1
|
González-Moyotl N, Huesca-Gómez C, Torres-Paz YE, Fuentevilla-Álvarez G, Romero-Maldonado S, Sámano R, Soto ME, Martínez-Rosas M, Domínguez-López A, Gamboa R. Paediatrics congenital heart disease is associated with plasma miRNAs. Pediatr Res 2024; 96:1220-1227. [PMID: 38755412 DOI: 10.1038/s41390-024-03230-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Congenital heart disease (CHD) are the most common malformations from birth. The severity of the different forms of CHD varies extensively from superficial mild lesions with follow-up for decades without any treatment to complex cyanotic malformations requiring urgent surgical intervention. microRNAs have been found to be crucial in cardiac development, giving rise to possible phenotypes in CHD. OBJECTIVES We aimed to evaluate the expression of miRNAs in 86 children with CHD and divided into cyanotic and non-cyanotic heart defects and 110 controls. METHODS The miRNAs expression of miR-21-5p, miR-155-5p, miR-221-3p, miR-26a-5p, and miR-144-3p were analyzed by RT-qPCR. In addition, the expressions of the miRNAs studied were correlated with the clinical characteristics of both the children and the mothers. RESULTS The expression levels of miR-21-5-5p, miR-15-5p5, miR-221-3p, and miR-26-5p significantly differed between CHD and control subjects. Moreover, miR-21-5p levels were higher in patients with cyanotic versus non-cyanotic CHD patients. CONCLUSION The expression levels of miRNAs of pediatric patients with CHD could participating in the development of cardiac malformations. Additionally, the high expression of miR-21-5p in cyanotic CHD children may be related to greater severity of illness relative to non-cyanotic CHD. IMPACT This study adds to knowledge of the association between microRNAs and congenital heart disease in children. The expression levels of miR-21-5-5p, miR-15-5p5, miR-221-3p, and miR-26-5p of pediatric patients with CHD could be involved in the development and phenotype present in pediatric patients. miR-21-5p may help to discriminate between cyanotic and non-cyanotic CHD. In the future, the miRNAs studied could have applications as clinical biomarkers.
Collapse
Affiliation(s)
- Nadia González-Moyotl
- Instituto Nacional de Cardiología Ignacio Chávez. Department of Physiology, México City, 14380, México
- Maestría en Ciencias de la Salud, Posgrado de la Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11350, México
| | - Claudia Huesca-Gómez
- Instituto Nacional de Cardiología Ignacio Chávez. Department of Physiology, México City, 14380, México
| | - Yazmín Estela Torres-Paz
- Instituto Nacional de Cardiología Ignacio Chávez. Department of Physiology, México City, 14380, México
| | | | - Silvia Romero-Maldonado
- Instituto Nacional de Perinatología, Coordination of the Human Milk Bank, México City, 11000, México
| | - Reyna Sámano
- Instituto Nacional de Perinatología. Coordination of Nutrition and Bioprogramming, México City, 11000, México
| | - María Elena Soto
- Instituto Nacional de Cardiología Ignacio Chávez. Research Direction, México City, 14380, México
| | - Martín Martínez-Rosas
- Instituto Nacional de Cardiología Ignacio Chávez. Department of Physiology, México City, 14380, México
| | - Aarón Domínguez-López
- Maestría en Ciencias de la Salud, Posgrado de la Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, 11350, México
| | - Ricardo Gamboa
- Instituto Nacional de Cardiología Ignacio Chávez. Department of Physiology, México City, 14380, México.
| |
Collapse
|
2
|
Emery L, Hughes A, Oji-Mmuo C, Silveyra P, Aluquin VPR, Donnelly A, Siddaiah R. Omic-signature of bronchopulmonary dysplasia associated pulmonary hypertension in <1500g-birth-weight-infants with hemodynamically significant intracardiac shunt. Pediatr Res 2024:10.1038/s41390-024-03541-5. [PMID: 39271902 DOI: 10.1038/s41390-024-03541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND PDA and ASD are common intracardiac shunts noted in prematurely born infants. While there is evidence of persistent PDA and ASD associated with a higher risk for developing bronchopulmonary dysplasia (ICS-BPD) and pulmonary hypertension (ICS-BPD-PH), the underlying pathogenesis is poorly understood and hence challenging to identify at-risk infants. Our study goal was to evaluate transcriptomic expression and associated pathways in tracheal aspirates (TAs) of low-birth-weight infants with hemodynamically significant cardiac shunt (ICS) that develop bronchopulmonary dysplasia (ICS-BPD) and pulmonary hypertension (ICS-BPD-PH). METHODS TAs were collected from preterm infants with ICS and a diagnosis of BPD or BPD-PH from a single center. 36 TA samples including 19 ICS-BPD and 17 ICS-BPD-PH were analyzed. MiRNA expression was determined via PCR arrays, and mRNA expression via RNA seq. Data were analyzed using limma. RESULTS 11 miRNAs and 10 mRNAs were differentially expressed (adjusted p < 0.05) in ICS-BPD-PH group when compared to ICS-BPD. Ingenuity Pathway Analysis identified associations with cellular growth, proliferation, death, and cell function pathways. CONCLUSION TAs from preterm infants show differentially expressed miRNAs and mRNAs in ICS-BPD-PH when compared to ICS-BPD, an in-silico model identified target molecules that could be playing a role in BPD-PH pathogenesis in low-birth-weight infants with ICS. IMPACT Pulmonary hypertension associated with severe BPD (BPD-PH) is a distinct disease in preterm infants with severe BPD and the role of intracardiac shunt (ICS) in its development is controversial and often challenging for clinical management. Our pilot study, researching specific endotyping of infants with pulmonary hypertension associated with BPD using multiomics approach has identified molecular markers and potential underlying pathways associated with this condition. These markers could aid in stratifying high risk infants with ICS that are at risk for developing BPD-PH and aid clinical management.
Collapse
Affiliation(s)
- Lucy Emery
- Penn State Health College of Medicine, Hershey, PA, USA
| | - Alexa Hughes
- Penn State Health College of Medicine, Hershey, PA, USA
| | - Christiana Oji-Mmuo
- Department of Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vincent P R Aluquin
- Department of Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Ann Donnelly
- Department of Respiratory Therapy, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Roopa Siddaiah
- Department of Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA.
| |
Collapse
|
3
|
Li KX, Li JR, Zuo SJ, Li X, Chen XT, Xiao PY, Li HT, Sun L, Qian T, Zhang HM, Zhu D, Yu XY, Chen G, Jiang XY. Identification of miR-20b-5p as an inhibitory regulator in cardiac differentiation via TET2 and DNA hydroxymethylation. Clin Epigenetics 2024; 16:42. [PMID: 38491513 PMCID: PMC10943922 DOI: 10.1186/s13148-024-01653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is a prevalent congenital cardiac malformation, which lacks effective early biological diagnosis and intervention. MicroRNAs, as epigenetic regulators of cardiac development, provide potential biomarkers for the diagnosis and treatment of CHD. However, the mechanisms underlying miRNAs-mediated regulation of cardiac development and CHD malformation remain to be further elucidated. This study aimed to explore the function of microRNA-20b-5p (miR-20b-5p) in cardiac development and CHD pathogenesis. METHODS AND RESULTS miRNA expression profiling identified that miR-20b-5p was significantly downregulated during a 12-day cardiac differentiation of human embryonic stem cells (hESCs), whereas it was markedly upregulated in plasma samples of atrial septal defect (ASD) patients. Our results further revealed that miR-20b-5p suppressed hESCs-derived cardiac differentiation by targeting tet methylcytosine dioxygenase 2 (TET2) and 5-hydroxymethylcytosine, leading to a reduction in key cardiac transcription factors including GATA4, NKX2.5, TBX5, MYH6 and cTnT. Additionally, knockdown of TET2 significantly inhibited cardiac differentiation, which could be partially restored by miR-20b-5p inhibition. CONCLUSIONS Collectively, this study provides compelling evidence that miR-20b-5p functions as an inhibitory regulator in hESCs-derived cardiac differentiation by targeting TET2, highlighting its potential as a biomarker for ASD.
Collapse
Affiliation(s)
- Ke-Xin Li
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jia-Ru Li
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Sheng-Jia Zuo
- Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Xudong Li
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xian-Tong Chen
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Pei-Yi Xiao
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hui-Tao Li
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, 518028, China
| | - Ling Sun
- Department of Cardiac Pediatrics, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tao Qian
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hao-Min Zhang
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Dongxing Zhu
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xi-Yong Yu
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xue-Yan Jiang
- Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
4
|
Zhang M, Han Y. MicroRNAs in chronic pediatric diseases (Review). Exp Ther Med 2024; 27:100. [PMID: 38356668 PMCID: PMC10865459 DOI: 10.3892/etm.2024.12388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/15/2023] [Indexed: 02/16/2024] Open
Abstract
MicroRNAs are small non-coding RNAs with a length of 20-24 nucleotides. They bind to the 3'-untranslated region of target genes to induce the degradation of target mRNAs or inhibit their translation. Therefore, they are involved in the regulation of development, apoptosis, proliferation, differentiation and other biological processes (including hormone secretion, signaling and viral infections). Chronic diseases in children may be difficult to treat and are often associated with malnutrition resulting from a poor diet. Consequently, further complications, disease aggravation and increased treatment costs impose a burden on patients and their families. Existing evidence suggests that microRNAs are involved in various chronic non-neoplastic diseases in children. The present review discusses the roles of microRNAs in five major chronic diseases in children, namely, diabetes mellitus, congenital heart diseases, liver diseases, bronchial asthma and epilepsy, providing a theoretical basis for them to become therapeutic biomarkers in chronic pediatric diseases.
Collapse
Affiliation(s)
- Mingyao Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanhua Han
- Department of Pediatrics, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
5
|
Zhu W, Lo CW. Insights into the genetic architecture of congenital heart disease from animal modeling. Zool Res 2023; 44:577-590. [PMID: 37147909 PMCID: PMC10236297 DOI: 10.24272/j.issn.2095-8137.2022.463] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/28/2023] [Indexed: 05/07/2023] Open
Abstract
Congenital heart disease (CHD) is observed in up to 1% of live births and is one of the leading causes of mortality from birth defects. While hundreds of genes have been implicated in the genetic etiology of CHD, their role in CHD pathogenesis is still poorly understood. This is largely a reflection of the sporadic nature of CHD, as well as its variable expressivity and incomplete penetrance. We reviewed the monogenic causes and evidence for oligogenic etiology of CHD, as well as the role of de novo mutations, common variants, and genetic modifiers. For further mechanistic insight, we leveraged single-cell data across species to investigate the cellular expression characteristics of genes implicated in CHD in developing human and mouse embryonic hearts. Understanding the genetic etiology of CHD may enable the application of precision medicine and prenatal diagnosis, thereby facilitating early intervention to improve outcomes for patients with CHD.
Collapse
Affiliation(s)
- Wenjuan Zhu
- Chinese University of Hong Kong, Hong Kong SAR, China
- Kunming Institute of Zoology-Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Hong Kong SAR, China
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201 USA. E-mail:
| |
Collapse
|
6
|
Yang Y, Yang H, Lian X, Yang S, Shen H, Wu S, Wang X, Lyu G. Circulating microRNA: Myocardium-derived prenatal biomarker of ventricular septal defects. Front Genet 2022; 13:899034. [PMID: 36035156 PMCID: PMC9403759 DOI: 10.3389/fgene.2022.899034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Recently, circulating microRNAs (miRNAs) from maternal blood and amniotic fluid have been used as biomarkers for ventricular septal defect (VSD) diagnosis. However, whether circulating miRNAs are associated with fetal myocardium remains unknown.Methods: Dimethadione (DMO) induced a VSD rat model. The miRNA expression profiles of the myocardium, amniotic fluid and maternal serum were analyzed. Differentially expressed microRNAs (DE-microRNAs) were verified by qRT–PCR. The target gene of miR-1-3p was confirmed by dual luciferase reporter assays. Expression of amniotic fluid-derived DE-microRNAs was verified in clinical samples.Results: MiRNAs were differentially expressed in VSD fetal rats and might be involved in cardiomyocyte differentiation and apoptosis. MiR-1-3p, miR-1b and miR-293-5p were downregulated in the myocardium and upregulated in amniotic fluid/maternal serum. The expression of amniotic fluid-derived DE-microRNAs (miR-1-3p, miR-206 and miR-184) was verified in clinical samples. Dual luciferase reporter assays confirmed that miR-1-3p directly targeted SLC8A1/NCX1.Conclusion: MiR-1-3p, miR-1b and miR-293-5p are downregulated in VSD myocardium and upregulated in circulation and may be released into circulation by cardiomyocytes. MiR-1-3p targets SLC8A1/NCX1 and participates in myocardial apoptosis. MiR-1-3p upregulation in circulation is a direct and powerful indicator of fetal VSD and is expected to serve as a prenatal VSD diagnostic marker.
Collapse
Affiliation(s)
- Yiru Yang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hainan Yang
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xihua Lian
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Shuping Yang
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Haolin Shen
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shufen Wu
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Xiali Wang
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology, Quanzhou Medical College, Quanzhou, Fujian, China
- *Correspondence: Guorong Lyu,
| |
Collapse
|
7
|
Sarkar S, Sen R. Insights into Cardiovascular Defects and Cardiac Epigenome in the Context of COVID-19. EPIGENOMES 2022; 6:epigenomes6020013. [PMID: 35645252 PMCID: PMC9150012 DOI: 10.3390/epigenomes6020013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Although few in number, studies on epigenome of the heart of COVID-19 patients show that epigenetic signatures such as DNA methylation are significantly altered, leading to changes in expression of several genes. It contributes to pathogenic cardiac phenotypes of COVID-19, e.g., low heart rate, myocardial edema, and myofibrillar disarray. DNA methylation studies reveal changes which likely contribute to cardiac disease through unknown mechanisms. The incidence of severe COVID-19 disease, including hospitalization, requiring respiratory support, morbidity, and mortality, is disproportionately higher in individuals with co-morbidities. This poses unprecedented strains on the global healthcare system. While their underlying conditions make patients more susceptible to severe COVID-19 disease, strained healthcare systems, lack of adequate support, or sedentary lifestyles from ongoing lockdowns have proved detrimental to their underlying health conditions, thus pushing them to severe risk of congenital heart disease (CHD) itself. Prophylactic vaccines against COVID-19 have ushered new hope for CHD. A common connection between COVID-19 and CHD is SARS-CoV-2’s host receptor ACE2, because ACE2 regulates and protects organs, including the heart, in various ways. ACE2 is a common therapeutic target against cardiovascular disease and COVID-19 which damages organs. Hence, this review explores the above regarding CHDs, cardiovascular damage, and cardiac epigenetics, in COVID-19 patients.
Collapse
Affiliation(s)
- Shreya Sarkar
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB E2L 4L2, Canada;
| | - Rwik Sen
- Active Motif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
- Correspondence:
| |
Collapse
|
8
|
Seelan RS, Pisano MM, Greene RM. MicroRNAs as Biomarkers for Birth Defects. Microrna 2022; 11:2-11. [PMID: 35168515 DOI: 10.2174/2211536611666220215123423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 11/22/2022]
Abstract
It is estimated that 2-4% of live births will have a birth defect (BD). The availability of biomarkers for the prenatal detection of BDs will facilitate early risk assessment, prompt medical intervention and ameliorating disease severity. miRNA expression levels are often found to be altered in many diseases. There is, thus, a growing interest in determining whether miRNAs, particularly extracellular miRNAs, can predict, diagnose, or monitor BDs. These miRNAs, typically encapsulated in exosomes, are released by cells (including those of the fetus and placenta) into the extracellular milieu, such as blood, urine, saliva and cerebrospinal fluid, thereby enabling interaction with target cells. Exosomal miRNAs are stable, protected from degradation, and retain functionality. The observation that placental and fetal miRNAs can be detected in maternal serum, provides a strong rationale for adopting miRNAs as noninvasive prenatal biomarkers for BDs. In this mini-review, we examine the current state of research involving the use of miRNAs as prognostic and diagnostic biomarkers for BD.
Collapse
Affiliation(s)
- Ratnam S Seelan
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, University of Louisville, Louisville, KY 40202, USA
| | - M Michele Pisano
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, University of Louisville, Louisville, KY 40202, USA
| | - Robert M Greene
- Department of Oral Immunology and Infectious Diseases, Division of Craniofacial Development and Anomalies, School of Dentistry, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
9
|
Zhang GQ, Wang SQ, Chen Y, Fu LY, Xu YN, Li L, Tao L, Shen XC. MicroRNAs Regulating Mitochondrial Function in Cardiac Diseases. Front Pharmacol 2021; 12:663322. [PMID: 34122082 PMCID: PMC8194257 DOI: 10.3389/fphar.2021.663322] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are the key organelles that supply cellular energy. As the most active organ in the body, the energy required to maintain the mechanical function of the heart requires a high quantity of high-quality mitochondria in cardiomyocytes. MicroRNAs (miRNAs) are single-stranded noncoding RNAs, approximately 22 nt in length, which play key roles in mediating post-transcriptional gene silencing. Numerous studies have confirmed that miRNAs can participate in the occurrence and development of cardiac diseases by regulating mitochondrial function-related genes and signaling pathways. Therefore, elucidating the crosstalk that occurs between miRNAs and mitochondria is important for the prevention and treatment of cardiac diseases. In this review, we discuss the biogenesis of miRNAs, the miRNA-mediated regulation of major genes involved in the maintenance of mitochondrial function, and the effects of miRNAs on mitochondrial function in cardiac diseases in order to provide a theoretical basis for the clinical prevention and treatment of cardiac disease and the development of new drugs.
Collapse
Affiliation(s)
- Guang-Qiong Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Sheng-Quan Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Ling-Yun Fu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Yi-Ni Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Ling Li
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Xiang-Chun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| |
Collapse
|
10
|
Zhang X, Gao Y, Zhang X, Zhang X, Xiang Y, Fu Q, Wang B, Xu Z. FGD5-AS1 Is a Hub lncRNA ceRNA in Hearts With Tetralogy of Fallot Which Regulates Congenital Heart Disease Genes Transcriptionally and Epigenetically. Front Cell Dev Biol 2021; 9:630634. [PMID: 34046402 PMCID: PMC8144506 DOI: 10.3389/fcell.2021.630634] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/30/2021] [Indexed: 01/19/2023] Open
Abstract
Heart development requires robust gene regulation, and the related disruption could lead to congenital heart disease (CHD). To gain insights into the regulation of gene expression in CHD, we obtained the expression profiles of long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) in 22 heart tissue samples with tetralogy of Fallot (TOF) through strand-specific transcriptomic analysis. Using a causal inference framework based on the expression correlations and validated microRNA (miRNA)–lncRNA–mRNA evidences, we constructed the competing endogenous RNA (ceRNA)-mediated network driven by lncRNAs. Four lncRNAs (FGD5-AS1, lnc-GNB4-1, lnc-PDK3-1, and lnc-SAMD5-1) were identified as hub lncRNAs in the network. FGD5-AS1 was selected for further study since all its targets were CHD-related genes (NRAS, PTEN, and SMAD4). Both FGD5-AS1 and SMAD4 could bind with hsa-miR-421, which has been validated using dual-luciferase reporter assays. Knockdown of FGD5-AS1 not only significantly reduced PTEN and SMAD4 expression in HEK 293 and the fetal heart cell line (CCC-HEH-2) but also increased the transcription of its interacted miRNAs in a cell-specific way. Besides ceRNA mechanism, RNAseq and ATACseq results showed that FGD5-AS1 might play repression roles in heart development by transcriptionally regulating CHD-related genes. In conclusion, we identified a ceRNA network driven by lncRNAs in heart tissues of TOF patients. Furthermore, we proved that FGD5-AS1, one hub lncRNA in the TOF heart ceRNA network, regulates multiple genes transcriptionally and epigenetically.
Collapse
Affiliation(s)
- Xingyu Zhang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunqian Gao
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Zhang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqing Zhang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Xiang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qihua Fu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Faculty of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Wang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Faculty of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuoming Xu
- Cardiac Intensive Care Unit, Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Feng D, Christensen JT, Yetman AT, Lindsey ML, Singh AB, Salomon JD. The microbiome’s relationship with congenital heart disease: more than a gut feeling. JOURNAL OF CONGENITAL CARDIOLOGY 2021. [DOI: 10.1186/s40949-021-00060-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AbstractPatients with congenital heart disease (CHD) are at risk for developing intestinal dysbiosis and intestinal epithelial barrier dysfunction due to abnormal gut perfusion or hypoxemia in the context of low cardiac output or cyanosis. Intestinal dysbiosis may contribute to systemic inflammation thereby worsening clinical outcomes in this patient population. Despite significant advances in the management and survival of patients with CHD, morbidity remains significant and questions have arisen as to the role of the microbiome in the inflammatory process. Intestinal dysbiosis and barrier dysfunction experienced in this patient population are increasingly implicated in critical illness. This review highlights possible CHD-microbiome interactions, illustrates underlying signaling mechanisms, and discusses future directions and therapeutic translation of the basic research.
Collapse
|
12
|
Joshi RO, Chellappan S, Kukshal P. Exploring the Role of Maternal Nutritional Epigenetics in Congenital Heart Disease. Curr Dev Nutr 2020; 4:nzaa166. [PMID: 33294766 PMCID: PMC7703391 DOI: 10.1093/cdn/nzaa166] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Congenital heart disease (CHD) is one of the major debilitating birth defects resulting in significant impact on neonatal and child mortality globally. The etiology of CHD is complex and multifactorial. Many causative genes responsible for CHDs have been identified from the familial forms previously. Still, the non-Mendelian inheritance and predominant sporadic cases have stimulated research to understand the epigenetic basis and environmental impact on the incidence of CHD. The fetal epigenetic programming affecting cardiac development is susceptible to the availability of key dietary factors during the crucial periconceptional period. This article highlights the need and importance of in-depth research in the new emerging area of maternal nutritional epigenetics and CHD. It summarizes the current research and underlines the limitations in these types of studies. This review will benefit the future research on nutrition as a modifiable environmental factor to decrease the incidence of CHD.
Collapse
Affiliation(s)
- Radha O Joshi
- Department of Genomics Research, Sri Sathya Sai Sanjeevani Research Foundation, Palwal, Haryana, India
| | - Subramanian Chellappan
- Department of Anesthesia, Sri Sathya Sai Sanjeevani International Centre for Child Heart Care and Research, Palwal, Haryana, India
| | - Prachi Kukshal
- Department of Genomics Research, Sri Sathya Sai Sanjeevani Research Foundation, Palwal, Haryana, India
| |
Collapse
|
13
|
Li Y, Li X, Wang L, Han N, Yin G. miR-375-3p contributes to hypoxia-induced apoptosis by targeting forkhead box P1 (FOXP1) and Bcl2 like protein 2 (Bcl2l2) in rat cardiomyocyte h9c2 cells. Biotechnol Lett 2020; 43:353-367. [PMID: 33128129 DOI: 10.1007/s10529-020-03013-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
miRNAs have been pointed to play critical role in the development of congenital heart disease (CHD). miRNA-375-3p (miR-375-3p) was involved in cardiac dysfunction and cardiogenesis. However, no prior study had established a therapeutic role of miR-375-3p in CHD. We intended to investigate the effect and mechanism of miR-375-3p on apoptosis in hypoxic cardiomyocytes in vitro. Expression of miR-375-3p, forkhead box P1 (FOXP1) and Bcl2 like protein 2 (Bcl2l2) was detected using real-time quantitative PCR and western blot. Apoptosis was measured with MTT assay, flow cytometry and caspase-3 activity assay. The potential target binding between miR-375-3p and FOXP1/Bcl2l2 was predicted on DianaTools, and was validated by luciferase reporter assay and RNA pull-down assay. As a result, miR-375-3p was upregulated and FOXP1/Bcl2l2 was downregulated in maternal serum of women with fetal CHD and hypoxia-induced rat cardiomyocyte h9c2 cells. Hypoxia induced apoptosis rate elevation, caspase-3 activity promotion and viability inhibition in h9c2 cells; overexpression of miR-375-3p promoted, whereas knockdown of miR-375-3p antagonized hypoxia-induced effects in h9c2 cells. In addition, miR-375-3p was validated to negatively regulate FOXP1 and Bcl2l2 expression through target binding, and silencing of FOXP1 and Bcl2l2 could independently abate the anti-apoptosis role of miR-375-3p knockdown in hypoxic h9c2 cells. Collectively, blocking miR-375-3p suppressed hypoxia-evoked apoptosis of cardiomyocytes by targeting and upregulating FOXP1 and Bcl2l2. Our results might suggest maternal serum miR-375-3p as a potential biomarker for prenatal detection of fetal CHD.
Collapse
Affiliation(s)
- Yuefan Li
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China
| | - Xiaofei Li
- Department of Acupuncture, Qingdao Central Hospital, Qingdao, 266042, Shandong, China
| | - Ling Wang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Na Han
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China
| | - Gang Yin
- Department of Cardiology, Qingdao Central Hospital, No. 127, Siliu South Road, Qingdao, 266042, Shandong, China.
| |
Collapse
|
14
|
Chen P, Zhou C, Li B, Yang C. Circular RNA MGAT1 regulates cell proliferation and apoptosis in hypoxia-induced cardiomyocytes through miR-34a/YAP1 axis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2474-2486. [PMID: 33165436 PMCID: PMC7642720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Congenital heart disease (CHD) has severe morbidity and mortality worldwide. Evidence suggests that circularRNAs (circRNAs) are involved in the pathogenesis of human CHD. However, the regulatory mechanism remains uncertain. This study aimed to explore that mechanism. The levels of circular RNA MGAT1 (circMGAT1) and miR-34a were measured by quantitative polymerase chain reaction (qRT-PCR). Expression of yes-associated protein isoform 1 (YAP1) was assessed by western blot. Caspase-3 activity was evaluated by Caspase 3 Activity Assay Kit. CCK-8 assay was carried out to detect cell proliferation of hypoxia-induced AC16 cells. Cell apoptosis was analyzed by flow cytometry. In addition, dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were performed to verify the relationship between miR-34a and circMGAT1 or YAP1 in vitro. The level of circMGAT1 was downregulated, while miR-34a was strikingly increased in CHD tissues and hypoxia-induced AC16 cells. CircMGAT1 was a sponge of miR-34a, and circMGAT1 targeted miR-34a to regulate cell proliferation and apoptosis in hypoxia-induced cardiomyocytes. Dual-luciferase reporter and RIP-assay verified that miR-34a directly targeted YAP1, and the expression of YAP1 was significantly suppressed by miR-34a mimics but was enhanced by miR-34a inhibitor. Interestingly, YAP1 restored the effect of miR-34a on cell proliferation and apoptosis in hypoxia-induced AC16 cells. Besides, circMGAT1 sponged miR-34a to regulate the expression of YAP1. In conclusion, circMGAT1 inhibited cell apoptosis and enhanced cell proliferation by regulating the miR-34a/YAP1 axis, providing a therapy target for the treatment of human CHD.
Collapse
Affiliation(s)
- Pengyuan Chen
- Department of Pediatrics, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Chaoran Zhou
- Department of Pediatrics, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Bo Li
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Chao Yang
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| |
Collapse
|
15
|
Zheng J, Peng B, Zhang Y, Ai F, Hu X. FOXD3-AS1 Knockdown Suppresses Hypoxia-Induced Cardiomyocyte Injury by Increasing Cell Survival and Inhibiting Apoptosis via Upregulating Cardioprotective Molecule miR-150-5p In Vitro. Front Pharmacol 2020; 11:1284. [PMID: 32973515 PMCID: PMC7469905 DOI: 10.3389/fphar.2020.01284] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
Congenital heart disease (CHD) is the most common type of human innate malformation in fetuses. LncRNAs have been pointed to play critical regulatory roles in various types of cardiac development and diseases including CHD. Our study aimed to explore the effects of lncRNA forkhead box D3 antisense RNA 1 (FOXD3-AS1) on hypoxia-induced injury in AC16 cardiomyocytes and the related molecular mechanism. In vitro cell model of CHD was established by stimulating AC16 cells with hypoxia (1% O2). Expression of FOXD3-AS1 and miR-150-5p was detected by qRT-PCR. Hypoxia-induced injury was evaluated by detecting cell survival, lactate dehydrogenase (LDH) release, apoptosis, and caspase-3/7 activity using MTT, LDH assay, flow cytometry analysis, and caspase-3/7 activity assay, respectively. The regulatory relationship between FOXD3-AS1 and miR-150-5p was explored by luciferase reporter assay, RNA immunoprecipitation (RIP), and qRT-PCR. Results showed that hypoxia exposure caused an upregulation of FOXD3-AS1 and a downregulation of miR-150-5p in AC16 cells. Knockdown of FOXD3-AS1 attenuated reduction of cell survival and increase of LDH release, apoptosis, caspase-3/7 activity, and Bcl-2 associated X (Bax) expression induced by hypoxia in AC16 cells. Notably, we demonstrated that FOXD3-AS1 directly interacted with miR-150-5p to inhibit its expression. miR-150-5p knockdown reinforced the reduction of survival and induction of apoptosis by hypoxia and attenuated the effects of FOXD3-AS1 silencing on the same parameters in AC16 cells. In conclusion, FOXD3-AS1 knockdown protected AC16 cardiomyocytes from hypoxia-induced injury by increasing cell survival and inhibiting apoptosis through upregulating miR-150-5p.
Collapse
Affiliation(s)
- Jiayong Zheng
- Department of Children's Heart Center, Henan Province People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Bangtian Peng
- Department of Children's Heart Center, Henan Province People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Yanwei Zhang
- Department of Children's Heart Center, Henan Province People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Feng Ai
- Department of Children's Heart Center, Henan Province People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Xiaosong Hu
- Department of Children's Heart Center, Henan Province People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| |
Collapse
|
16
|
Circ_0068655 Promotes Cardiomyocyte Apoptosis via miR-498/PAWR Axis. Tissue Eng Regen Med 2020; 17:659-670. [PMID: 32767028 DOI: 10.1007/s13770-020-00270-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The cardiomyocyte apoptosis is considered as one of major contributions to cardiac remodeling after myocardial infarction (MI). Numerous studies find that circular RNAs (circRNAs) play pivotal roles in a variety of biological functions. However, the role of circ_0068655 in MI and human induced pluripotent stem-derived cardiomyocytes (HCMs) remains unknown. METHODS The expression of circ_0068655, miR-498, and PRKC apoptosis WT1 regulator (PAWR) in human MI heart tissues and hypoxia subjected HCMs was evaluated with qRT-PCR and Western blot. The effects of circ_0068655 on hypoxia-induced apoptotic death and cell migration in HCMs were evaluated with qRT-PCR, cell viability, cell death ELISA (POD), and Caspase-3 activity assay, and Trans-well assay, respectively. Furthermore, luciferase assay, qRT-PCR, biotin-labeled miRNA pulldown assay, and Western blot were employed in the functional studies. RESULTS We found that the expression of circ_0068655 and PAWR was enhanced in MI patients and hypoxia subjected HCMs; by contrast, the expression of miR-498 decreased. Inhibited expression of circ_0068655 in HMCs counteracted hypoxia-induced apoptotic death and impaired cell migration, in sharp contrast to circ_0068655 knockdown. We identified that circ_0068655 sponged an endogenous miR-498 to sequester and inhibit its activity, leading to the increased PAWR expression. CONCLUSIONS Our findings reveal that the expression of circ_0068655 can promote cardiomyocyte apoptosis through the modulation of miR-498-PAWR axis in vitro, which highlights the diagnostic and therapeutic value of circ_0068655 in patients with MI.
Collapse
|
17
|
Li X, Zhong J, Zeng Z, Wang H, Li J, Liu X, Yang X. MiR-181c protects cardiomyocyte injury by preventing cell apoptosis through PI3K/Akt signaling pathway. Cardiovasc Diagn Ther 2020; 10:849-858. [PMID: 32968640 DOI: 10.21037/cdt-20-490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Cardiomyocyte apoptosis plays an important role in the development of heart failure, which leads to high mortality in patients with cardiovascular diseases. In this study, we are focused to identify the role of miRNA-181c in the regulating of myocardial tissue apoptosis in the doxorubicin (DOX) or hypoxia/reoxygenation (H/R) induced H9C2 cardiomyocyte injury. Methods DOX-induced heart failure animal model was established using mice. Total RNA was extracted from tissue and cell using Trizol. RT-PCR was conducted for real-time RNA quantification. H9c2 cells were collected and labeled using an Annexin V-PI apoptosis kit. Flow cytometry was conducted to identify the cell apoptosis. Rat cardiomyocyte H9c2 cell was treated by 16 hours' hypoxia and 2 hours' reoxygenation to induce cell apoptosis. TUNEL assay was employed for myocardial tissue apoptosis analysis. Results It was revealed that miR-181c was suppressed on the heart tissue of DOX-induced heart failure animal model. We observed miR-181c overexpression reduced apoptosis through TUNEL assay, which suggested the inhibitory effect of miR-181c on myocardial tissue apoptosis. Transfection of miR-181c mimic could decrease cell apoptosis in H/R treated H9C2 cells in vitro. Under the stimulation of H/R or DOX, miR-181c could downregulate protein expression of Fas, IL-6 and TNF-α, and upregulated Bcl2 and the phosphorylation of Akt. Conclusions Our study revealed that miR-181c protected heart failure by impeding cardiomyocyte apoptosis through PI3K/Akt pathway, implying the therapeutic role of miR-181c during the exacerbation of the cardiovascular disease.
Collapse
Affiliation(s)
- Xiaoli Li
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jiuchang Zhong
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhen Zeng
- Geriatric Department, Chui Yang Liu Hospital Affiliated to Tsinghua University, Beijing, China
| | - Hongjiang Wang
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jing Li
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinchun Yang
- Department of Cardiology, Chaoyang Hospital affiliated to Capital Medical University, Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Abu-Halima M, Oberhoffer FS, El Rahman MA, Jung AM, Zemlin M, Rohrer TR, Kahraman M, Keller A, Meese E, Abdul-Khaliq H. Insights from circulating microRNAs in cardiovascular entities in turner syndrome patients. PLoS One 2020; 15:e0231402. [PMID: 32271829 PMCID: PMC7145016 DOI: 10.1371/journal.pone.0231402] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Turner syndrome (TS) is a chromosomal disorder, in which a female is partially or entirely missing one of the two X chromosomes, with a prevalence of 1:2500 live female births. The present study aims to identify a circulating microRNA (miRNA) signature for TS patients with and without congenital heart disease (CHD). Methods Microarray platform interrogating 2549 miRNAs were used to detect the miRNA abundance levels in the blood of 33 TS patients and 14 age-matched healthy volunteer controls (HVs). The differentially abundant miRNAs between the two groups were further validated by RT-qPCR. Results We identified 60 differentially abundant miRNA in the blood of TS patients compared to HVs, from which, 41 and 19 miRNAs showed a higher and a lower abundance levels in TS patients compared to HVs, respectively. RT-qPCR confirmed the significantly higher abundance levels of eight miRNAs namely miR-374b-5p, miR-199a-5p, miR-340-3p, miR-125b-5p, miR-30e-3p, miR-126-3p, miR-5695, and miR-26b-5p in TS patients as compared with the HVs. The abundance level of miR-5695 was higher in TS patients displaying CHD as compared to TS patients without CHD (p = 0.0265; log2-fold change 1.99); whereas, the abundance level of miR-126-3p was lower in TS patients with congenital aortic valve disease (AVD) compared to TS patients without BAV (p = 0.0139, log2-fold change 1.52). The clinical feature statistics revealed that miR-126-3p had a significant correlation with sinotubular junction Z-score (r = 0.42; p = 0.0154). Conclusion The identified circulating miRNAs signature for TS patients with manifestations associated with cardiovascular diseases provide new insights into the molecular mechanism of TS that may guide the development of novel diagnostic approaches.
Collapse
Affiliation(s)
- Masood Abu-Halima
- Institute of Human Genetics, Saarland University, Homburg/Saar, Germany
- * E-mail:
| | | | - Mohammed Abd El Rahman
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Anna-Maria Jung
- Department of Pediatric Endocrinology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Michael Zemlin
- Department of Pediatric Endocrinology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Tilman R. Rohrer
- Department of Pediatric Endocrinology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Mustafa Kahraman
- Chair for Clinical Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Homburg/Saar, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
19
|
Abstract
Cardiovascular diseases are one of the most common causes of death in both developing and developed countries worldwide. Even though there have been improvements in primary prevention, the prevalence of cardiovascular diseases continues to increase in recent years. Hence, it is crucial to both investigate the molecular pathophysiology of cardiovascular diseases in-depth and find novel biomarkers regarding the early and proper prevention and diagnosis of these diseases. MicroRNAs, or miRNAs, are endogenous, conserved, single-stranded non-coding RNAs of 21-25 nucleotides in length. miRNAs have important roles in various cellular events such as embryogenesis, proliferation, vasculogenesis, apoptosis, cell growth, differentiation, and tumorigenesis. They also have potential roles in the cardiovascular system, including angiogenesis, cardiac cell contractility, control of lipid metabolism, plaque formation, the arrangement of cardiac rhythm, and cardiac cell growth. Circulating miRNAs are promising novel biomarkers for purposes of the diagnosis and prognosis of cardiovascular diseases. Cell or tissue specificity, stability in serum or plasma, resistance to degradative factors such as freeze-thaw cycles or enzymes in the blood, and fast-release kinetics, provide the potential for miRNAs to be surrogate markers for the early and accurate diagnosis of disease and for predicting middle- or long-term prognosis. Moreover, it may be a logical approach to combine miRNAs with traditional biomarkers to improve risk stratification and long-term prognosis. In addition to their efficacy in both diagnosis and prognosis, miRNA-based therapeutics may be beneficial for treating cardiovascular diseases using novel platforms and computational tools and in combination with traditional methods of analysis. microRNAs are promising, novel therapeutic agents, which can affect multiple genes using different signaling pathways. miRNAs therapeutic modulation techniques have been used in the settings of atherosclerosis, acute myocardial infarction, restenosis, vascular remodeling, arrhythmias, hypertrophy and fibrosis, angiogenesis and cardiogenesis, aortic aneurysm, pulmonary hypertension, and ischemic injury. This review presents detailed information about miRNAs regarding structure and biogenesis, stages of synthesis and functions, expression profiles in serum/plasma of living organisms, diagnostic and prognostic potential as novel biomarkers, and therapeutic applications in various diseases.
Collapse
Affiliation(s)
| | - Mehmet Demir
- Department of Cardiology, University of Health Sciences, Bursa Yüksek İhtisas Research and Training Hospital, Bursa, Turkey
| |
Collapse
|
20
|
Raggi F, Cangelosi D, Becherini P, Blengio F, Morini M, Acquaviva M, Belli ML, Panizzon G, Cervo G, Varesio L, Eva A, Bosco MC. Transcriptome analysis defines myocardium gene signatures in children with ToF and ASD and reveals disease-specific molecular reprogramming in response to surgery with cardiopulmonary bypass. J Transl Med 2020; 18:21. [PMID: 31924244 PMCID: PMC6954611 DOI: 10.1186/s12967-020-02210-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 01/03/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tetralogy of Fallot (ToF) and Atrial Septal Defects (ASD) are the most common types of congenital heart diseases and a major cause of childhood morbidity and mortality. Cardiopulmonary bypass (CPB) is used during corrective cardiac surgery to support circulation and heart stabilization. However, this procedure triggers systemic inflammatory and stress response and consequent increased risk of postoperative complications. The aim of this study was to define the molecular bases of ToF and ASD pathogenesis and response to CPB and identify new potential biomarkers. METHODS Comparative transcriptome analysis of right atrium specimens collected from 10 ToF and 10 ASD patients was conducted before (Pre-CPB) and after (Post-CPB) corrective surgery. Total RNA isolated from each sample was individually hybridized on Affymetrix HG-U133 Plus Array Strips containing 38,500 unique human genes. Differences in the gene expression profiles and functional enrichment/network analyses were assessed using bioinformatic tools. qRT-PCR analysis was used to validate gene modulation. RESULTS Pre-CPB samples showed significant differential expression of a total of 72 genes, 28 of which were overexpressed in ToF and 44 in ASD. According to Gene Ontology annotation, the mostly enriched biological processes were represented by matrix organization and cell adhesion in ToF and by muscle development and contractility in ASD specimens. GSEA highlighted the specific enrichment of hypoxia gene sets in ToF samples, pointing to a role for hypoxia in disease pathogenesis. The post-CPB myocardium exhibited significant alterations in the expression profile of genes related to transcription regulation, growth/apoptosis, inflammation, adhesion/matrix organization, and oxidative stress. Among them, only 70 were common to the two disease groups, whereas 110 and 24 were unique in ToF and ASD, respectively. Multiple functional interactions among differentially expressed gene products were predicted by network analysis. Interestingly, gene expression changes in ASD samples followed a consensus hypoxia profile. CONCLUSION Our results provide a comprehensive view of gene reprogramming in right atrium tissues of ToF and ASD patients before and after CPB, defining specific molecular pathways underlying disease pathophysiology and myocardium response to CPB. These findings have potential translational value because they identify new candidate prognostic markers and targets for tailored cardioprotective post-surgical therapies.
Collapse
Affiliation(s)
- Federica Raggi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Davide Cangelosi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Pamela Becherini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy
| | - Fabiola Blengio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,INSERM U955 Equipe 16, Creteil, France
| | - Martina Morini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Massimo Acquaviva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology INSPE, Ospedale San Raffaele, Milano, Italy
| | - Maria Luisa Belli
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Cytomorphology Laboratory, Heamo-Onco-TMO Department, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Panizzon
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Cervo
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.
| |
Collapse
|
21
|
Lv Y, Liu Z, Huang J, Yu J, Dong Y, Wang J. LncRNA nuclear-enriched abundant transcript 1 regulates hypoxia-evoked apoptosis and autophagy via mediation of microRNA-181b. Mol Cell Biochem 2019; 464:193-203. [PMID: 31853799 DOI: 10.1007/s11010-019-03660-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/16/2019] [Indexed: 12/22/2022]
Abstract
Nuclear-enriched abundant transcript 1 (NEAT1), a vital long noncoding RNA (lncRNA), exhibits the functions in disparate cancers. Nevertheless, the influences of NEAT1 in congenital heart disease (CHD) remain unreported. The research delves into whether NEAT1 affects H9c2 cells apoptosis and autophagy under the hypoxia condition. Overexpressed NEAT1 vector was transfected into H9c2 cells; then, functions of NEAT1 in cell viability, apoptosis, autophagy, PI3K/AKT/mTOR and JAK1/STAT3 pathways were detected in H9c2 cells under hypoxia condition. Expression of NEAT1 and miR-181b in hypoxia and blood samples from CHD was evaluated. After miR-181b inhibitor transfection, functions of miR-181b repression in the above-mentioned cell behavior and PI3K/AKT/mTOR and JAK1/STAT3 pathways were reassessed. Overexpressed NEAT1 clearly allayed hypoxia-triggered H9c2 cells apoptosis and autophagy. The decreased NEAT1 and miR-181b were showcased in hypoxia and blood samples from CHD; meanwhile, elevated miR-181b evoked by overexpressed NEAT1 was observed in hypoxia-managed H9c2 cells. More importantly, miR-181b inhibition obviously overturned the influences of NEAT1 in hypoxia-affected H9c2 cells apoptosis and autophagy. Besides, overexpressed NEAT1 facilitated PI3K/AKT/mTOR and JAK1/STAT3 activations via enhancing miR-181b. The research exposed that NEAT1 eased hypoxia-triggered H9c2 cells apoptosis and autophagy by expediting PI3K/AKT/mTOR and JAK1/STAT3 pathways via elevating miR-181b.
Collapse
Affiliation(s)
- Ying Lv
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, 050031, Hebei, China
| | - Zhaoming Liu
- Department of Pediatric Surgery, Shijiazhuang Maternity & Child Healthcare Hospital, No. 9 Jianguo Road, Shijiazhuang, 050051, Hebei, China
| | - Jiancheng Huang
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, 050031, Hebei, China
| | - Jie Yu
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, 050031, Hebei, China
| | - Yanbo Dong
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, 050031, Hebei, China
| | - Jun Wang
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, 050031, Hebei, China.
| |
Collapse
|
22
|
Abu-Halima M, Weidinger J, Poryo M, Henn D, Keller A, Meese E, Abdul-Khaliq H. Micro-RNA signatures in monozygotic twins discordant for congenital heart defects. PLoS One 2019; 14:e0226164. [PMID: 31805172 PMCID: PMC6894838 DOI: 10.1371/journal.pone.0226164] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Background MicroRNAs (miRNAs) are small RNAs regulating gene expression post-transcriptionally. Recent studies demonstrated that miRNAs are involved in the development of congenital heart defects (CHD). In this study, we aimed at identifying the specific patterns of miRNAs in blood of monozygotic twin pairs discordant for CHD and to assess whether miRNAs might be involved in the development or reflect the consequences of CHD. Methods miRNA microarray analysis and Real-Time Quantitative PCR (RT-qPCR) were employed to determine the miRNA abundance level from 12 monozygotic twins discordant for CHD and their non-CHD co-twins (n = 12). Enrichment analyses of altered miRNAs were performed using bioinformatics tools. Results Compared with non-CHD co-twins, profiling analysis indicated 34 miRNAs with a significant difference in abundance level (p<0.05, fold change ≥ 1.3), of which 11 miRNAs were up-regulated and 23 miRNAs were down-regulated. Seven miRNAs were validated with RT-qPCR including miR-511-3p, miR-1306-5p, miR-421, miR-4707-3p, miR-4732-3p, miR-5189-3p, and miR-890, and the results were consistent with microarray analysis. Five miRNAs namely miR-511-3p, miR-1306-5p, miR-4732-3p, miR-5189-3p, and miR-890 were found to be significantly up-regulated in twins < 10 years old. Bioinformatics analysis showed that the 7 validated miRNAs were involved in phosphatidylinositol signaling, gap junction signaling, and adrenergic signaling in cardiomyocytes. Conclusions Our data show deregulated miRNA abundance levels in the peripheral blood of monozygotic twins discordant for CHD, and identify new candidates for further analysis, which may contribute to understanding the development of CHD in the future. Bioinformatics analysis indicated that the target genes of these miRNAs are likely involved in signaling and communication of cardiomyocytes.
Collapse
Affiliation(s)
- Masood Abu-Halima
- Institute of Human Genetics, Saarland University, Homburg/Saar, Germany
| | - Josephin Weidinger
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dominic Henn
- Department of Hand, Plastic and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Homburg/Saar, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
23
|
Vizitiu AC, Stambouli D, Pavel AG, Muresan MC, Anastasiu DM, Bejinar C, Alexa A, Marian C, Sirbu IO, Sima L. Mature miR-99a Upregulation in the Amniotic Fluid Samples from Female Fetus Down Syndrome Pregnancies: A Pilot Study. ACTA ACUST UNITED AC 2019; 55:medicina55110728. [PMID: 31703316 PMCID: PMC6915350 DOI: 10.3390/medicina55110728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/25/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Background and Objective: Although Down syndrome is the most frequent aneuploidy, its pathogenic molecular mechanisms are not yet fully understood. The aim of our study is to quantify-by qRT-PCR-the expression levels of both the mature forms and the pri-miRNAs of the microRNAs resident on chromosome 21 (miR(21)) in the amniotic fluid samples from Down syndrome singleton pregnancies and to estimate the impact of the differentially expressed microRNAs on Down syndrome fetal heart and amniocytes transcriptomes. Materials and methods: We collected amniotic fluid samples harvested by trained obstetricians as part of the second trimester screening/diagnostic procedure for aneuploidies to assess the trisomy 21 status by QF-PCR and karyotyping. Next, we evaluated-by Taqman qRT-PCR-the expression levels of both the mature forms and the pri-miRNA precursors of the microRNAs resident on chromosome 21 in amniotic fluid samples from singleton Down syndrome and euploid pregnancies. Further, we combined miRWalk 3.0 microRNA target prediction with GEO DataSets analysis to estimate the impact of hsa-miR-99a abnormal expression on Down syndrome heart and amniocytes transcriptome. Results: We found a statistically significant up-regulation of the mature form of miR-99a, but not pri-miR-99a, in the amniotic fluid samples from Down syndrome pregnancies with female fetuses. GATHER functional enrichment analysis of miRWalk3.0-predicted targets from Down syndrome amniocytes and fetal hearts transcriptome GEODataSets outlined both focal adhesion and cytokine-cytokine receptor interaction signaling as novel signaling pathways impacted by miR-99a and associated with cardiac defects in female Down syndrome patients. Conclusions: The significant overexpression of miR-99a, but not pri-miR-99a, points towards an alteration of the post-transcriptional mechanisms of hsa-miR-99a maturation and/or stability in the female trisomic milieu, with a potential impact on signaling pathways important for proper development of the heart.
Collapse
Affiliation(s)
- Anda-Cornelia Vizitiu
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, Eftimie Murgu Nr. 2, Timisoara 300041, Romania;
| | - Danae Stambouli
- CytoGenomic Medical Laboratory, Calea Floreasca Nr. 35, Sector 1, Bucharest 014451, Romania; (D.S.); (A.-G.P.)
| | - Anca-Gabriela Pavel
- CytoGenomic Medical Laboratory, Calea Floreasca Nr. 35, Sector 1, Bucharest 014451, Romania; (D.S.); (A.-G.P.)
| | - Maria-Cezara Muresan
- Obstetrics and Gynecology Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania (D.M.A.)
| | - Diana Maria Anastasiu
- Obstetrics and Gynecology Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania (D.M.A.)
| | - Cristina Bejinar
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania; (C.B.); (A.A.); (C.M.)
| | - Anda Alexa
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania; (C.B.); (A.A.); (C.M.)
| | - Catalin Marian
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania; (C.B.); (A.A.); (C.M.)
| | - Ioan Ovidiu Sirbu
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania; (C.B.); (A.A.); (C.M.)
- Correspondence: ; Tel.: +40-756-136-272
| | - Laurentiu Sima
- Surgical Semiology Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, Timisoara 300041, Romania;
| |
Collapse
|
24
|
Zhang H, Liu L, Tian J. Molecular mechanisms of congenital heart disease in down syndrome. Genes Dis 2019; 6:372-377. [PMID: 31832516 PMCID: PMC6889238 DOI: 10.1016/j.gendis.2019.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022] Open
Abstract
Down syndrome (DS), as a typical genomic aneuploidy, is a common cause of various birth defects, among which is congenital heart disease (CHD). 40-60% neonates with DS have some kinds of CHD. However, the molecular pathogenic mechanisms of DS associated CHD are still not fully understood. This review summarizes available studies on DS associated CHD from seven aspects so as to provide a crucial and updated overview of what we known so far in this domain.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| | - Lingjuan Liu
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| | - Jie Tian
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| |
Collapse
|
25
|
Nagy O, Baráth S, Ujfalusi A. The role of microRNAs in congenital heart disease. EJIFCC 2019; 30:165-178. [PMID: 31263391 PMCID: PMC6599193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Congenital heart diseases (CHDs) are the leading inherited cause of perinatal and infant mortality. CHD refers to structural anomalies of the heart and blood vessels that arise during cardiac development and represents a broad spectrum of malformations, including septal and valve defects, lesions affecting the outflow tract and ventricules. Advanced treatment strategies have greatly improved life expectancy and led to expanded population of adult patients with CHD. Thus, a better understanding of the pathogenesis and molecular mechanisms underlying CHDs is essential to improve the diagnosis and prognosis of patients. The etiology of CHD is largely unknown, genetic and environmental factors may contribute to the disease. In addition to the mutations affecting genomic DNA, epigenetic changes are being increasingly acknowledged as key factors in the development and progression of CHDs. The posttranscriptional regulation of gene expression by microRNAs (miRs) controls the highly complex multi-cell lineage process of cardiac tissue formation. In recent years, multiplex experimental models have provided evidence that changes in expression levels of miRs are associated with human cardiovascular disease, including CHD. The newly described correlations between miRs and heart development suggest the potential importance of miRs as diagnostic markers in human cardiovascular diseases. In the future, more intensive research is likely to be carried out to clarify their contribution to personalized management and treatment of CHD patients. In this paper, we discuss the current knowledge on the causative role of miRs in cardiac development and CHDs.
Collapse
Affiliation(s)
| | | | - Anikó Ujfalusi
- Corresponding author: Anikó Ujfalusi Department of Laboratory Medicine Faculty of Medicine University of Debrecen Nagyerdei krt. 98. Debrecen H-4032 Hungary Phone: +36 52 340 006 Fax: +36 52 417 631 E-mail:
| |
Collapse
|
26
|
Dueñas A, Expósito A, Aranega A, Franco D. The Role of Non-Coding RNA in Congenital Heart Diseases. J Cardiovasc Dev Dis 2019; 6:E15. [PMID: 30939839 PMCID: PMC6616598 DOI: 10.3390/jcdd6020015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular development is a complex developmental process starting with the formation of an early straight heart tube, followed by a rightward looping and the configuration of atrial and ventricular chambers. The subsequent step allows the separation of these cardiac chambers leading to the formation of a four-chambered organ. Impairment in any of these developmental processes invariably leads to cardiac defects. Importantly, our understanding of the developmental defects causing cardiac congenital heart diseases has largely increased over the last decades. The advent of the molecular era allowed to bridge morphogenetic with genetic defects and therefore our current understanding of the transcriptional regulation of cardiac morphogenesis has enormously increased. Moreover, the impact of environmental agents to genetic cascades has been demonstrated as well as of novel genomic mechanisms modulating gene regulation such as post-transcriptional regulatory mechanisms. Among post-transcriptional regulatory mechanisms, non-coding RNAs, including therein microRNAs and lncRNAs, are emerging to play pivotal roles. In this review, we summarize current knowledge on the functional role of non-coding RNAs in distinct congenital heart diseases, with particular emphasis on microRNAs and long non-coding RNAs.
Collapse
Affiliation(s)
- Angel Dueñas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Almudena Expósito
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Amelia Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain.
| |
Collapse
|
27
|
Radhakrishna U, Albayrak S, Zafra R, Baraa A, Vishweswaraiah S, Veerappa AM, Mahishi D, Saiyed N, Mishra NK, Guda C, Ali-Fehmi R, Bahado-Singh RO. Placental epigenetics for evaluation of fetal congenital heart defects: Ventricular Septal Defect (VSD). PLoS One 2019; 14:e0200229. [PMID: 30897084 PMCID: PMC6428297 DOI: 10.1371/journal.pone.0200229] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 03/11/2019] [Indexed: 12/19/2022] Open
Abstract
Ventricular Septal Defect (VSD), the most common congenital heart defect, is characterized by a hole in the septum between the right and left ventricles. The pathogenesis of VSD is unknown in most clinical cases. There is a paucity of data relevant to epigenetic changes in VSD. The placenta is a fetal tissue crucial in cardiac development and a potentially useful surrogate for evaluating the development of heart tissue. To understand epigenetic mechanisms that may play a role in the development of VSD, genome-wide DNA methylation assay on placentas of 8 term subjects with isolated VSD and no known or suspected genetic syndromes and 10 unaffected controls was performed using the Illumina HumanMethylation450 BeadChip assay. We identified a total of 80 highly accurate potential CpGs in 80 genes for detection of VSD; area under the receiver operating characteristic curve (AUC ROC) 1.0 with significant 95% CI (FDR) p-values < 0.05 for each individual locus. The biological processes and functions for many of these differentially methylated genes are previously known to be associated with heart development or disease, including cardiac ventricle development (HEY2, ISL1), heart looping (SRF), cardiac muscle cell differentiation (ACTC1, HEY2), cardiac septum development (ISL1), heart morphogenesis (SRF, HEY2, ISL1, HEYL), Notch signaling pathway (HEY2, HEYL), cardiac chamber development (ISL1), and cardiac muscle tissue development (ACTC1, ISL1). In addition, we identified 8 microRNAs that have the potential to be biomarkers for the detection of VSD including: miR-191, miR-548F1, miR-148A, miR-423, miR-92B, miR-611, miR-2110, and miR-548H4. To our knowledge this is the first report in which placental analysis has been used for determining the pathogenesis of and predicting VSD.
Collapse
Affiliation(s)
- Uppala Radhakrishna
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, Michigan, United States of America
- * E-mail:
| | - Samet Albayrak
- Department of Obstetrics and Gynaecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Rita Zafra
- Department of Obstetrics and Gynaecology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Alosh Baraa
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Sangeetha Vishweswaraiah
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, Michigan, United States of America
| | - Avinash M. Veerappa
- Department of Studies in Genetics and Genomics, Laboratory of Genomic Sciences, University of Mysore, Mysore, India
| | - Deepthi Mahishi
- Department of Studies in Genetics and Genomics, Laboratory of Genomic Sciences, University of Mysore, Mysore, India
| | - Nazia Saiyed
- Biotechnology, Nirma Institute of Science, Nirma University, Ahmedabad, India
| | - Nitish K. Mishra
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Centre Omaha, Nebraska, United States of America
| | - Chittibabu Guda
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Centre Omaha, Nebraska, United States of America
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Ray O. Bahado-Singh
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, Michigan, United States of America
| |
Collapse
|
28
|
A Matched Case-Control Study on the Association Between Colds, Depressive Symptoms during Pregnancy and Congenital Heart Disease in Northwestern China. Sci Rep 2019; 9:589. [PMID: 30679633 PMCID: PMC6345882 DOI: 10.1038/s41598-018-36968-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/28/2018] [Indexed: 11/09/2022] Open
Abstract
The purpose of this study was to explore the association between colds, depressive symptoms during pregnancy and offspring congenital heart disease (CHD). A 1:2 matching case-control study was conducted in Northwest China. Information was gathered by a structured questionnaire and was reviewed by investigators on the spot. Multivariate logistic regressions and nonlinear mixed effect model were performed. 614 cases and 1228 controls were available in this study. After adjusting for potential confounders, the colds during the entire pregnancy were associated with increased risk of offspring CHD (OR = 1.44(1.12-1.85)). Similarly, there was a higher depression score in CHD group than the control group (OR = 1.89(1.48-2.41)). In addition, the women with both colds and higher depression scores had a higher risk of offspring CHD (OR = 2.72(1.87-3.93)) than their counterparts with only colds (OR = 1.48(1.04-2.09)) or with only higher depression scores (OR = 1.94(1.37-2.74)). The combined effects were significant in the multiplication model (OR = 2.04(1.47-2.83)) but not in the additive model (S = 1.40(0.70-2.81), AP = 0.19(-0.15-0.53) and RERI = 0.55(-0.54-1.64)). In conclusion, the colds and depressive symptoms during pregnancy were found associated with increased risk of offspring CHD and we found for the first time that there existed a statistically multiplying interaction effect of colds and depression on increasing risk of offspring CHD.
Collapse
|
29
|
Mantakaki A, Fakoya AOJ, Sharifpanah F. Recent advances and challenges on application of tissue engineering for treatment of congenital heart disease. PeerJ 2018; 6:e5805. [PMID: 30386701 PMCID: PMC6204240 DOI: 10.7717/peerj.5805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022] Open
Abstract
Congenital heart disease (CHD) affects a considerable number of children and adults worldwide. This implicates not only developmental disorders, high mortality, and reduced quality of life but also, high costs for the healthcare systems. CHD refers to a variety of heart and vascular malformations which could be very challenging to reconstruct the malformed region surgically, especially when the patient is an infant or a child. Advanced technology and research have offered a better mechanistic insight on the impact of CHD in the heart and vascular system of infants, children, and adults and identified potential therapeutic solutions. Many artificial materials and devices have been used for cardiovascular surgery. Surgeons and the medical industry created and evolved the ball valves to the carbon-based leaflet valves and introduced bioprosthesis as an alternative. However, with research further progressing, contracting tissue has been developed in laboratories and tissue engineering (TE) could represent a revolutionary answer for CHD surgery. Development of engineered tissue for cardiac and aortic reconstruction for developing bodies of infants and children can be very challenging. Nevertheless, using acellular scaffolds, allograft, xenografts, and autografts is already very common. Seeding of cells on surface and within scaffold is a key challenging factor for use of the above. The use of different types of stem cells has been investigated and proven to be suitable for tissue engineering. They are the most promising source of cells for heart reconstruction in a developing body, even for adults. Some stem cell types are more effective than others, with some disadvantages which may be eliminated in the future.
Collapse
Affiliation(s)
| | | | - Fatemeh Sharifpanah
- Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany
| |
Collapse
|
30
|
Islas JF, Moreno-Cuevas JE. A MicroRNA Perspective on Cardiovascular Development and Diseases: An Update. Int J Mol Sci 2018; 19:E2075. [PMID: 30018214 PMCID: PMC6073753 DOI: 10.3390/ijms19072075] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/02/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
In this review, we summarize the latest research pertaining to MicroRNAs (miRs) related to cardiovascular diseases. In today's molecular age, the key clinical aspects of diagnosing and treating these type of diseases are crucial, and miRs play an important role. Therefore, we have made a thorough analysis discussing the most important candidate protagonists of many pathways relating to such conditions as atherosclerosis, heart failure, myocardial infarction, and congenital heart disorders. We approach miRs initially from the fundamental molecular aspects and look at their role in developmental pathways, as well as regulatory mechanisms dysregulated under specific cardiovascular conditions. By doing so, we can better understand their functional roles. Next, we look at therapeutic aspects, including delivery and inhibition techniques. We conclude that a personal approach for treatment is paramount, and so understanding miRs is strategic for cardiovascular health.
Collapse
Affiliation(s)
- Jose Francisco Islas
- Tecnologico de Monterrey, Grupo de Investigación con Enfoque Estratégico en Bioingeniería y Medicina Regenerativa, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey, NL 64710, Mexico.
| | - Jorge Eugenio Moreno-Cuevas
- Tecnologico de Monterrey, Grupo de Investigación con Enfoque Estratégico en Bioingeniería y Medicina Regenerativa, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey, NL 64710, Mexico.
| |
Collapse
|
31
|
Zhong J, Wang S, Shen WB, Kaushal S, Yang P. The current status and future of cardiac stem/progenitor cell therapy for congenital heart defects from diabetic pregnancy. Pediatr Res 2018; 83:275-282. [PMID: 29016556 PMCID: PMC5876137 DOI: 10.1038/pr.2017.259] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023]
Abstract
Pregestational maternal diabetes induces congenital heart defects (CHDs). Cardiac dysfunction after palliative surgical procedures contributes to the high mortality of CHD patients. Autologous or allogeneic stem cell therapies are effective for improving cardiac function in animal models and clinical trials. c-kit+ cardiac progenitor cells (CPCs), the most recognized CPCs, have the following basic properties of stem cells: self-renewal, multicellular clone formation, and differentiation into multiple cardiac lineages. However, there is ongoing debate regarding whether c-kit+ CPCs can give rise to sufficient cardiomyocytes. A new hypothesis to address the beneficial effect of c-kit+ CPCs is that these cells stimulate endogenous cardiac cells through a paracrine function in producing a robust secretome and exosomes. The values of other cardiac CPCs, including Sca1+ CPCs and cardiosphere-derived cells, are beginning to be revealed. These cells may be better choices than c-kit+ CPCs for generating cardiomyocytes. Adult mesenchymal stem cells are considered immune-incompetent and effective for improving cardiac function. Autologous CPC therapy may be limited by the observation that maternal diabetes adversely affects the biological function of embryonic stem cells and CPCs. Future studies should focus on determining the mechanistic action of these cells, identifying new CPC markers, selecting highly effective CPCs, and engineering cell-free products.
Collapse
Affiliation(s)
- Jianxiang Zhong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shengbing Wang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sunjay Kaushal
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Kaul Z, Chakrabarti O. Tumor susceptibility gene 101 regulates predisposition to apoptosis via ESCRT machinery accessory proteins. Mol Biol Cell 2017; 28:2106-2122. [PMID: 28539405 PMCID: PMC5509423 DOI: 10.1091/mbc.e16-12-0855] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/11/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022] Open
Abstract
ESCRT proteins are implicated in myriad cellular processes, including endosome formation, fusion of autophagosomes/amphisomes with lysosomes, and apoptosis. The role played by these proteins in either facilitating or protecting against apoptosis is unclear. In this study, while trying to understand how deficiency of Mahogunin RING finger 1 (MGRN1) affects cell viability, we uncovered a novel role for its interactor, the ESCRT-I protein TSG101: it directly participates in mitigating ER stress-mediated apoptosis. The association of TSG101 with ALIX prevents predisposition to apoptosis, whereas ALIX-ALG-2 interaction favors a death phenotype. Altered Ca2+ homeostasis in cells and a simultaneous increase in the protein levels of ALIX and ALG-2 are required to elicit apoptosis by activating ER stress-associated caspase 4/12. We further demonstrate that in the presence of membrane-associated, disease-causing prion protein CtmPrP, increased ALIX and ALG-2 levels are detected along with ER stress markers and associated caspases in transgenic brain lysates and cells. These effects were rescued by overexpression of TSG101. This is significant because MGRN1 deficiency is closely associated with neurodegeneration and prenatal and neonatal mortality, which could be due to excess cell death in selected brain regions or myocardial apoptosis during embryonic development.
Collapse
Affiliation(s)
- Zenia Kaul
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata 700064, India
| | - Oishee Chakrabarti
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata 700064, India
| |
Collapse
|
33
|
Abu-Halima M, Meese E, Keller A, Abdul-Khaliq H, Rädle-Hurst T. Analysis of circulating microRNAs in patients with repaired Tetralogy of Fallot with and without heart failure. J Transl Med 2017; 15:156. [PMID: 28693530 PMCID: PMC5504636 DOI: 10.1186/s12967-017-1255-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/24/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are a class of regulatory RNAs that regulate gene expression post-transcriptionally. Little, however, is known on the expression profile of circulating miRNAs in Tetralogy of Fallot (TOF) patients late after surgical repair. In this study, we aimed to identify the specific patterns of circulating miRNAs in blood of patients with repaired, non-syndromic TOF and to assess whether these specific miRNAs may be useful to differentiate patients with and without heart failure. METHODS SurePrint™ 8 × 60 K Human v16 miRNA arrays were used to determine miRNA expression profiles in 15 healthy controls and 37 patients after TOF repair of whom 3 had symptomatic right heart failure. The expression levels of selected miRNAs have been validated by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Enrichment analyses of altered miRNA expression were predicted using bioinformatic tools. RESULTS Compared with healthy controls, a total of 49, 58 and 77 miRNAs were found to be significantly altered in TOF patients (TOF-all), TOF patients with (TOF-HF) and without symptomatic right heart failure (TOF-noHF) (>2.0-fold change, adjusted P < 0.05), respectively. Three miRNAs namely miR-181d-5p, miR-206 and miR-625-5p were validated by RT-qPCR in all TOF groups. The area under the receiver operating characteristic curve (AUC) for miR-181d-5p, miR-206 and miR-625-5p were 0.987, 0.993 and 0.769 in TOF-all and 0.990, 0.994 and 0.749 in TOF-noHF, respectively. Moreover, expression levels of miR-625-5p, miR-1233-3p and miR-421 were lower in TOF-HF compared to TOF-noHF (P = 0.012). CONCLUSIONS Altered expression levels of circulating miRNAs were found in TOF patients late after surgical repair and are different to those seen in the right ventricular myocardium of infants with TOF. Expression levels of miR-421, miR-1233-3p and miR-625-5p are lower in TOF patients with symptomatic right heart failure and thus may indicate disease progression in these patients.
Collapse
Affiliation(s)
- Masood Abu-Halima
- Department of Human Genetics, Saarland University, 66421, Homburg/Saar, Germany. .,Department of Human Genetics, Saarland University Medical Center, Kirrberger Straße 100, 66421, Homburg/Saar, Germany.
| | - Eckart Meese
- Department of Human Genetics, Saarland University, 66421, Homburg/Saar, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66041, Saarbruecken, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421, Homburg/Saar, Germany
| | - Tanja Rädle-Hurst
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421, Homburg/Saar, Germany
| |
Collapse
|
34
|
Touma M, Reemtsen B, Halnon N, Alejos J, Finn JP, Nelson SF, Wang Y. A Path to Implement Precision Child Health Cardiovascular Medicine. Front Cardiovasc Med 2017; 4:36. [PMID: 28620608 PMCID: PMC5451507 DOI: 10.3389/fcvm.2017.00036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022] Open
Abstract
Congenital heart defects (CHDs) affect approximately 1% of live births and are a major source of childhood morbidity and mortality even in countries with advanced healthcare systems. Along with phenotypic heterogeneity, the underlying etiology of CHDs is multifactorial, involving genetic, epigenetic, and/or environmental contributors. Clear dissection of the underlying mechanism is a powerful step to establish individualized therapies. However, the majority of CHDs are yet to be clearly diagnosed for the underlying genetic and environmental factors, and even less with effective therapies. Although the survival rate for CHDs is steadily improving, there is still a significant unmet need for refining diagnostic precision and establishing targeted therapies to optimize life quality and to minimize future complications. In particular, proper identification of disease associated genetic variants in humans has been challenging, and this greatly impedes our ability to delineate gene–environment interactions that contribute to the pathogenesis of CHDs. Implementing a systematic multileveled approach can establish a continuum from phenotypic characterization in the clinic to molecular dissection using combined next-generation sequencing platforms and validation studies in suitable models at the bench. Key elements necessary to advance the field are: first, proper delineation of the phenotypic spectrum of CHDs; second, defining the molecular genotype/phenotype by combining whole-exome sequencing and transcriptome analysis; third, integration of phenotypic, genotypic, and molecular datasets to identify molecular network contributing to CHDs; fourth, generation of relevant disease models and multileveled experimental investigations. In order to achieve all these goals, access to high-quality biological specimens from well-defined patient cohorts is a crucial step. Therefore, establishing a CHD BioCore is an essential infrastructure and a critical step on the path toward precision child health cardiovascular medicine.
Collapse
Affiliation(s)
- Marlin Touma
- Department of Pediatrics, Children's Discovery and Innovation Institute, University of California at Los Angeles, Los Angeles, CA, United States.,Cardiovascular Research Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - Brian Reemtsen
- Department of Cardiothoracic Surgery, University of California at Los Angeles, Los Angeles, CA, United States
| | - Nancy Halnon
- Department of Pediatrics, University of California at Los Angeles, Los Angeles, CA, United States
| | - Juan Alejos
- Department of Pediatrics, University of California at Los Angeles, Los Angeles, CA, United States
| | - J Paul Finn
- Department of Radiology, Cardiovascular Imaging, University of California at Los Angeles, Los Angeles, CA, United States
| | - Stanley F Nelson
- Department of Human Genetics, University of California at Los Angeles, Los Angeles, CA, United States
| | - Yibin Wang
- Cardiovascular Research Laboratory, University of California at Los Angeles, Los Angeles, CA, United States.,Department of Anesthesiology, Physiology and Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
35
|
Hoelscher SC, Doppler SA, Dreßen M, Lahm H, Lange R, Krane M. MicroRNAs: pleiotropic players in congenital heart disease and regeneration. J Thorac Dis 2017; 9:S64-S81. [PMID: 28446969 DOI: 10.21037/jtd.2017.03.149] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital heart disease (CHD) is the leading cause of infant death, affecting approximately 4-14 live births per 1,000. Although surgical techniques and interventions have improved significantly, a large number of infants still face poor clinical outcomes. MicroRNAs (miRs) are known to coordinately regulate cardiac development and stimulate pathological processes in the heart, including fibrosis or hypertrophy and impair angiogenesis. Dysregulation of these regulators could therefore contribute (I) to the initial development of CHD and (II) at least partially to the observed clinical outcomes of many CHD patients by stimulating the aforementioned pathways. Thus, miRs may exhibit great potential as therapeutic targets in regenerative medicine. In this review we provide an overview of miR function and elucidate their role in selected CHDs, including hypoplastic left heart syndrome (HLHS), tetralogy of Fallot (TOF), ventricular septal defects (VSDs) and Holt-Oram syndrome (HOS). We then bridge this knowledge to the potential usefulness of miRs and/or their targets in therapeutic strategies for regenerative purposes in CHDs.
Collapse
Affiliation(s)
- Sarah C Hoelscher
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Stefanie A Doppler
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Martina Dreßen
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Harald Lahm
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Rüdiger Lange
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
36
|
Liang Q, Gong W, Zheng D, Zhong R, Wen Y, Wang X. The influence of maternal exposure history to virus and medicine during pregnancy on congenital heart defects of fetus. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:5628-5632. [PMID: 28039625 DOI: 10.1007/s11356-016-8198-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/01/2016] [Indexed: 05/09/2023]
Abstract
Congenital heart disease (CHD) is the most common birth defect. It is due to dysfunction of the heart and great vessels during embryo development stage, or the channel was not closed after birth. This study focuses on investigating the influence of virus infection and medicine history during pregnancy on the incidence rate of CHD of fetus. We conducted a retrospective birth cohort study of infant born in the maternal and child health hospital of Fanyu district in Guangzhou. Five thousand three hundred eighty one cases with complete medical records, including mothers, fathers, and infants, were enrolled. The exposure history of mothers to virus and medicine from 6 months before pregnancy to prenatal examination was investigated, including mflu, mumps, measles, rubella, chickenpox, and hepatitis and antibiotics, tocolytic agent, anticonvulsants, antipyretic and analgesic, antitumor drug, folic acid supplement, and contraceptive. The relationship between virus infection and medicine history during pregnancy and CHD was analyzed. There was statistical difference between a normal group and a defected group in influenza infection and tocolytic agent and contraceptive pill. The exposure history to influenza and medicines, such as tocolytic agent and contraceptive pill, during pregnancy influenced the incidence rate of CHD of fetus.
Collapse
Affiliation(s)
- Qianhong Liang
- Department of Ultrasound, Panyu He Xian Memorial Hospital, No. 2 East Qinghe Road, Panyu District, Guangzhou City, Guangdong Province, China.
| | - Wei Gong
- Department of Ultrasound, Panyu He Xian Memorial Hospital, No. 2 East Qinghe Road, Panyu District, Guangzhou City, Guangdong Province, China
| | - Dongming Zheng
- Department of Ultrasound, Panyu He Xian Memorial Hospital, No. 2 East Qinghe Road, Panyu District, Guangzhou City, Guangdong Province, China
| | - Risheng Zhong
- Department of Ultrasound, Panyu He Xian Memorial Hospital, No. 2 East Qinghe Road, Panyu District, Guangzhou City, Guangdong Province, China
| | - Yunjie Wen
- Guangzhou Huayin Medical Laboratory Center. CO. Ltd, Guangzhou, 510663, China
| | - Xiaodan Wang
- Guangzhou Huayin Medical Laboratory Center. CO. Ltd, Guangzhou, 510663, China
| |
Collapse
|
37
|
Muntean I, Togănel R, Benedek T. Genetics of Congenital Heart Disease: Past and Present. Biochem Genet 2016; 55:105-123. [PMID: 27807680 DOI: 10.1007/s10528-016-9780-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022]
Abstract
Congenital heart disease is the most common congenital anomaly, representing an important cause of infant morbidity and mortality. Congenital heart disease represents a group of heart anomalies that include septal defects, valve defects, and outflow tract anomalies. The exact genetic, epigenetic, or environmental basis of congenital heart disease remains poorly understood, although the exact mechanism is likely multifactorial. However, the development of new technologies including copy number variants, single-nucleotide polymorphism, next-generation sequencing are accelerating the detection of genetic causes of heart anomalies. Recent studies suggest a role of small non-coding RNAs, micro RNA, in congenital heart disease. The recently described epigenetic factors have also been found to contribute to cardiac morphogenesis. In this review, we present past and recent genetic discoveries in congenital heart disease.
Collapse
Affiliation(s)
- Iolanda Muntean
- Institute of Cardiovascular Diseases and Transplantation, Clinic of Pediatric Cardiology, University of Medicine and Pharmacy Tîrgu Mureş, 50 Gh Marinescu St, 540136, Tirgu Mures, Romania
| | - Rodica Togănel
- Institute of Cardiovascular Diseases and Transplantation, Clinic of Pediatric Cardiology, University of Medicine and Pharmacy Tîrgu Mureş, 50 Gh Marinescu St, 540136, Tirgu Mures, Romania.
| | - Theodora Benedek
- Clinic of Cardiology, University of Medicine and Pharmacy Tîrgu Mureş, Tirgu Mures, Romania
| |
Collapse
|