1
|
Pradeep SR, Thirunavukkarasu M, Accorsi D, Swaminathan S, Lim ST, Cernuda B, Kemerley A, Hubbard J, Campbell J, Wilson RL, Coca-Soliz V, Tapias L, Selvaraju V, Jellison ER, Yee SP, Palesty JA, Maulik N. Novel approaches to determine the functional role of cardiomyocyte specific E3 ligase, Pellino-1 following myocardial infarction. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166899. [PMID: 37778482 DOI: 10.1016/j.bbadis.2023.166899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVES Ubiquitination plays a vital role in controlling vascular inflammation, cellular protein quality control, and minimizing misfolded protein toxicity. Pellino-1 (Peli1), a type of E3 ubiquitin ligase, has emerged as a critical regulator of the innate immune response; however, its role in the repair and regeneration of ischemic myocardium remains to be elucidated. METHODS Mice (8-12 weeks old, male and females) were divided into (i) Wild type (ii) cardiomyocyte-specific Peli1 overexpressed (AMPEL1Tg/+), (iii) cardiomyocyte-specific Peli1 knockout (CP1KO) and were subjected to sham and left anterior descending artery ligation. The tissues were collected at various time points after surgery for Western blot, and immunohistochemical analyses. Echocardiography is performed 30 days after myocardial infarction. Cardiomyocytes isolated from wild-type, Peli1 overexpressed and knockout mice were used to study the interaction between cardiomyocytes and endothelial cells in vitro under oxidative stress and cells were used for Western blot, flow cytometric analysis, and scratch assay. RESULTS We observed faster wound closure and increased expression of angiogenic factors with MCECs treated with conditioned media obtained from the AMPEL1Tg/+ cardiomyocytes compared to CPIKO and WT cardiomyocytes. Again, AMPEL1Tg/+MI mice showed preserved systolic function and reduced fibrosis compared to the CPIKOMI and WTMI groups. Capillary and arteriolar density were found to be increased in AMPEL1Tg/+MI compared to CP1KOMI. Increased survival and angiogenic factors such as p-Akt, p-MK2, p-IkBα, VEGF, cIAP2, and Bcl2 were observed in AMPEL1Tg/+ compared to CP1KO and WT mice subjected to MI. CONCLUSION The present study uncovers the crucial role of cardiac Peli1 as a regulator of the repair and regeneration of ischemic myocardium by using multiple genetically engineered mouse models.
Collapse
Affiliation(s)
- Seetur R Pradeep
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Mahesh Thirunavukkarasu
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Diego Accorsi
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Santosh Swaminathan
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Sue Ting Lim
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Bryan Cernuda
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Andrew Kemerley
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Jennifer Hubbard
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Jacob Campbell
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Rickesha L Wilson
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Vladimir Coca-Soliz
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Leonidas Tapias
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Vaithinathan Selvaraju
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Evan R Jellison
- Department of Immunology, University of Connecticut Health, School of Medicine, Farmington, CT, USA
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, University of Connecticut Health, School of Medicine, Farmington, CT, USA
| | - J Alexander Palesty
- Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Nilanjana Maulik
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA.
| |
Collapse
|
2
|
Yan L, Cui Y, Feng J. Biology of Pellino1: a potential therapeutic target for inflammation in diseases and cancers. Front Immunol 2023; 14:1292022. [PMID: 38179042 PMCID: PMC10765590 DOI: 10.3389/fimmu.2023.1292022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Pellino1 (Peli1) is a highly conserved E3 Ub ligase that exerts its biological functions by mediating target protein ubiquitination. Extensive evidence has demonstrated the crucial role of Peli1 in regulating inflammation by modulating various receptor signaling pathways, including interleukin-1 receptors, Toll-like receptors, nuclear factor-κB, mitogen-activated protein kinase, and phosphoinositide 3-kinase/AKT pathways. Peli1 has been implicated in the development of several diseases by influencing inflammation, apoptosis, necrosis, pyroptosis, autophagy, DNA damage repair, and glycolysis. Peli1 is a risk factor for most cancers, including breast cancer, lung cancer, and lymphoma. Conversely, Peli1 protects against herpes simplex virus infection, systemic lupus erythematosus, esophageal cancer, and toxic epidermolysis bullosa. Therefore, Peli1 is a potential therapeutic target that warrants further investigation. This comprehensive review summarizes the target proteins of Peli1, delineates their involvement in major signaling pathways and biological processes, explores their role in diseases, and discusses the potential clinical applications of Peli1-targeted therapy, highlighting the therapeutic prospects of Peli1 in various diseases.
Collapse
Affiliation(s)
| | | | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Thirunavukkarasu M, Swaminathan S, Kemerley A, Pradeep SR, Lim ST, Accorsi D, Wilson R, Campbell J, Saad I, Yee SP, Palesty JA, McFadden DW, Maulik N. Role of Pellino-1 in Inflammation and Cardioprotection following Severe Sepsis: A Novel Mechanism in a Murine Severe Sepsis Model †. Cells 2023; 12:1527. [PMID: 37296648 PMCID: PMC10252528 DOI: 10.3390/cells12111527] [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: 01/27/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVES Intra-abdominal sepsis is commonly diagnosed in the surgical population and remains the second most common cause of sepsis overall. Sepsis-related mortality remains a significant burden in the intensive care unit despite advances in critical care. Nearly a quarter of the deaths in people with heart failure are caused by sepsis. We have observed that overexpression of mammalian Pellino-1 (Peli1), an E3 ubiquitin ligase, causes inhibition of apoptosis, oxidative stress, and preservation of cardiac function in a myocardial infarction model. Given these manifold applications, we investigated the role of Peli1 in sepsis using transgenic and knockout mouse models specific to this protein. Therefore, we aimed to explore further the myocardial dysfunction seen in sepsis through its relation to the Peli 1 protein by using the loss of function and gain-of-function strategy. METHODS A series of genetic animals were created to understand the role of Peli1 in sepsis and the preservation of heart function. Wild-type, global Peli1 knock out (Peli1-/-), cardiomyocyte-specific Peli1 deletion (CP1KO), and cardiomyocyte-specific Peli1 overexpressing (alpha MHC (αMHC) Peli1; AMPEL1Tg/+) animals were divided into sham and cecal ligation and puncture (CLP) surgical procedure groups. Cardiac function was determined by two-dimensional echocardiography pre-surgery and at 6- and 24-h post-surgery. Serum IL-6 and TNF-alpha levels (ELISA) (6 h), cardiac apoptosis (TUNEL assay), and Bax expression (24 h) post-surgery were measured. Results are expressed as mean ± S.E.M. RESULTS AMPEL1Tg/+ prevents sepsis-induced cardiac dysfunction assessed by echocardiographic analysis, whereas global and cardiomyocyte-specific deletion of Peli1 shows significant deterioration of cardiac functions. Cardiac function was similar across the sham groups in all three genetically modified mice. ELISA assay displayed how Peli 1 overexpression decreased cardo-suppressive circulating inflammatory cytokines (TNF-alpha, IL-6) compared to both the knockout groups. The proportion of TUNEL-positive cells varied according to Peli1 expression, with overexpression (AMPEL1Tg/+) leading to a significant reduction and Peli1 gene knockout (Peli1-/- and CP1KO) leading to a significant increase in their presence. A similar trend was also observed with Bax protein expression. The improved cellular survival associated with Peli1 overexpression was again shown with the reduction of oxidative stress marker 4-Hydroxy-2-Nonenal (4-HNE). CONCLUSION Our results indicate that overexpression of Peli1 is a novel approach that not only preserved cardiac function but reduced inflammatory markers and apoptosis following severe sepsis in a murine genetic model.
Collapse
Affiliation(s)
- Mahesh Thirunavukkarasu
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Santosh Swaminathan
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Andrew Kemerley
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Seetur R. Pradeep
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Sue Ting Lim
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Diego Accorsi
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Rickesha Wilson
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Jacob Campbell
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Ibnalwalid Saad
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, University of Connecticut Health School of Medicine, Farmington, CT 06032, USA
| | - J. Alexander Palesty
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - David W. McFadden
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Nilanjana Maulik
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| |
Collapse
|
4
|
Koo SY, Park EJ, Noh HJ, Jo SM, Ko BK, Shin HJ, Lee CW. Ubiquitination Links DNA Damage and Repair Signaling to Cancer Metabolism. Int J Mol Sci 2023; 24:ijms24098441. [PMID: 37176148 PMCID: PMC10179089 DOI: 10.3390/ijms24098441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Changes in the DNA damage response (DDR) and cellular metabolism are two important factors that allow cancer cells to proliferate. DDR is a set of events in which DNA damage is recognized, DNA repair factors are recruited to the site of damage, the lesion is repaired, and cellular responses associated with the damage are processed. In cancer, DDR is commonly dysregulated, and the enzymes associated with DDR are prone to changes in ubiquitination. Additionally, cellular metabolism, especially glycolysis, is upregulated in cancer cells, and enzymes in this metabolic pathway are modulated by ubiquitination. The ubiquitin-proteasome system (UPS), particularly E3 ligases, act as a bridge between cellular metabolism and DDR since they regulate the enzymes associated with the two processes. Hence, the E3 ligases with high substrate specificity are considered potential therapeutic targets for treating cancer. A number of small molecule inhibitors designed to target different components of the UPS have been developed, and several have been tested in clinical trials for human use. In this review, we discuss the role of ubiquitination on overall cellular metabolism and DDR and confirm the link between them through the E3 ligases NEDD4, APC/CCDH1, FBXW7, and Pellino1. In addition, we present an overview of the clinically important small molecule inhibitors and implications for their practical use.
Collapse
Affiliation(s)
- Seo-Young Koo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Eun-Ji Park
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Ji Noh
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Su-Mi Jo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Bo-Kyoung Ko
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Jin Shin
- Team of Radiation Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
5
|
Yang J, Tong T, Zhu C, Zhou M, Jiang Y, Chen H, Que L, Liu L, Zhu G, Ha T, Chen Q, Li C, Xu Y, Li J, Li Y. Peli1 contributes to myocardial ischemia/reperfusion injury by impairing autophagy flux via its E3 ligase mediated ubiquitination of P62. J Mol Cell Cardiol 2022; 173:30-46. [PMID: 36179399 DOI: 10.1016/j.yjmcc.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 01/18/2023]
Abstract
Autophagy flux is impaired during myocardial ischemia/reperfusion (M-I/R) via the accumulation of autophagosome and insufficient clearance, which exacerbates cardiomyocyte death. Peli1 (Pellion1) is a RING finger domain-containing ubiquitin E3 ligase that could catalyze the polyubiquitination of substrate proteins. Peli1 has been demonstrated to play an important role in ischemic cardiac diseases. However, little is known about whether Peli1 is involved in the regulation of autophagy flux during M-I/R. The present study investigated whether M-I/R induced impaired autophagy flux could be mediated through Peli1 dependent mechanisms. We induced M-I/R injury in wild type (WT) and Peli1 knockout mice and observed that M-I/R significantly decreased cardiac function that was associated with increased cardiac Peli1 expression and upregulated autophagy-associated protein LC3II and P62. In contrast, Peli1 knockout mice exhibited significant improvement of M-I/R induced cardiac dysfunction and decreased LC3II and P62 expression. Besides, inhibitors of autophagy also increased the infarct size in Peli1 knockout mice after 24 h of reperfusion. Mechanistic studies demonstrated that in vivo I/R or in vitro hypoxia/reoxygenation (H/R) markedly increased the Peli1 E3 ligase activity which directly promoted the ubiquitination of P62 at lysine(K)7 via K63-linkage to inhibit its dimerization and autophagic degradation. Co-immunoprecipitation and GST-pull down assay indicated that Peli1 interacted with P62 via the Ring domain. In addition, Peli1 deficiency also decreased cardiomyocyte apoptosis. Together, our work demonstrated a critical link between increased expression and activity of Peli1 and autophagy flux blockage in M-I/R injury, providing insight into a promising strategy for treating myocardium M-I/R injury.
Collapse
Affiliation(s)
- Jie Yang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tingting Tong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chenghao Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Miao Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yuqing Jiang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hao Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Pathology, Wannan Medical College, Wuhu 241002, Anhui, China
| | - Linli Que
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Li Liu
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Guoqing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tuanzhu Ha
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Qi Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Institute of Biomedical Research, Liaocheng University, Liaocheng 252000, Shandong, China
| | - Jiantao Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
6
|
Protective Effect of Cardiomyocyte-Specific Prolyl-4-Hydroxylase 2 Inhibition on Ischemic Injury in a Mouse MI Model. J Am Coll Surg 2022; 235:240-254. [PMID: 35758926 DOI: 10.1097/xcs.0000000000000241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Our earlier studies showed that inhibiting prolyl-4-hydroxylase enzymes (PHD-1 and PHD-3) improves angiogenesis, heart function, and limb perfusion in mouse models via stabilizing hypoxia-inducible transcription factor-alpha (HIF-1α). The present study explored the effects of the prolyl-4-hydroxylase enzyme, PHD-2, on ischemic heart failure using cardiac-specific PHD-2 gene knockout (KO) mice (PHD2-/-). STUDY DESIGN Adult wild-type (WT) and PHD2-/- mice, 8-12 weeks old, were subjected to myocardial infarction (MI) by irreversibly ligating the left anterior descending (LAD) coronary artery. All sham group mice underwent surgery without LAD ligation. Animals were divided into four groups 1) Wild-type Sham (WTS); Wild-Type myocardial infarction (WTMI); 3) PHD2KO Sham (PHD2-/-S); 4) PHD2KO myocardial infarction (PHD2-/-MI). Left ventricular tissue samples collected at various time points following surgery were used for microRNA expression profiling, Western blotting, immunohistochemical, and echocardiographic analysis. RESULTS Volcano plot analysis revealed 19 differentially expressed miRNAs in the PHD2-/-MI compared to the WTMI group. Target analysis using Ingenuity Pathway Analysis showed several differentially regulated miRNAs targeting key signaling pathways such as Akt, VEGF, Ang-1, PTEN, apoptosis, and hypoxia pathways. Compared to the WTMI group, Western blot analysis showed increased HIF-1α, VEGF, phospho-AKT, and β-catenin expression and reduced Bax expression for the PHD2-/-MI group post-MI. Echocardiographic analysis showed preserved heart functions, and picrosirius red staining revealed decreased fibrosis in PHD2-/-MI compared to the WTMI group. CONCLUSION PHD2 inhibition showed preserved heart function, enhanced angiogenic factor expression, and decreased apoptotic markers after MI. Overall, PHD2 gene inhibition is a promising candidate for managing cardiovascular diseases.
Collapse
|
7
|
Zhang E, Li X. The Emerging Roles of Pellino Family in Pattern Recognition Receptor Signaling. Front Immunol 2022; 13:728794. [PMID: 35197966 PMCID: PMC8860249 DOI: 10.3389/fimmu.2022.728794] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/14/2022] [Indexed: 12/03/2022] Open
Abstract
The Pellino family is a novel and well-conserved E3 ubiquitin ligase family and consists of Pellino1, Pellino2, and Pellino3. Each family member exhibits a highly conserved structure providing ubiquitin ligase activity without abrogating cell and structure-specific function. In this review, we mainly summarized the crucial roles of the Pellino family in pattern recognition receptor-related signaling pathways: IL-1R signaling, Toll-like signaling, NOD-like signaling, T-cell and B-cell signaling, and cell death-related TNFR signaling. We also summarized the current information of the Pellino family in tumorigenesis, microRNAs, and other phenotypes. Finally, we discussed the outstanding questions of the Pellino family in immunity.
Collapse
Affiliation(s)
- E Zhang
- Marine College, Shandong University, Weihai, China
| | - Xia Li
- Marine College, Shandong University, Weihai, China
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
- *Correspondence: Xia Li,
| |
Collapse
|
8
|
Thirunavukkarasu M, Pradeep SR, Ukani G, Abunnaja S, Youssef M, Accorsi D, Swaminathan S, Lim ST, Parker V, Campbell J, Rishi MT, Palesty JA, Maulik N. Gene therapy with Pellino-1 improves perfusion and decreases tissue loss in Flk-1 heterozygous mice but fails in MAPKAP Kinase-2 knockout murine hind limb ischemia model. Microvasc Res 2022; 141:104311. [PMID: 34999110 PMCID: PMC9250804 DOI: 10.1016/j.mvr.2022.104311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 10/19/2022]
Abstract
OBJECTIVES In the United States, over 8.5 million people suffer from peripheral arterial disease (PAD). Previously we reported that Pellino-1(Peli1) gene therapy reduces ischemic damage in the myocardium and skin flaps in Flk-1 [Fetal Liver kinase receptor-1 (Flk-1)/ Vascular endothelial growth factor receptor-2/VEGFR2] heterozygous (Flk-1+/-) mice. The present study compares the angiogenic response and perfusion efficiency following hind limb ischemia (HLI) in, Flk-1+/- and, MAPKAPKINASE2 (MK2-/-) knockout (KO) mice to their control wild type (WT). We also demonstrated the use of Peli1 gene therapy to improve loss of function following HLI. STUDY DESIGN AND METHODS Femoral artery ligation (HLI) was performed in both Flk-1+/-and MK2-/-mice along with their corresponding WT. Another set of Flk-1+/- and MK2-/- were injected with either Adeno-LacZ (Ad.LacZ) or Adeno-Peli1 (Ad.Peli1) after HLI. Hind limb perfusion was assessed by laser doppler imaging at specific time points. A standardized scoring scale is used to quantify the extent of ischemia. Histology analysis performed includes capillary density, fibrosis, pro-angiogenic and anti-apoptotic proteins. RESULTS Flk-1+/- and MK2-/- had a slower recovery of perfusion efficiency in the ischemic limbs than controls. Both Flk-1+/-and MK2-/-KO mice showed decreased capillary density and capillary myocyte ratios with increased fibrosis than their corresponding wild types. Ad.Peli1 injected ischemic Flk-1+/- limb showed improved perfusion, increased capillary density, and pro-angiogenic molecules with reduced fibrosis compared to Ad.LacZ group. No significant improvement in perfusion was observed in MK2-/- ischemic limb after Ad. Peli1 injection. CONCLUSION Deletion of Flk-1 and MK2 impairs neovascularization and perfusion following HLI. Treatment with Ad. Peli1 results in increased angiogenesis and improved perfusion in Flk-1+/- mice but fails to rectify perfusion in MK2 KO mice. Overall, Peli1 gene therapy is a promising candidate for the treatment of PAD.
Collapse
Affiliation(s)
- Mahesh Thirunavukkarasu
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA
| | - Seetur R Pradeep
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA
| | - Gopi Ukani
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Salim Abunnaja
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Mark Youssef
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Diego Accorsi
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Santosh Swaminathan
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Sue Ting Lim
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Virginia Parker
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Jacob Campbell
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA
| | - Muhammad Tipu Rishi
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - J Alexander Palesty
- Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut School of Medicine, University of Connecticut Health, Farmington 06030, CT, USA.
| |
Collapse
|
9
|
Wu D, Kampmann E, Qian G. Novel Insights Into the Role of Mitochondria-Derived Peptides in Myocardial Infarction. Front Physiol 2021; 12:750177. [PMID: 34777013 PMCID: PMC8582487 DOI: 10.3389/fphys.2021.750177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
Mitochondria-derived peptides (MDPs) are a new class of bioactive peptides encoded by small open reading frames (sORFs) within known mitochondrial DNA (mtDNA) genes. MDPs may affect the expression of nuclear genes and play cytoprotective roles against chronic and age-related diseases by maintaining mitochondrial function and cell viability in the face of metabolic stress and cytotoxic insults. In this review, we summarize clinical and experimental findings indicating that MDPs act as local and systemic regulators of glucose homeostasis, immune and inflammatory responses, mitochondrial function, and adaptive stress responses, and focus on evidence supporting the protective effects of MDPs against myocardial infarction. These insights into MDPs actions suggest their potential in the treatment of cardiovascular diseases and should encourage further research in this field.
Collapse
Affiliation(s)
- Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Enny Kampmann
- School of Life Sciences, City College of San Francisco, San Francisco, CA, United States
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
10
|
Cheng D, Zheng J, Hu F, Lv W, Lu C. Abnormal Mitochondria-Endoplasmic Reticulum Communication Promotes Myocardial Infarction. Front Physiol 2021; 12:717187. [PMID: 34413791 PMCID: PMC8369510 DOI: 10.3389/fphys.2021.717187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023] Open
Abstract
Myocardial infarction is characterized by cardiomyocyte death, and can be exacerbated by mitochondrial damage and endoplasmic reticulum injury. In the present study, we investigated whether communication between mitochondria and the endoplasmic reticulum contributes to cardiomyocyte death after myocardial infarction. Our data demonstrated that hypoxia treatment (mimicking myocardial infarction) promoted cardiomyocyte death by inducing the c-Jun N-terminal kinase (JNK) pathway. The activation of JNK under hypoxic conditions was dependent on overproduction of mitochondrial reactive oxygen species (mtROS) in cardiomyocytes, and mitochondrial division was identified as the upstream inducer of mtROS overproduction. Silencing mitochondrial division activators, such as B cell receptor associated protein 31 (BAP31) and mitochondrial fission 1 (Fis1), repressed mitochondrial division, thereby inhibiting mtROS overproduction and preventing JNK-induced cardiomyocyte death under hypoxic conditions. These data revealed that a novel death-inducing mechanism involving the BAP31/Fis1/mtROS/JNK axis promotes hypoxia-induced cardiomyocyte damage. Considering that BAP31 is localized within the endoplasmic reticulum and Fis1 is localized in mitochondria, abnormal mitochondria-endoplasmic reticulum communication may be a useful therapeutic target after myocardial infarction.
Collapse
Affiliation(s)
- Degang Cheng
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Jia Zheng
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Fang Hu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Wei Lv
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
11
|
Melatonin Attenuates Cardiac Ischemia-Reperfusion Injury through Modulation of IP3R-Mediated Mitochondria-ER Contact. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1370862. [PMID: 34422206 PMCID: PMC8371645 DOI: 10.1155/2021/1370862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/17/2021] [Accepted: 07/24/2021] [Indexed: 02/07/2023]
Abstract
Although the interplay between mitochondria and ER has been identified as a crucial regulator of cellular homeostasis, the pathogenic impact of alterations in mitochondria-ER contact sites (MERCS) during myocardial postischemic reperfusion (I/R) injury remains incompletely understood. Therefore, in our study, we explored the beneficial role played by melatonin in protecting cardiomyocytes against reperfusion injury via stabilizing mitochondria-ER interaction. In vitro exposure of H9C2 rat cardiomyocytes to hypoxia/reoxygenation (H/R) augmented mitochondrial ROS synthesis, suppressed both mitochondrial potential and ATP generation, and increased the mitochondrial permeability transition pore (mPTP) opening rate. Furthermore, H/R exposure upregulated the protein content of CHOP and caspase-12, two markers of ER stress, and enhanced the transcription of main MERCS tethering proteins, namely, Fis1, BAP31, Mfn2, and IP3R. Interestingly, all the above changes could be attenuated or reversed by melatonin treatment. Suggesting that melatonin-induced cardioprotection works through normalization of mitochondria-ER interaction, overexpression of IP3R abolished the protective actions offered by melatonin on mitochondria-ER fitness. These results expand our knowledge on the cardioprotective actions of melatonin during myocardial postischemic reperfusion damage and suggest that novel, more effective treatments for acute myocardial reperfusion injury might be achieved through modulation of mitochondria-ER interaction in cardiac cells.
Collapse
|
12
|
Zhang J, Zhang F, Wang Y. Mitofusin-2 Enhances Mitochondrial Contact With the Endoplasmic Reticulum and Promotes Diabetic Cardiomyopathy. Front Physiol 2021; 12:707634. [PMID: 34305656 PMCID: PMC8298037 DOI: 10.3389/fphys.2021.707634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetic cardiomyopathy has been associated with mitochondrial damage. Mitochondria–endoplasmic reticulum (ER) contact is an important determinant of mitochondrial function and ER homeostasis. We therefore investigated whether hyperglycemia can damage the mitochondria by increasing their contact with the ER in cardiomyocytes. We found that hyperglycemia induced mitochondria–ER contact in cardiomyocytes, as evidenced by the increased MMM1, MDM34, and BAP31 expressions. Interestingly, the silencing of Mfn2 reduced the cooperation between the mitochondria and the ER in cardiomyocytes. Mfn2 silencing improved cardiomyocyte viability and function under hyperglycemic conditions. Additionally, the silencing of Mfn2 markedly attenuated the release of calcium from the ER to the mitochondria, thereby preserving mitochondrial metabolism in cardiomyocytes under hyperglycemic conditions. Mfn2 silencing reduced mitochondrial reactive oxygen species production, which reduced mitochondria-dependent apoptosis in hyperglycemia-treated cardiomyocytes. Finally, Mfn2 silencing attenuated ER stress in cardiomyocytes subjected to high-glucose stress. These results demonstrate that Mfn2 promotes mitochondria–ER contact in hyperglycemia-treated cardiomyocytes. The silencing of Mfn2 sustained mitochondrial function, suppressed mitochondrial calcium overload, prevented mitochondrial apoptosis, and reduced ER stress, thereby enhancing cardiomyocyte survival under hyperglycemic conditions.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Tianjin First Central Hospital, Tianjing, China
| | - Feng Zhang
- Department of Cardiology, Tianjin First Central Hospital, Tianjing, China
| | - Yanou Wang
- Health Management Department, Tianjin First Central Hospital, Tianjing, China
| |
Collapse
|
13
|
Heat shock protein A12B gene therapy improves perfusion, promotes neovascularization, and decreases fibrosis in a murine model of hind limb ischemia. Surgery 2021; 170:969-977. [PMID: 34092373 DOI: 10.1016/j.surg.2021.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Heat shock protein A12B expressed in endothelial cells is important and required for angiogenesis to form functional vessels in ischemic tissue. We have previously shown the cardioprotective effects of heat shock protein A12B overexpression in a rat model of diabetic myocardial infarction. In this study, we aim to explore the role of heat shock protein A12B in a surgically-induced murine hind-limb ischemia model. MATERIALS AND METHODS Adult 8- to 12-week-old C57BL/6J mice were divided into 2 groups: treated with Adeno.LacZ (control group) and with Adeno.HSPA12B (experimental group) and, with both groups subjected to right femoral artery ligation. Immediately after surgery, mice in both groups received either Adeno.HSPA12B or Adeno.LacZ (1 × 109 plaque forming units) in both the semimembranosus and gastrocnemius muscles of the right limb. The left limb served as the internal control. Both groups underwent serial laser Doppler imaging preoperatively, and again postoperatively until 28 days. Immunohistochemical analysis was performed 3 and 28 days post-surgery. RESULTS Mice in the Adeno.HSPA12B gene therapy group showed improved motor function and a significantly higher blood perfusion ratio on postoperative days 21 and 28, along with better motor function. Immunohistochemical analysis showed increased expression of vascular endothelial growth factor, thioredoxin-1, heme oxygenase, and hypoxia-inducible factor 1α, along with a decreased expression of A-kinase-anchoring protein 12 and thioredoxin-interacting protein levels. The Adeno.HSPA12B-treated group also showed increased capillary and arteriolar density and an increased capillary-myocyte ratio, along with reduced fibrosis compared to the Adeno.LacZ group. CONCLUSION Our study demonstrates that targeted Adeno.HSPA12B gene delivery into ischemic muscle enhances perfusion and angiogenic protein expression. This molecule shows promise for the management of peripheral vascular disease as a potential target for clinical trials and subsequent drug therapy.
Collapse
|
14
|
Ruan Y, Jin Q, Zeng J, Ren F, Xie Z, Ji K, Wu L, Wu J, Li L. Grape Seed Proanthocyanidin Extract Ameliorates Cardiac Remodelling After Myocardial Infarction Through PI3K/AKT Pathway in Mice. Front Pharmacol 2020; 11:585984. [PMID: 33343353 PMCID: PMC7747856 DOI: 10.3389/fphar.2020.585984] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Myocardial infarction is one of the most serious fatal diseases in the world, which is due to acute occlusion of coronary arteries. Grape seed proanthocyanidin extract (GSPE) is an active compound extracted from grape seeds that has anti-oxidative, anti-inflammatory and anti-tumor pharmacological effects. Natural products are cheap, easy to obtain, widely used and effective. It has been used to treat numerous diseases, such as cancer, brain injury and diabetes complications. However, there are limited studies on its role and associated mechanisms in myocardial infarction in mice. This study showed that GSPE treatment in mice significantly reduced cardiac dysfunction and improved the pathological changes due to MI injury. In vitro, GSPE inhibited the apoptosis of H9C2 cells after hypoxia culture, resulting in the expression of Bax decreased and the expression of Bcl-2 increased. The high expression of p-PI3K and p-AKT was detected in MI model in vivo and in vitro. The use of the specific PI3K/AKT pathway inhibitor LY294002 regressed the cardio-protection of GSPE. Our results showed that GSPE could improve the cardiac dysfunction and remodeling induced by MI and inhibit cardiomyocytes apoptosis in hypoxic conditions through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yongxue Ruan
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qike Jin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Zeng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fangfang Ren
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zuoyi Xie
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kangting Ji
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lianpin Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingguo Wu
- Department of General Internal Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Emergency, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|