1
|
Huang Y, Qiu H, Chen W, Meng Z, Cai Y, Qiao D, Yue X. Identification of TRAF2, CAMK2G, and TIMM17A as biomarkers distinguishing mechanical asphyxia from sudden cardiac death base on 4D-DIA Proteomics: A pilot study. J Pharm Biomed Anal 2025; 258:116730. [PMID: 39921950 DOI: 10.1016/j.jpba.2025.116730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/10/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
In the context of forensic medicine, the differential diagnosis between mechanical asphyxia and sudden cardiac death is very important regarding the establishment of the cause of death. Traditional autopsy findings have generally been very nonspecific; accordingly, highlighting the need for more specific molecular biomarkers. This study employed four-dimensional data-independent acquisition (4D-DIA) proteomics technology, in combination with both animal models and human samples, to conduct a comprehensive protein expression analysis of cardiac tissues, identifying 7557 proteins, among which 142 shared differentially expressed proteins (DEPS) were screened out. Based on the protein interaction network and through rigorous screening, this study identified three proteins, namely TNF receptor-associated factor 2 (TRAF2), Calcium/calmodulin-dependent protein kinase II gamma (CAMK2G), and translocase of inner mitochondrial membrane 17 homolog A (TIMM17A), as biomarkers for differentiating mechanical asphyxia from sudden cardiac death. Further verification using Western Blot (WB) and immunohistochemistry (IHC) proved the differential expression of these biomarkers in both animal and human samples. These findings, besides deepening the molecular understanding of the pathophysiological differences between sudden cardiac death and mechanical asphyxia, also provided new biomarkers for forensic applications that could enable the improvement of accuracy and reliability in the determination of the cause of death.
Collapse
Affiliation(s)
- Yuebing Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hai Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wen Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zilin Meng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu Cai
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Dongfang Qiao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
2
|
Ravindran R, Gustafsson ÅB. Mitochondrial quality control in cardiomyocytes: safeguarding the heart against disease and ageing. Nat Rev Cardiol 2025:10.1038/s41569-025-01142-1. [PMID: 40113864 DOI: 10.1038/s41569-025-01142-1] [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] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
Mitochondria are multifunctional organelles that are important for many different cellular processes, including energy production and biosynthesis of fatty acids, haem and iron-sulfur clusters. Mitochondrial dysfunction leads to a disruption in these processes, the generation of excessive reactive oxygen species, and the activation of inflammatory and cell death pathways. The consequences of mitochondrial dysfunction are particularly harmful in energy-demanding organs such as the heart. Loss of terminally differentiated cardiomyocytes leads to cardiac remodelling and a reduced ability to sustain contraction. Therefore, cardiomyocytes rely on multilayered mitochondrial quality control mechanisms to maintain a healthy population of mitochondria. Mitochondrial chaperones protect against protein misfolding and aggregation, and resident proteases eliminate damaged proteins through proteolysis. Irreparably damaged mitochondria can also be degraded through mitochondrial autophagy (mitophagy) or ejected from cells inside vesicles. The accumulation of dysfunctional mitochondria in cardiomyocytes is a hallmark of ageing and cardiovascular disease. This accumulation is driven by impaired mitochondrial quality control mechanisms and contributes to the development of heart failure. Therefore, there is a strong interest in developing therapies that directly target mitochondrial quality control in cardiomyocytes. In this Review, we discuss the current knowledge of the mechanisms involved in regulating mitochondrial quality in cardiomyocytes, how these pathways are altered with age and in disease, and the therapeutic potential of targeting mitochondrial quality control pathways in cardiovascular disease.
Collapse
Affiliation(s)
- Rishith Ravindran
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Islam M, Rawnsley DR, Ma X, Navid W, Zhao C, Guan X, Foroughi L, Murphy JT, Navid H, Weinheimer CJ, Kovacs A, Nigro J, Diwan A, Chang RP, Kumari M, Young ME, Razani B, Margulies KB, Abdellatif M, Sedej S, Javaheri A, Covey DF, Mani K, Diwan A. Phosphorylation of CRYAB induces a condensatopathy to worsen post-myocardial infarction left ventricular remodeling. J Clin Invest 2025; 135:e163730. [PMID: 39932799 PMCID: PMC11957698 DOI: 10.1172/jci163730] [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: 07/22/2022] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Protein aggregates are emerging therapeutic targets in rare monogenic causes of cardiomyopathy and amyloid heart disease, but their role in more prevalent heart-failure syndromes remains mechanistically unexamined. We observed mislocalization of desmin and sarcomeric proteins to aggregates in human myocardium with ischemic cardiomyopathy and in mouse hearts with post-myocardial infarction ventricular remodeling, mimicking findings of autosomal-dominant cardiomyopathy induced by the R120G mutation in the cognate chaperone protein CRYAB. In both syndromes, we demonstrate increased partitioning of CRYAB phosphorylated on serine 59 to NP40-insoluble aggregate-rich biochemical fraction. While CRYAB undergoes phase separation to form condensates, the phosphomimetic mutation of serine 59 to aspartate (S59D) in CRYAB mimics R120G-CRYAB mutants with reduced condensate fluidity, formation of protein aggregates, and increased cell death. Conversely, changing serine to alanine (phosphorylation-deficient mutation) at position 59 (S59A) restored condensate fluidity and reduced both R120G-CRYAB aggregates and cell death. In mice, S59D CRYAB knockin was sufficient to induce desmin mislocalization and myocardial protein aggregates, while S59A CRYAB knockin rescued left ventricular systolic dysfunction after myocardial infarction and preserved desmin localization with reduced myocardial protein aggregates. 25-Hydroxycholesterol attenuated CRYAB serine 59 phosphorylation and rescued post-myocardial infarction adverse remodeling. Thus, targeting CRYAB phosphorylation-induced condensatopathy is an attractive strategy to counter ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Moydul Islam
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - David R. Rawnsley
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiucui Ma
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Walter Navid
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chen Zhao
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xumin Guan
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Layla Foroughi
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John T. Murphy
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Honora Navid
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carla J. Weinheimer
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Attila Kovacs
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jessica Nigro
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaradhya Diwan
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan P. Chang
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Minu Kumari
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Martin E. Young
- Division of Cardiology and Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Babak Razani
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kenneth B. Margulies
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mahmoud Abdellatif
- Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Simon Sedej
- Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Ali Javaheri
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
| | - Douglas F. Covey
- Department of Developmental Biology and
- Department of Anesthesiology, Psychiatry, and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kartik Mani
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
- Cardiovascular Service Line, HCA Midwest Health, Overland Park, Kansas, USA
| | - Abhinav Diwan
- Division of Cardiology and
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
- Departments of Cell Biology and Physiology, Obstetrics and Gynecology, and Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Shaikh K, Bowman M, McCormick SM, Gao L, Zhang J, White J, Tawil J, Kapoor A, Arav-Boger R, Norbury CC, Harhaj EW. ZFAND6 promotes TRAF2-dependent mitophagy to restrain cGAS-STING signaling. iScience 2025; 28:111544. [PMID: 39811672 PMCID: PMC11731517 DOI: 10.1016/j.isci.2024.111544] [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: 08/02/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
ZFAND6 is a zinc finger protein that interacts with TNF receptor-associated factor 2 (TRAF2) and polyubiquitin chains and has been linked to tumor necrosis factor (TNF) signaling. Here, we report a previously undescribed function of ZFAND6 in maintaining mitochondrial homeostasis by promoting mitophagy. Deletion of ZFAND6 in bone marrow-derived macrophages (BMDMs) upregulates reactive oxygen species (ROS) and the accumulation of damaged mitochondria due to impaired mitophagy. Consequently, mitochondrial DNA (mtDNA) is released into the cytoplasm, triggering the spontaneous expression of interferon-stimulated genes (ISGs) in a stimulator of interferon genes (STING) dependent manner, which leads to enhanced viral resistance. Mechanistically, ZFAND6 bridges a TRAF2-cIAP1 interaction and mediates the recruitment of TRAF2 to damaged mitochondria, which is required for the initiation of ubiquitin-dependent mitophagy. Our results suggest that ZFAND6 promotes the interactions of TRAF2 and cIAP1 and the clearance of damaged mitochondria by mitophagy to maintain mitochondrial homeostasis.
Collapse
Affiliation(s)
- Kashif Shaikh
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Melissa Bowman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Sarah M. McCormick
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Linlin Gao
- Department of Microbiology and Immunology, The University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Jiawen Zhang
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Jesse White
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - John Tawil
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Arun Kapoor
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Ravit Arav-Boger
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Christopher C. Norbury
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Edward W. Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
5
|
Rawnsley DR, Islam M, Zhao C, Kargar Gaz Kooh Y, Mendoza A, Navid H, Kumari M, Guan X, Murphy JT, Nigro J, Kovacs A, Mani K, Huebsch N, Ma X, Diwan A. Mitophagy Facilitates Cytosolic Proteostasis to Preserve Cardiac Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.24.624947. [PMID: 39651239 PMCID: PMC11623534 DOI: 10.1101/2024.11.24.624947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Protein quality control (PQC) is critical for maintaining sarcomere structure and function in cardiac myocytes, and mutations in PQC pathway proteins, such as CRYAB (arginine to glycine at position 120, R120G) and BAG3 (proline to lysine at position 209, P209L) induce protein aggregate pathology with cardiomyopathy in humans. Novel observations in yeast and mammalian cells demonstrate mitochondrial uptake of cytosolic protein aggregates. We hypothesized that mitochondrial uptake of cytosolic protein aggregates and their removal by mitophagy, a lysosomal degradative pathway essential for myocardial homeostasis, facilitates cytosolic protein quality control in cardiac myocytes. Methods Mice with inducible cardiac myocyte specific ablation of TRAF2 (TRAF2icKO), which impairs mitophagy, were assessed for protein aggregates with biochemical fractionation and super-resolution imaging in comparison to floxed controls. Induced pluripotent stem cell (iPSC)-derived cardiac myocytes with R120G knock-in to the CRYAB locus were assessed for localization of the CRYAB protein. Transgenic mice expressing R120G CRYAB protein (R120G-TG) were subjected to both TRAF2 gain-of-function (with AAV9-cardiac Troponin T promoter-driven TRAF2 transduction) and TRAF2 loss-of-function (with tamoxifen-inducible ablation of one Traf2 allele) in cardiac myocytes to determine the effect of mitophagy modulation on cardiac structure, function, and protein aggregate pathology. Results Cardiomyocyte-specific ablation of TRAF2 results accumulation of mitochondrial and cytosolic protein aggregates and DESMIN mis-localization to protein aggregates. Isolated mitochondria take up cardiomyopathy-associated aggregate-prone cytosolic chaperone proteins, namely arginine to glycine (R120G) CRYAB mutant and proline to lysine (P209L) BAG3 mutant. R120G-CRYAB mutant protein increasingly localizes to mitochondria in human and mouse cardiomyocytes. R120G-TG mice demonstrate upregulation of TRAF2 in the mitochondrial fraction with increased mitophagy as compared with wild type. Adult-onset inducible haplo-insufficiency of TRAF2 resulted in accelerated mortality, impaired left ventricular systolic function and increased protein aggregates in R120G-TG mice as compared with controls. Conversely, AAV9-mediated TRAF2 transduction in R120G-TG mice reduced mortality and attenuated left ventricular systolic dysfunction, with reduced protein aggregates and restoration of normal localization of DESMIN, a cytosolic scaffolding protein chaperoned by CRYAB, as compared with control AAV9-GFP group. Conclusions TRAF2-mediated mitophagy in cardiac myocytes facilitates removal of cytosolic protein aggregates and can be stimulated to ameliorate proteotoxic cardiomyopathy.
Collapse
|
6
|
Zhao L, Li Z. LncRNA DANCR suppresses acute myocardial infarction in mice via mediating p-RXRA/TRAF2/NIK/IKK/NF-κB signaling pathway. Aging (Albany NY) 2024; 16:13356-13370. [PMID: 39546553 PMCID: PMC11719107 DOI: 10.18632/aging.206150] [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: 05/16/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES This study aimed to investigate the role of LncRNA differentiation antagonizing non-protein coding RNA (DANCR) in acute myocardial infarction (AMI). METHODS A mouse model of AMI was established, and the cardiac contractile function was detected. Next, cardiomyocytes treated with oxygen-glucose deprivation (OGD) were used for gain- and loss-function experiments in vitro. RIP analysis was used to verify the binding of DANCR and Retinoid X receptor alpha (RXRA), and Co-IP assay was used to measure the binding of phosphorylated RXRA to TNF receptor associated factor 2 (TRAF2). RESULTS The expression of DANCR in myocardial tissues of AMI mice were significantly downregulated. Overexpression of DANCR decreased myocardial infarct size and enhanced cardiac contractile function in AMI mice. Moreover, overexpression of DANCR promoted proliferation and inhibited apoptosis in OGD-induced cardiomyocytes. Mechanism studies demonstrated that DANCR interacted with RXRA and promoted glycogen synthase kinase 3beta (GSK3β)-mediated phosphorylation of RXRA, and phosphorylated RXRA interacted with TRAF2 protein to downregulate TRAF2 protein level. Bexarotene (Bex), an activator of RXRA, inhibited TRAF2 protein expression, while RXRA overexpression had no effect on TRAF2 protein expression. Bex treatment or silencing TRAF2 promoted proliferation and inhibited apoptosis in cardiomyocytes. In addition, silencing DANCR inhibited cardiomyocyte proliferation and induced apoptosis by activating the NIK/IKK/NF-κB pathway, while B022, an inhibitor of NIK, counteracted the effects of DANCR silencing on cardiomyocytes. CONCLUSIONS Studies demonstrated that DANCR suppressed AMI in mice by mediating p-RXRA/TRAF2/NIK/IKK/NF-κB pathway.
Collapse
Affiliation(s)
- Li Zhao
- Department of Cardiovascular, Affiliated Hospital of Yanan University, Yanan, China
| | - Zhi Li
- Department of Cardiovascular, Affiliated Hospital of Yanan University, Yanan, China
| |
Collapse
|
7
|
Zhang F, Zhang L, Hu G, Chen X, Liu H, Li C, Guo X, Huang C, Sun F, Li T, Cui Z, Guo Y, Yan W, Xia Y, Liu Z, Lin Z, Duan W, Lu L, Wang X, Wang Z, Wang S, Tao L. Rectifying METTL4-Mediated N 6-Methyladenine Excess in Mitochondrial DNA Alleviates Heart Failure. Circulation 2024; 150:1441-1458. [PMID: 38686562 DOI: 10.1161/circulationaha.123.068358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/08/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Myocardial mitochondrial dysfunction underpins the pathogenesis of heart failure (HF), yet therapeutic options to restore myocardial mitochondrial function are scarce. Epigenetic modifications of mitochondrial DNA (mtDNA), such as methylation, play a pivotal role in modulating mitochondrial homeostasis. However, their involvement in HF remains unclear. METHODS Experimental HF models were established through continuous angiotensin II and phenylephrine (AngII/PE) infusion or prolonged myocardial ischemia/reperfusion injury. The landscape of N6-methyladenine (6mA) methylation within failing cardiomyocyte mtDNA was characterized using high-resolution mass spectrometry and methylated DNA immunoprecipitation sequencing. A tamoxifen-inducible cardiomyocyte-specific Mettl4 knockout mouse model and adeno-associated virus vectors designed for cardiomyocyte-targeted manipulation of METTL4 (methyltransferase-like protein 4) expression were used to ascertain the role of mtDNA 6mA and its methyltransferase METTL4 in HF. RESULTS METTL4 was predominantly localized within adult cardiomyocyte mitochondria. 6mA modifications were significantly more abundant in mtDNA than in nuclear DNA. Postnatal cardiomyocyte maturation presented with a reduction in 6mA levels within mtDNA, coinciding with a decrease in METTL4 expression. However, an increase in both mtDNA 6mA level and METTL4 expression was observed in failing adult cardiomyocytes, suggesting a shift toward a neonatal-like state. METTL4 preferentially targeted mtDNA promoter regions, which resulted in interference with transcription initiation complex assembly, mtDNA transcriptional stalling, and ultimately mitochondrial dysfunction. Amplifying cardiomyocyte mtDNA 6mA through METTL4 overexpression led to spontaneous mitochondrial dysfunction and HF phenotypes. The transcription factor p53 was identified as a direct regulator of METTL4 transcription in response to HF-provoking stress, thereby revealing a stress-responsive mechanism that controls METTL4 expression and mtDNA 6mA. Cardiomyocyte-specific deletion of the Mettl4 gene eliminated mtDNA 6mA excess, preserved mitochondrial function, and mitigated the development of HF upon continuous infusion of AngII/PE. In addition, specific silencing of METTL4 in cardiomyocytes restored mitochondrial function and offered therapeutic relief in mice with preexisting HF, irrespective of whether the condition was induced by AngII/PE infusion or myocardial ischemia/reperfusion injury. CONCLUSIONS Our findings identify a pivotal role of cardiomyocyte mtDNA 6mA and the corresponding methyltransferase, METTL4, in the pathogenesis of mitochondrial dysfunction and HF. Targeted suppression of METTL4 to rectify mtDNA 6mA excess emerges as a promising strategy for developing mitochondria-focused HF interventions.
Collapse
Affiliation(s)
- Fuyang Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guangyu Hu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiyao Chen
- Geriatrics (X.C.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiong Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chong Huang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fangfang Sun
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tongzheng Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Cui
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongzhen Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjun Yan
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunlong Xia
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyuan Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Lin
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weixun Duan
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Linhe Lu
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyi Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhengyang Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shan Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Tao
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Islam M, Rawnsley DR, Ma X, Navid W, Zhao C, Foroughi L, Murphy JT, Navid H, Weinheimer CJ, Kovacs A, Nigro J, Diwan A, Chang R, Kumari M, Young ME, Razani B, Margulies KB, Abdellatif M, Sedej S, Javaheri A, Covey DF, Mani K, Diwan A. Phosphorylation of CRYAB Induces a Condensatopathy to Worsen Post-Myocardial Infarction Left Ventricular Remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610556. [PMID: 39282298 PMCID: PMC11398338 DOI: 10.1101/2024.08.30.610556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
Protein aggregates are emerging therapeutic targets in rare monogenic causes of cardiomyopathy and amyloid heart disease, but their role in more prevalent heart failure syndromes remains mechanistically unexamined. We observed mis-localization of desmin and sarcomeric proteins to aggregates in human myocardium with ischemic cardiomyopathy and in mouse hearts with post-myocardial infarction ventricular remodeling, mimicking findings of autosomal-dominant cardiomyopathy induced by R120G mutation in the cognate chaperone protein, CRYAB. In both syndromes, we demonstrate increased partitioning of CRYAB phosphorylated on serine-59 to NP40-insoluble aggregate-rich biochemical fraction. While CRYAB undergoes phase separation to form condensates, the phospho-mimetic mutation of serine-59 to aspartate (S59D) in CRYAB mimics R120G-CRYAB mutants with reduced condensate fluidity, formation of protein aggregates and increased cell death. Conversely, changing serine to alanine (phosphorylation-deficient mutation) at position 59 (S59A) restored condensate fluidity, and reduced both R120G-CRYAB aggregates and cell death. In mice, S59D CRYAB knock-in was sufficient to induce desmin mis-localization and myocardial protein aggregates, while S59A CRYAB knock-in rescued left ventricular systolic dysfunction post-myocardial infarction and preserved desmin localization with reduced myocardial protein aggregates. 25-Hydroxycholesterol attenuated CRYAB serine-59 phosphorylation and rescued post-myocardial infarction adverse remodeling. Thus, targeting CRYAB phosphorylation-induced condensatopathy is an attractive strategy to counter ischemic cardiomyopathy.
Collapse
|
9
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
10
|
Wang W, Li E, Zou J, Qu C, Ayala J, Wen Y, Islam MS, Weintraub NL, Fulton DJ, Liang Q, Zhou J, Liu J, Li J, Sun Y, Su H. Ubiquitin Ligase RBX2/SAG Regulates Mitochondrial Ubiquitination and Mitophagy. Circ Res 2024; 135:e39-e56. [PMID: 38873758 PMCID: PMC11264309 DOI: 10.1161/circresaha.124.324285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Clearance of damaged mitochondria via mitophagy is crucial for cellular homeostasis. Apart from Parkin, little is known about additional Ub (ubiquitin) ligases that mediate mitochondrial ubiquitination and turnover, particularly in highly metabolically active organs such as the heart. METHODS In this study, we have combined in silico analysis and biochemical assay to identify CRL (cullin-RING ligase) 5 as a mitochondrial Ub ligase. We generated cardiomyocytes and mice lacking RBX2 (RING-box protein 2; also known as SAG [sensitive to apoptosis gene]), a catalytic subunit of CRL5, to understand the effects of RBX2 depletion on mitochondrial ubiquitination, mitophagy, and cardiac function. We also performed proteomics analysis and RNA-sequencing analysis to define the impact of loss of RBX2 on the proteome and transcriptome. RESULTS RBX2 and CUL (cullin) 5, 2 core components of CRL5, localize to mitochondria. Depletion of RBX2 inhibited mitochondrial ubiquitination and turnover, impaired mitochondrial membrane potential and respiration, increased cardiomyocyte cell death, and has a global impact on the mitochondrial proteome. In vivo, deletion of the Rbx2 gene in adult mouse hearts suppressed mitophagic activity, provoked accumulation of damaged mitochondria in the myocardium, and disrupted myocardial metabolism, leading to the rapid development of dilated cardiomyopathy and heart failure. Similarly, ablation of RBX2 in the developing heart resulted in dilated cardiomyopathy and heart failure. The action of RBX2 in mitochondria is not dependent on Parkin, and Parkin gene deletion had no impact on the onset and progression of cardiomyopathy in RBX2-deficient hearts. Furthermore, RBX2 controls the stability of PINK1 (PTEN-induced kinase 1) in mitochondria. CONCLUSIONS These findings identify RBX2-CRL5 as a mitochondrial Ub ligase that regulates mitophagy and cardiac homeostasis in a Parkin-independent, PINK1-dependent manner.
Collapse
Affiliation(s)
- Wenjuan Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510510, China
| | - Ermin Li
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Jianqiu Zou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Chen Qu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Juan Ayala
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Yuan Wen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Md Sadikul Islam
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - David J. Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Qiangrong Liang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York 11568, United States
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510510, China
| | - Jie Li
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| | - Yi Sun
- Cancer Institute of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, United States
| |
Collapse
|
11
|
Yu H, Gan D, Luo Z, Yang Q, An D, Zhang H, Hu Y, Ma Z, Zeng Q, Xu D, Ren H. α-Ketoglutarate improves cardiac insufficiency through NAD +-SIRT1 signaling-mediated mitophagy and ferroptosis in pressure overload-induced mice. Mol Med 2024; 30:15. [PMID: 38254035 PMCID: PMC10804789 DOI: 10.1186/s10020-024-00783-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND In heart failure (HF), mitochondrial dysfunction and metabolic remodeling lead to a reduction in energy productivity and aggravate cardiomyocyte injury. Supplementation with α-ketoglutarate (AKG) alleviated myocardial hypertrophy and fibrosis in mice with HF and improved cardiac insufficiency. However, the myocardial protective mechanism of AKG remains unclear. We verified the hypothesis that AKG improves mitochondrial function by upregulating NAD+ levels and activating silent information regulator 2 homolog 1 (SIRT1) in cardiomyocytes. METHODS In vivo, 2% AKG was added to the drinking water of mice undergoing transverse aortic constriction (TAC) surgery. Echocardiography and biopsy were performed to evaluate cardiac function and pathological changes. Myocardial metabolomics was analyzed by liquid chromatography‒mass spectrometry (LC‒MS/MS) at 8 weeks after surgery. In vitro, the expression of SIRT1 or PINK1 proteins was inhibited by selective inhibitors and siRNA in cardiomyocytes stimulated with angiotensin II (AngII) and AKG. NAD+ levels were detected using an NAD test kit. Mitophagy and ferroptosis levels were evaluated by Western blotting, qPCR, JC-1 staining and lipid peroxidation analysis. RESULTS AKG supplementation after TAC surgery could alleviate myocardial hypertrophy and fibrosis and improve cardiac function in mice. Metabolites of the malate-aspartate shuttle (MAS) were increased, but the TCA cycle and fatty acid metabolism pathway could be inhibited in the myocardium of TAC mice after AKG supplementation. Decreased NAD+ levels and SIRT1 protein expression were observed in heart of mice and AngII-treated cardiomyocytes. After AKG treatment, these changes were reversed, and increased mitophagy, inhibited ferroptosis, and alleviated damage in cardiomyocytes were observed. When the expression of SIRT1 was inhibited by a selective inhibitor and siRNA, the protective effect of AKG was suppressed. CONCLUSION Supplementation with AKG can improve myocardial hypertrophy, fibrosis and chronic cardiac insufficiency caused by pressure overload. By increasing the level of NAD+, the SIRT-PINK1 and SIRT1-GPX4 signaling pathways are activated to promote mitophagy and inhibit ferroptosis in cardiomyocytes, which ultimately alleviates cardiomyocyte damage.
Collapse
Affiliation(s)
- Hao Yu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Daojing Gan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qilin Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dongqi An
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
12
|
Pedersen LN, Valenzuela Ripoll C, Ozcan M, Guo Z, Lotfinaghsh A, Zhang S, Ng S, Weinheimer C, Nigro J, Kovacs A, Diab A, Klaas A, Grogan F, Cho Y, Ataran A, Luehmann H, Heck A, Kolb K, Strong L, Navara R, Walls GM, Hugo G, Samson P, Cooper D, Reynoso FJ, Schwarz JK, Moore K, Lavine K, Rentschler SL, Liu Y, Woodard PK, Robinson C, Cuculich PS, Bergom C, Javaheri A. Cardiac radiation improves ventricular function in mice and humans with cardiomyopathy. MED 2023; 4:928-943.e5. [PMID: 38029754 PMCID: PMC10994563 DOI: 10.1016/j.medj.2023.10.006] [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: 02/28/2023] [Revised: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Rapidly dividing cells are more sensitive to radiation therapy (RT) than quiescent cells. In the failing myocardium, macrophages and fibroblasts mediate collateral tissue injury, leading to progressive myocardial remodeling, fibrosis, and pump failure. Because these cells divide more rapidly than cardiomyocytes, we hypothesized that macrophages and fibroblasts would be more susceptible to lower doses of radiation and that cardiac radiation could therefore attenuate myocardial remodeling. METHODS In three independent murine heart failure models, including models of metabolic stress, ischemia, and pressure overload, mice underwent 5 Gy cardiac radiation or sham treatment followed by echocardiography. Immunofluorescence, flow cytometry, and non-invasive PET imaging were employed to evaluate cardiac macrophages and fibroblasts. Serial cardiac magnetic resonance imaging (cMRI) from patients with cardiomyopathy treated with 25 Gy cardiac RT for ventricular tachycardia (VT) was evaluated to determine changes in cardiac function. FINDINGS In murine heart failure models, cardiac radiation significantly increased LV ejection fraction and reduced end-diastolic volume vs. sham. Radiation resulted in reduced mRNA abundance of B-type natriuretic peptide and fibrotic genes, and histological assessment of the LV showed reduced fibrosis. PET and flow cytometry demonstrated reductions in pro-inflammatory macrophages, and immunofluorescence demonstrated reduced proliferation of macrophages and fibroblasts with RT. In patients who were treated with RT for VT, cMRI demonstrated decreases in LV end-diastolic volume and improvements in LV ejection fraction early after treatment. CONCLUSIONS These results suggest that 5 Gy cardiac radiation attenuates cardiac remodeling in mice and humans with heart failure. FUNDING NIH, ASTRO, AHA, Longer Life Foundation.
Collapse
Affiliation(s)
- Lauren N Pedersen
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | | | - Mualla Ozcan
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Zhen Guo
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Aynaz Lotfinaghsh
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Shiyang Zhang
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sherwin Ng
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Carla Weinheimer
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jessica Nigro
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ahmed Diab
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Amanda Klaas
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Felicia Grogan
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yoonje Cho
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Anahita Ataran
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Hannah Luehmann
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Abigail Heck
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kollin Kolb
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Lori Strong
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Rachita Navara
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Gerard M Walls
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT97AE, Northern Ireland
| | - Geoff Hugo
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Pamela Samson
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Cooper
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Francisco J Reynoso
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kaitlin Moore
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kory Lavine
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Stacey L Rentschler
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yongjian Liu
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Pamela K Woodard
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Clifford Robinson
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Phillip S Cuculich
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Carmen Bergom
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Ali Javaheri
- Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; John J. Cochran Veterans Affairs Medical Center, St. Louis, MO 63106, USA.
| |
Collapse
|
13
|
Liu X, Zhu Y, Wang D, Feng R, Chen Z, Zheng Z, Li Y, Xu L, Zheng H, Fan Y, Yin Y, Xiao S. The natural compound Sanggenon C inhibits PRRSV infection by regulating the TRAF2/NF-κB signalling pathway. Vet Res 2023; 54:114. [PMID: 38037100 PMCID: PMC10691163 DOI: 10.1186/s13567-023-01245-y] [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: 08/22/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a serious infectious disease and one of the major causes of death in the global pig industry. PRRS virus (PRRSV) strains have complex and diverse genetic characteristics and cross-protection between strains is low, which complicates vaccine selection; thus, the current vaccination strategy has been greatly compromised. Therefore, it is necessary to identify effective natural compounds for the clinical treatment of PRRS. A small molecule library composed of 720 natural compounds was screened in vitro, and we found that Sanggenon C (SC) was amongst the most effective natural compound inhibitors of PRRSV infection. Compared with ribavirin, SC more significantly inhibited PRRSV infection at both the gene and protein levels and reduced the viral titres and levels of protein expression and inflammatory cytokine secretion to more effectively protect cells from PRRSV infection and damage. Mechanistically, SC inhibits activation of the NF-κB signalling pathway by promoting TRAF2 expression, thereby reducing PRRSV replication. In conclusion, by screening natural compounds, we found that SC suppresses PRRSV infection by regulating the TRAF2/NF-κB signalling pathway. This study contributes to a deeper understanding of the therapeutic targets and pathogenesis of PRRSV infection. More importantly, our results demonstrate that SC has potential as a candidate for the treatment of PRRS.
Collapse
Affiliation(s)
- Xiao Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yanan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Dan Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ran Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhihao Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zifang Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Lele Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yunpeng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Yupeng Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Shuqi Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
14
|
Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol 2023; 118:42. [PMID: 37798455 PMCID: PMC10556134 DOI: 10.1007/s00395-023-01009-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.
Collapse
Affiliation(s)
- Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|
15
|
Rocca C, Soda T, De Francesco EM, Fiorillo M, Moccia F, Viglietto G, Angelone T, Amodio N. Mitochondrial dysfunction at the crossroad of cardiovascular diseases and cancer. J Transl Med 2023; 21:635. [PMID: 37726810 PMCID: PMC10507834 DOI: 10.1186/s12967-023-04498-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
A large body of evidence indicates the existence of a complex pathophysiological relationship between cardiovascular diseases and cancer. Mitochondria are crucial organelles whose optimal activity is determined by quality control systems, which regulate critical cellular events, ranging from intermediary metabolism and calcium signaling to mitochondrial dynamics, cell death and mitophagy. Emerging data indicate that impaired mitochondrial quality control drives myocardial dysfunction occurring in several heart diseases, including cardiac hypertrophy, myocardial infarction, ischaemia/reperfusion damage and metabolic cardiomyopathies. On the other hand, diverse human cancers also dysregulate mitochondrial quality control to promote their initiation and progression, suggesting that modulating mitochondrial homeostasis may represent a promising therapeutic strategy both in cardiology and oncology. In this review, first we briefly introduce the physiological mechanisms underlying the mitochondrial quality control system, and then summarize the current understanding about the impact of dysregulated mitochondrial functions in cardiovascular diseases and cancer. We also discuss key mitochondrial mechanisms underlying the increased risk of cardiovascular complications secondary to the main current anticancer strategies, highlighting the potential of strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction and tumorigenesis. It is hoped that this summary can provide novel insights into precision medicine approaches to reduce cardiovascular and cancer morbidities and mortalities.
Collapse
Affiliation(s)
- Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Teresa Soda
- Department of Health Science, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
- National Institute of Cardiovascular Research (I.N.R.C.), 40126, Bologna, Italy.
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy.
| |
Collapse
|
16
|
Diwan A. Preserving mitochondria to treat hypertrophic cardiomyopathy: From rare mitochondrial DNA mutation to heart failure therapy? J Clin Invest 2023; 133:e171965. [PMID: 37463442 PMCID: PMC10348762 DOI: 10.1172/jci171965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Hypertrophic cardiomyopathy and pathological cardiac hypertrophy are characterized by mitochondrial structural and functional abnormalities. In this issue of the JCI, Zhuang et al. discovered 1-deoxynojirimycin (DNJ) through a screen of mitochondrially targeted compounds. The authors described the effects of DNJ in restoring mitochondria and preventing cardiac myocyte hypertrophy in cellular models carrying a mutant mitochondrial gene, MT-RNR2, which is causally implicated in familial hypertrophic cardiomyopathy. DNJ worked via stabilization of the mitochondrial inner-membrane GTPase OPA1 and other, hitherto unknown, mechanisms to preserve mitochondrial crista and respiratory chain components. The discovery is likely to spur development of a class of therapeutics that restore mitochondrial health to prevent cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Abhinav Diwan
- Department of Medicine
- Department of Cell Biology and Physiology
- Department of Obstetrics and Gynecology
- Department of Neurology
- Center for Cardiovascular Research, and
- Hope Center for Neurologic Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
17
|
Marshall AG, Neikirk K, Vue Z, Beasley HK, Garza-Lopez E, Vang L, Barongan T, Evans Z, Crabtree A, Spencer E, Anudokem J, Parker R, Davis J, Stephens D, Damo S, Pham TT, Gomez JA, Exil V, Dai DF, Murray SA, Entman ML, Taffet GE, Hinton AO, Reddy AK. Cardiovascular hemodynamics in mice with tumor necrosis factor receptor-associated factor 2 mediated cytoprotection in the heart. Front Cardiovasc Med 2023; 10:1064640. [PMID: 37229235 PMCID: PMC10203617 DOI: 10.3389/fcvm.2023.1064640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/03/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Many studies in mice have demonstrated that cardiac-specific innate immune signaling pathways can be reprogrammed to modulate inflammation in response to myocardial injury and improve outcomes. While the echocardiography standard parameters of left ventricular (LV) ejection fraction, fractional shortening, end-diastolic diameter, and others are used to assess cardiac function, their dependency on loading conditions somewhat limits their utility in completely reflecting the contractile function and global cardiovascular efficiency of the heart. A true measure of global cardiovascular efficiency should include the interaction between the ventricle and the aorta (ventricular-vascular coupling, VVC) as well as measures of aortic impedance and pulse wave velocity. Methods We measured cardiac Doppler velocities, blood pressures, along with VVC, aortic impedance, and pulse wave velocity to evaluate global cardiac function in a mouse model of cardiac-restricted low levels of TRAF2 overexpression that conferred cytoprotection in the heart. Results While previous studies reported that response to myocardial infarction and reperfusion was improved in the TRAF2 overexpressed mice, we found that TRAF2 mice had significantly lower cardiac systolic velocities and accelerations, diastolic atrial velocity, aortic pressures, rate-pressure product, LV contractility and relaxation, and stroke work when compared to littermate control mice. Also, we found significantly longer aortic ejection time, isovolumic contraction and relaxation times, and significantly higher mitral early/atrial ratio, myocardial performance index, and ventricular vascular coupling in the TRAF2 overexpression mice compared to their littermate controls. We found no significant differences in the aortic impedance and pulse wave velocity. Discussion While the reported tolerance to ischemic insults in TRAF2 overexpression mice may suggest enhanced cardiac reserve, our results indicate diminished cardiac function in these mice.
Collapse
Affiliation(s)
- Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Taylor Barongan
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Zoe Evans
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Elsie Spencer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Josephs Anudokem
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, United States
| | - Remi Parker
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, United States
| | - Jamaine Davis
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, United States
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, United States
| | - Steven Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, United States
| | - Thuy T. Pham
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Jose A. Gomez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Vernat Exil
- Department of Pediatrics, Div. of Cardiology, St. Louis University School of Medicine, St. Louis, MO, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Dao-fu Dai
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sandra A. Murray
- Department of Cell Biology, College of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Mark L. Entman
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - George E. Taffet
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Antentor O. Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Anilkumar K. Reddy
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| |
Collapse
|
18
|
Sakr N, Glazova O, Shevkova L, Onyanov N, Kaziakhmedova S, Shilova A, Vorontsova MV, Volchkov P. Characterizing and Quenching Autofluorescence in Fixed Mouse Adrenal Cortex Tissue. Int J Mol Sci 2023; 24:3432. [PMID: 36834842 PMCID: PMC9968082 DOI: 10.3390/ijms24043432] [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: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Tissue autofluorescence of fixed tissue sections is a major concern of fluorescence microscopy. The adrenal cortex emits intense intrinsic fluorescence that interferes with signals from fluorescent labels, resulting in poor-quality images and complicating data analysis. We used confocal scanning laser microscopy imaging and lambda scanning to characterize the mouse adrenal cortex autofluorescence. We evaluated the efficacy of tissue treatment methods in reducing the intensity of the observed autofluorescence, such as trypan blue, copper sulfate, ammonia/ethanol, Sudan Black B, TrueVIEWTM Autofluorescence Quenching Kit, MaxBlockTM Autofluorescence Reducing Reagent Kit, and TrueBlackTM Lipofuscin Autofluorescence Quencher. Quantitative analysis demonstrated autofluorescence reduction by 12-95%, depending on the tissue treatment method and excitation wavelength. TrueBlackTM Lipofuscin Autofluorescence Quencher and MaxBlockTM Autofluorescence Reducing Reagent Kit were the most effective treatments, reducing the autofluorescence intensity by 89-93% and 90-95%, respectively. The treatment with TrueBlackTM Lipofuscin Autofluorescence Quencher preserved the specific fluorescence signals and tissue integrity, allowing reliable detection of fluorescent labels in the adrenal cortex tissue. This study demonstrates a feasible, easy-to-perform, and cost-effective method to quench tissue autofluorescence and improve the signal-to-noise ratio in adrenal tissue sections for fluorescence microscopy.
Collapse
Affiliation(s)
- Nawar Sakr
- Endocrinology Research Centre, Moscow 117292, Russia
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Olga Glazova
- Endocrinology Research Centre, Moscow 117292, Russia
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Liudmila Shevkova
- Endocrinology Research Centre, Moscow 117292, Russia
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Nikita Onyanov
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Samira Kaziakhmedova
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Alena Shilova
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 27-1, Lomonosovsky Prospect, Moscow 117192, Russia
| | - Maria V. Vorontsova
- Endocrinology Research Centre, Moscow 117292, Russia
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Pavel Volchkov
- Endocrinology Research Centre, Moscow 117292, Russia
- Genome Engineering Lab, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| |
Collapse
|
19
|
Dhingra R, Rabinovich-Nikitin I, Rothman S, Guberman M, Gang H, Margulets V, Jassal DS, Alagarsamy KN, Dhingra S, Ripoll CV, Billia F, Diwan A, Javaheri A, Kirshenbaum LA. Proteasomal Degradation of TRAF2 Mediates Mitochondrial Dysfunction in Doxorubicin-Cardiomyopathy. Circulation 2022; 146:934-954. [PMID: 35983756 PMCID: PMC10043946 DOI: 10.1161/circulationaha.121.058411] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Cytokines such as tumor necrosis factor-α (TNFα) have been implicated in cardiac dysfunction and toxicity associated with doxorubicin (DOX). Although TNFα can elicit different cellular responses, including survival or death, the mechanisms underlying these divergent outcomes in the heart remain cryptic. The E3 ubiquitin ligase TRAF2 (TNF receptor associated factor 2) provides a critical signaling platform for K63-linked polyubiquitination of RIPK1 (receptor interacting protein 1), crucial for nuclear factor-κB (NF-κB) activation by TNFα and survival. Here, we investigate alterations in TNFα-TRAF2-NF-κB signaling in the pathogenesis of DOX cardiotoxicity. METHODS Using a combination of in vivo (4 weekly injections of DOX 5 mg·kg-1·wk-1) in C57/BL6J mice and in vitro approaches (rat, mouse, and human inducible pluripotent stem cell-derived cardiac myocytes), we monitored TNFα levels, lactate dehydrogenase, cardiac ultrastructure and function, mitochondrial bioenergetics, and cardiac cell viability. RESULTS In contrast to vehicle-treated mice, ultrastructural defects, including cytoplasmic swelling, mitochondrial perturbations, and elevated TNFα levels, were observed in the hearts of mice treated with DOX. While investigating the involvement of TNFα in DOX cardiotoxicity, we discovered that NF-κB was readily activated by TNFα. However, TNFα-mediated NF-κB activation was impaired in cardiac myocytes treated with DOX. This coincided with loss of K63- linked polyubiquitination of RIPK1 from the proteasomal degradation of TRAF2. Furthermore, TRAF2 protein abundance was markedly reduced in hearts of patients with cancer treated with DOX. We further established that the reciprocal actions of the ubiquitinating and deubiquitinating enzymes cellular inhibitors of apoptosis 1 and USP19 (ubiquitin-specific peptidase 19), respectively, regulated the proteasomal degradation of TRAF2 in DOX-treated cardiac myocytes. An E3-ligase mutant of cellular inhibitors of apoptosis 1 (H588A) or gain of function of USP19 prevented proteasomal degradation of TRAF2 and DOX-induced cell death. Furthermore, wild-type TRAF2, but not a RING finger mutant defective for K63-linked polyubiquitination of RIPK1, restored NF-κB signaling and suppressed DOX-induced cardiac cell death. Last, cardiomyocyte-restricted expression of TRAF2 (cardiac troponin T-adeno-associated virus 9-TRAF2) in vivo protected against mitochondrial defects and cardiac dysfunction induced by DOX. CONCLUSIONS Our findings reveal a novel signaling axis that functionally connects the cardiotoxic effects of DOX to proteasomal degradation of TRAF2. Disruption of the critical TRAF2 survival pathway by DOX sensitizes cardiac myocytes to TNFα-mediated necrotic cell death and DOX cardiotoxicity.
Collapse
Affiliation(s)
- Rimpy Dhingra
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Sonny Rothman
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Matthew Guberman
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Hongying Gang
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Victoria Margulets
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Davinder S. Jassal
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Keshav N. Alagarsamy
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Regenerative Medicine Program, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Sanjiv Dhingra
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Regenerative Medicine Program, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Carla Valenzuela Ripoll
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Filio Billia
- Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada, Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada
| | - Abhinav Diwan
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ali Javaheri
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Lorrie A. Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Department of Pharmacology and Therapeutics, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| |
Collapse
|