL levels in miR-23a-transfected cells were lower than those in the control groups. Ectopic expression of miR-23a in K562 cells led to cellular senescence. Moreover, when K562 cells were treated with 5-aza-2'-deoxycytidine, a DNA methylation inhibitor, BCR/ABL expression was upregulated, which indicates epigenetic silencing of miR-23a in leukemic cells. BCR/ABL and miR-23a expressions were inversely related to CML, and BCR/ABL expression was regulated by miR-23a in leukemic cells. The epigenetic silencing of miR-23a led to derepression of BCR/ABL expression, and consequently contributes to CML development and progression.
Collapse
|
46
|
MIMURA SHIMA, IWAMA HISAKAZU, KATO KIYOHITO, NOMURA KEI, KOBAYASHI MITSUYOSHI, YONEYAMA HIROHITO, MIYOSHI HISAAKI, TANI JOJI, MORISHITA ASAHIRO, HIMOTO TAKASHI, DEGUCHI AKIHIRO, NOMURA TAKAKO, SAKAMOTO TEPPEI, FUJITA KOJI, MAEDA EMIKO, IZUISHI KUNIHIKO, OKANO KEIICHI, SUZUKI YASUYUKI, MASAKI TSUTOMU. Profile of microRNAs associated with aging in rat liver. Int J Mol Med 2014; 34:1065-72. [DOI: 10.3892/ijmm.2014.1892] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 07/30/2014] [Indexed: 11/05/2022] Open
|
47
|
Li S, Ren J, Xu N, Zhang J, Geng Q, Cao C, Lee C, Song J, Li J, Chen H. MicroRNA-19b functions as potential anti-thrombotic protector in patients with unstable angina by targeting tissue factor. J Mol Cell Cardiol 2014; 75:49-57. [PMID: 24998411 DOI: 10.1016/j.yjmcc.2014.06.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/12/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
The activation of a hemostatic system plays a critical role in the incidence of acute coronary events. Hemostatic proteins may be regulated by microRNAs (miRNAs). Microparticles (MPs) are the major carrier of circulating miRNAs. The aim of this study was to determine the potential role of miRNAs in regulating gene expression involved in the hemostatic system in patients with unstable angina (UA). MiRNA expression profiles in the plasma from patients with UA (UA group, n=9) compared with individuals with clinical suspicion of coronary artery disease (CAD) but negative angiography (control group, n=9) showed that among 36 differentially expressed miRNAs, miR-19b was the most obvious one. Using real-time PCR, 5 selected miRNA levels in plasma (UA group, n=20; control group, n=30) and plasma MPs (UA group n=6; control group n=6) were proved to be consistent with the miRNA array. Flow cytometry analysis indicated that the amounts of plasma endothelial microparticles (EMPs) were increased in UA patients (UA group, n=4) compared to controls (control group, n=4). In cultured endothelial cells (ECs), TNF-α increased miR-19b release and expression. Tissue factor (TF) was predicted to be the target of miR-19b by bioinformatics analysis. Luciferase reporter assays demonstrated that miR-19b binds to TF mRNA. Overexpression of miR-19b inhibited TF expression and procoagulant activity. This study indicates that in UA patients, the increase of miR-19b wrapped in EMPs due to endothelial dysfunction may partially contribute to the circulating miR-19b elevation and miR-19b may play an anti-thrombotic role by inhibiting the expression of TF in ECs.
Collapse
Affiliation(s)
- Sufang Li
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Jingyi Ren
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Ning Xu
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jing Zhang
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Qiang Geng
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Chengfu Cao
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Chongyou Lee
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Junxian Song
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Jingjin Li
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
48
|
Heme-bound iron activates placenta growth factor in erythroid cells via erythroid Krüppel-like factor. Blood 2014; 124:946-54. [PMID: 24916507 DOI: 10.1182/blood-2013-11-539718] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In adults with sickle cell disease (SCD), markers of iron burden are associated with excessive production of the angiogenic protein placenta growth factor (PlGF) and high estimated pulmonary artery pressure. Enforced PlGF expression in mice stimulates production of the potent vasoconstrictor endothelin-1, producing pulmonary hypertension. We now demonstrate heme-bound iron (hemin) induces PlGF mRNA >200-fold in a dose- and time-dependent fashion. In murine and human erythroid cells, expression of erythroid Krüppel-like factor (EKLF) precedes PlGF, and its enforced expression in human erythroid progenitor cells induces PlGF mRNA. Hemin-induced expression of PlGF is abolished in EKLF-deficient murine erythroid cells but rescued by conditional expression of EKLF. Chromatin immunoprecipitation reveals that EKLF binds to the PlGF promoter region. SCD patients show higher level expression of both EKLF and PlGF mRNA in circulating blood cells, and markers of iron overload are associated with high PlGF and early mortality. Finally, PlGF association with iron burden generalizes to other human diseases of iron overload. Our results demonstrate a specific mechanistic pathway induced by excess iron that is linked in humans with SCD and in mice to markers of vasculopathy and pulmonary hypertension. These trials were registered at www.clinicaltrials.gov as #NCT00007150, #NCT00023296, #NCT00081523, and #NCT00352430.
Collapse
|
49
|
Wijnen WJ, van der Made I, van den Oever S, Hiller M, de Boer BA, Picavet DI, Chatzispyrou IA, Houtkooper RH, Tijsen AJ, Hagoort J, van Veen H, Everts V, Ruijter JM, Pinto YM, Creemers EE. Cardiomyocyte-specific miRNA-30c over-expression causes dilated cardiomyopathy. PLoS One 2014; 9:e96290. [PMID: 24789369 PMCID: PMC4008570 DOI: 10.1371/journal.pone.0096290] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/07/2014] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) regulate many aspects of cellular function and their deregulation has been implicated in heart disease. MiRNA-30c is differentially expressed in the heart during the progression towards heart failure and in vitro studies hint to its importance in cellular physiology. As little is known about the in vivo function of miRNA-30c in the heart, we generated transgenic mice that specifically overexpress miRNA-30c in cardiomyocytes. We show that these mice display no abnormalities until about 6 weeks of age, but subsequently develop a severely dilated cardiomyopathy. Gene expression analysis of the miRNA-30c transgenic hearts before onset of the phenotype indicated disturbed mitochondrial function. This was further evident by the downregulation of mitochondrial oxidative phosphorylation (OXPHOS) complexes III and IV at the protein level. Taken together these data indicate impaired mitochondrial function due to OXPHOS protein depletion as a potential cause for the observed dilated cardiomyopathic phenotype in miRNA-30c transgenic mice. We thus establish an in vivo role for miRNA-30c in cardiac physiology, particularly in mitochondrial function.
Collapse
Affiliation(s)
- Wino J. Wijnen
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
- Interuniversitair Cardiologisch Instituut Nederland (ICIN-NHI), Utrecht, The Netherlands
| | | | | | - Monika Hiller
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Bouke A. de Boer
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Daisy I. Picavet
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Iliana A. Chatzispyrou
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Anke J. Tijsen
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Jaco Hagoort
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Henk van Veen
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Vincent Everts
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Jan M. Ruijter
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Yigal M. Pinto
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther E. Creemers
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
50
|
Kong X, Xu X, Yan Y, Guo F, Li J, Hu Y, Zhou H, Xun Q. Estrogen regulates the tumour suppressor MiRNA-30c and its target gene, MTA-1, in endometrial cancer. PLoS One 2014; 9:e90810. [PMID: 24595016 PMCID: PMC3940948 DOI: 10.1371/journal.pone.0090810] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 02/04/2014] [Indexed: 11/18/2022] Open
Abstract
MicroRNA-30c (miR-30c) has been reported to be a tumour suppressor in endometrial cancer (EC). We demonstrate that miR-30c is down-regulated in EC tissue and is highly expressed in estrogen receptor (ER)-negative HEC-1-B cells. MiR-30c directly inhibits MTA-1 expression and functions as a tumour suppressor via the miR-30c-MTA-1 signalling pathway. Furthermore, miR-30c is decreased upon E2 treatment in both ER-positive Ishikawa and ER-negative HEC-1-B cells. Taken together, our results suggest that miR-30c is an important deregulated miRNA in EC and might serve as a potential biomarker and novel therapeutic target for EC.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - XiaoFeng Xu
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yuhua Yan
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Feifei Guo
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Jian Li
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yali Hu
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Huaijun Zhou
- Department of Gynecology and Obstetrics, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Qingying Xun
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| |
Collapse
|