1
|
Varone E, Retini M, Cherubini A, Chernorudskiy A, Marrazza A, Guidarelli A, Cagnotto A, Beeg M, Gobbi M, Fumagalli S, Bolis M, Guarrera L, Barbera MC, Grasselli C, Bleve A, Generali D, Milani M, Mari M, Salmona M, Piersanti G, Bottegoni G, Broggini M, Janssen-Heininger YMW, Cho J, Cantoni O, Zito E. Small molecule-mediated inhibition of the oxidoreductase ERO1A restrains aggressive breast cancer by impairing VEGF and PD-L1 in the tumor microenvironment. Cell Death Dis 2025; 16:105. [PMID: 39962052 PMCID: PMC11833095 DOI: 10.1038/s41419-025-07426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Cancer cells adapt to harsh environmental conditions by inducing the Unfolded Protein Response (UPR), of which ERO1A is a mediator. ERO1A aids protein folding by acting as a protein disulfide oxidase, and under cancer-related hypoxia conditions, it favors the folding of angiogenic VEGFA, leading tumor cells to thrive and spread. The upregulation of ERO1A in cancer cells, oppositely to the dispensability of ERO1A activity in healthy cells, renders ERO1A a perfect target for cancer therapy. Here, we report the upregulation of ERO1A in a cohort of aggressive triple-negative breast cancer (TNBC) patients in which ERO1A levels correlate with a higher risk of breast tumor recurrence and metastatic spread. For ERO1A target validation and therapy in TNBC, we designed new ERO1A inhibitors in a structure-activity campaign of the prototype EN460. Cell-based screenings showed that the presence of the Micheal acceptor in the compound is necessary to engage the cysteine 397 of ERO1A but not sufficient to set out the inhibitory effect on ERO1A. Indeed, the ERO1 inhibitor must adopt a non-coplanar rearrangement within the ERO1A binding site. I2 and I3, two new EN460 analogs with different phenyl-substituted moieties, efficiently inhibited ERO1A, blunting VEGFA secretion. Accordingly, in vitro assays to measure ERO1A engagement and inhibition confirmed that I2 and I3 bind ERO1A and restrain its activity with a IC50 in a low micromolar range. EN460, I2 and I3 triggered breast cancer cytotoxicity while specifically inhibiting ERO1A in a dose-dependent manner. I2 more efficiently impaired cancer-relevant features such as VEGFA secretion and related cell migration. I2 also acted on the tumor microenvironment and viability of xenografts and syngeneic TNBC. Thus, small molecule-mediated ERO1A pharmacological inhibition is feasible and promises to lead to effective therapy for the still incurable TNBC.
Collapse
Affiliation(s)
- Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alexander Chernorudskiy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Alice Marrazza
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alfredo Cagnotto
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Chiara Grasselli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Augusto Bleve
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daniele Generali
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Manuela Milani
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Giovanni Bottegoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Massimo Broggini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
2
|
Ji X, Chen Z, Wang Y, Huo X, Liang X, Wang H, Xu M. ERP44 could serve as a bridge mediating prognosis and immunity for glioma via single-cell and bulk RNA-sequencing. Gene 2025; 933:148963. [PMID: 39341519 DOI: 10.1016/j.gene.2024.148963] [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: 03/29/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND There was evidence that ERP44 played vital roles in a variety of cancers. However, currently, ERP44 was rarely mentioned in gliomas. Therefore, we firstly integrated proteomics, bulk, as well as single-cell RNA-sequencing (scRNA-seq) to study the possible functions of ERP44 in glioma patients. METHODS From online databases, we obtained bulk RNA-seq, scRNA-seq, and proteomic data of ERP44 in gliomas and verified the expression of ERP44 by qRT-PCR. Then, the Noman diagram, gene set enrichment analysis (GSEA), and univariate/multivariate Cox regression analysis were all carried out in turn. Further discussions were also conducted regarding tumor immunity and ERP44 expression. RESULTS ERP44 in glioma tissues was found to be considerably higher than that in normal tissues (P<0.05) in the TCGA dataset, as well as the verification of GSE50161, GSE4290, and qRT-PCR results. High ERP44 expression indicated poorer overall survival (OS) for glioma (P<0.05), and it might also be used to predict gliomas' OS independently (P<0.05). In order to estimate these patients' survival prognosis, a Noman chart was created with effectiveness. According to GSEA analysis, ERP44 might be implicated in five significant pathways in gliomas. The levels of immune cell infiltration of LGG, the tumor immune microenvironments, the immunological checkpoints of LGG, and GBM were all strongly linked with ERP44 in terms of tumor immunity (P<0.05). Further scRNA-seq analysis revealed that ERP44 could be expressed in various cell types, including T cells, Mono/Macrophages, and malignant cells. CONCLUSIONS ERP44 was an oncogenic gene in gliomas, serving as a bridge mediating prognosis and immunity.
Collapse
Affiliation(s)
- Xiang Ji
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Zhenglou Chen
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Yunjiang Wang
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Xuqi Huo
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Xiaodong Liang
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Hongsheng Wang
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China
| | - Min Xu
- Department of Neurosurgery, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, China; Department of Neurosurgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China; Department of Neurosurgery, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China; Department of Neurosurgery, The Affiliated Yancheng Hospital of Southeast University, Yancheng, Jiangsu, China.
| |
Collapse
|
3
|
Chen F, Gao K, Li Y, Li Y, Wu Y, Dong L, Yang Z, Shi J, Guo K, Gao Q, Lu H, Zhang S. ST3GAL1 Promotes Malignant Phenotypes in Intrahepatic Cholangiocarcinoma. Mol Cell Proteomics 2024; 23:100821. [PMID: 39069074 PMCID: PMC11385758 DOI: 10.1016/j.mcpro.2024.100821] [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: 10/22/2023] [Revised: 07/14/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) has a poor prognosis, and elucidation of the molecular mechanisms underlying iCCA malignancy is of great significance. Glycosylation, an important post-translational modification, is closely associated with tumor progression. Altered glycosylation, including aberrant sialylation resulting from abnormal expression of sialyltransferases (STs) and neuraminidases (NEUs), is a significant feature of cancer cells. However, there is limited information on the roles of STs and NEUs in iCCA malignancy. Here, utilizing our proteogenomic resources from a cohort of 262 patients with iCCA, we identified ST3GAL1 as a prognostically relevant molecule in iCCA. Moreover, overexpression of ST3GAL1 promoted proliferation, migration, and invasion and inhibited apoptosis of iCCA cells in vitro. Through proteomic analyses, we identified the downstream pathway potentially regulated by ST3GAL1, which was the NF-κB signaling pathway, and further demonstrated that this pathway was positively correlated with malignancy in iCCA cells. Notably, glycoproteomics showed that O-glycosylation was changed in iCCA cells with high ST3GAL1 expression. Importantly, the altered O-glycopeptides underscored the potential utility of O-glycosylation profiling as a discriminatory marker for iCCA cells with ST3GAL1 overexpression. Additionally, miR-320b was identified as a post-transcriptional regulator of ST3GAL1, capable of suppressing ST3GAL1 expression and then reducing the proliferation, migration, and invasion abilities of iCCA cell lines. Taken together, these results suggest ST3GAL1 could serve as a promising therapeutic target for iCCA.
Collapse
Affiliation(s)
- Fanghua Chen
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Ke Gao
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Yan Li
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Yin Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingcheng Wu
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Liangqing Dong
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Zijian Yang
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Jieyi Shi
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Qiang Gao
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Haojie Lu
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China; Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Shu Zhang
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Bovo E, Jamrozik T, Kahn D, Karkut P, Robia SL, Zima AV. Phosphorylation of phospholamban promotes SERCA2a activation by dwarf open reading frame (DWORF). Cell Calcium 2024; 121:102910. [PMID: 38823350 PMCID: PMC11247691 DOI: 10.1016/j.ceca.2024.102910] [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: 03/22/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024]
Abstract
In cardiac myocytes, the type 2a sarco/endoplasmic reticulum Ca-ATPase (SERCA2a) plays a key role in intracellular Ca regulation. Due to its critical role in heart function, SERCA2a activity is tightly regulated by different mechanisms, including micropeptides. While phospholamban (PLB) is a well-known SERCA2a inhibitor, dwarf open reading frame (DWORF) is a recently identified SERCA2a activator. Since PLB phosphorylation is the most recognized mechanism of SERCA2a activation during adrenergic stress, we studied whether PLB phosphorylation also affects SERCA2a regulation by DWORF. By using confocal Ca imaging in a HEK293 expressing cell system, we analyzed the effect of the co-expression of PLB and DWORF using a bicistronic construct on SERCA2a-mediated Ca uptake. Under these conditions of matched expression of PLB and DWORF, we found that SERCA2a inhibition by non-phosphorylated PLB prevails over DWORF activating effect. However, when PLB is phosphorylated at PKA and CaMKII sites, not only PLB's inhibitory effect was relieved, but SERCA2a was effectively activated by DWORF. Förster resonance energy transfer (FRET) analysis between SERCA2a and DWORF showed that DWORF has a higher relative affinity for SERCA2a when PLB is phosphorylated. Thus, SERCA2a regulation by DWORF responds to the PLB phosphorylation status, suggesting that DWORF might contribute to SERCA2a activation during conditions of adrenergic stress.
Collapse
Affiliation(s)
- Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Thomas Jamrozik
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Patryk Karkut
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
5
|
Germani S, Van Ho AT, Cherubini A, Varone E, Chernorudskiy A, Renna GM, Fumagalli S, Gobbi M, Lucchetti J, Bolis M, Guarrera L, Craparotta I, Rastelli G, Piccoli G, de Napoli C, Nogara L, Poggio E, Brini M, Cattaneo A, Bachi A, Simmen T, Calì T, Quijano-Roy S, Boncompagni S, Blaauw B, Ferreiro A, Zito E. SEPN1-related myopathy depends on the oxidoreductase ERO1A and is druggable with the chemical chaperone TUDCA. Cell Rep Med 2024; 5:101439. [PMID: 38402623 PMCID: PMC10982971 DOI: 10.1016/j.xcrm.2024.101439] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/06/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024]
Abstract
Selenoprotein N (SEPN1) is a protein of the endoplasmic reticulum (ER) whose inherited defects originate SEPN1-related myopathy (SEPN1-RM). Here, we identify an interaction between SEPN1 and the ER-stress-induced oxidoreductase ERO1A. SEPN1 and ERO1A, both enriched in mitochondria-associated membranes (MAMs), are involved in the redox regulation of proteins. ERO1A depletion in SEPN1 knockout cells restores ER redox, re-equilibrates short-range MAMs, and rescues mitochondrial bioenergetics. ERO1A knockout in a mouse background of SEPN1 loss blunts ER stress and improves multiple MAM functions, including Ca2+ levels and bioenergetics, thus reversing diaphragmatic weakness. The treatment of SEPN1 knockout mice with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) mirrors the results of ERO1A loss. Importantly, muscle biopsies from patients with SEPN1-RM exhibit ERO1A overexpression, and TUDCA-treated SEPN1-RM patient-derived primary myoblasts show improvement in bioenergetics. These findings point to ERO1A as a biomarker and a viable target for intervention and to TUDCA as a pharmacological treatment for SEPN1-RM.
Collapse
Affiliation(s)
- Serena Germani
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Andrew Tri Van Ho
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | - Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Bioinformatics Core Unit, Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giorgia Rastelli
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgia Piccoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cosimo de Napoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Elena Poggio
- Department of Biology, University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Pharmaceutical Sciences, University of Padova, Padova, Italy; Department of Biology, University of Padova, Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy
| | | | - Angela Bachi
- IFOM-ETS AIRC Institute of Molecular Oncology, Milan, Italy
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Susana Quijano-Roy
- APHP-Université Paris-Saclay, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, FILNEMUS, ERN-Euro-NMD, Creteil, France; Pediatric Neurology and ICU Department, DMU Santé Enfant Adolescent (SEA), Raymond Poincaré University Hospital, Garches, France
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France; APHP, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, Neuromyology Department, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
6
|
Zhang X, Zhou Y, Chang X, Wu Q, Liu Z, Liu R. Tongyang Huoxue decoction (TYHX) ameliorating hypoxia/reoxygenation-induced disequilibrium of calcium homeostasis via regulating β-tubulin in rabbit sinoatrial node cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117006. [PMID: 37544340 DOI: 10.1016/j.jep.2023.117006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE β-tubulin is a skeletal protein of sinoatrial node cells (SANCs) that maintains the physiological structure of SANCs and inhibits calcium overload. Tongyang Huoxue decoction (TYHX) is widely used to treat sick sinus syndrome (SSS) owing to its effects on calcium channels regulation and SANCs protection. AIM OF THE STUDY This study focuses on the mechanism of TYHX in improving the hypoxia/reoxygenation (H/R)-induced disequilibrium of calcium homeostasis in SANCs via regulating β-tubulin. MATERIALS AND METHODS Real-Time PCR (RT-PCR) and Western Blot were adopted to detect the mRNA and protein expression levels of calcium channel regulatory molecules. Laser confocal method was employed to examine β-tubulin structure and fluorescence expression levels in SANCs, as well as calcium wave and calcium release levels. RESULTS It was found that the fluorescence expression level decreased and the β-tubulin structure of SANCs was damaged after H/R treatment. The mRNA and protein expression levels of SERCA2a/CaV1.3/NCX and β-tubulin decreased, while the mRNA and protein expression of RyR2 increased. The results of calcium wave and calcium transient experiments showed that the fluorescence expression level of Ca2+ increased and calcium overload occurred in SANCs. After treatment with TYHX, the mRNA and protein expression levels of SERCA2a/CaV1.3/NCX and β-tubulin increased, while the mRNA and protein expression levels of RyR2 decreased and the cell structure was restored. Interestingly, the regulation of TYHX on calcium homeostasis was further enhanced after Ad-β-tubulin treatment and counteracted after siRNA-β-tubulin treatment. These results suggest that TYHX could maintain calcium homeostasis via regulating β-tubulin, thus protecting against H/R-induced SANCs injury, which may be a new target for SSS treatment.
Collapse
Affiliation(s)
- Xinai Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yutong Zhou
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing Chang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaomin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiming Liu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Riuxiu Liu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Urrutia PJ, Bórquez DA. Expanded bioinformatic analysis of Oximouse dataset reveals key putative processes involved in brain aging and cognitive decline. Free Radic Biol Med 2023; 207:200-211. [PMID: 37473875 DOI: 10.1016/j.freeradbiomed.2023.07.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
The theory that aging is driven by the damage produced by reactive oxygen species (ROS) derived from oxidative metabolism dominated geroscience studies during the second half of the 20th century. However, increasing evidence that ROS also plays a key role in the physiological regulation of numerous processes through the reversible oxidation of cysteine residues in proteins, has challenged this notion. Currently, the scope of redox signaling has reached proteomic dimensions through mass spectrometry techniques. Here, we perform a comprehensive bioinformatics analysis of cysteine oxidation changes during mouse brain aging, using the quantitative data provided in the Oximouse dataset. Interestingly, our unbiased analysis identified hundreds of putative cysteine redox switches covering several pathways previously associated with aging. These include the ubiquitin-proteasome pathway and one-carbon metabolism (folate cycle, methionine cycle, transsulfuration and polyamine pathways). Surprisingly, cysteine oxidation changes are enriched in synaptic proteins in a highly asymmetric distribution: while postsynaptic proteins tend to increase cysteine oxidation with age, the opposite occurs for presynaptic proteins. Additionally, cysteine oxidation changes during aging are associated with proteins involved in the regulation of the mitochondrial transition pore opening and synaptic calcium homeostasis. Our analysis reinforces the concept that brain aging is associated with selective changes in the oxidation state of key proteins, rather than an overall trend toward increased oxidation. Also, we provide a prioritized list of specific cysteine residues with putative impact in aging processes for future experimental validation.
Collapse
Affiliation(s)
- Pamela J Urrutia
- Institute for Nutrition & Food Technology (INTA), Universidad de Chile, El Líbano 5524, Santiago, 7830490, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, 7800003, Chile
| | - Daniel A Bórquez
- Laboratory of Cell Signaling & Bioinformatics, Center for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, Ejército Libertador 141, Santiago, 8370007, Chile.
| |
Collapse
|
8
|
Dries E, Gilbert G, Roderick HL, Sipido KR. The ryanodine receptor microdomain in cardiomyocytes. Cell Calcium 2023; 114:102769. [PMID: 37390591 DOI: 10.1016/j.ceca.2023.102769] [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: 05/02/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
The ryanodine receptor type 2 (RyR) is a key player in Ca2+ handling during excitation-contraction coupling. During each heartbeat, RyR channels are responsible for linking the action potential with the contractile machinery of the cardiomyocyte by releasing Ca2+ from the sarcoplasmic reticulum. RyR function is fine-tuned by associated signalling molecules, arrangement in clusters and subcellular localization. These parameters together define RyR function within microdomains and are subject to disease remodelling. This review describes the latest findings on RyR microdomain organization, the alterations with disease which result in increased subcellular heterogeneity and emergence of microdomains with enhanced arrhythmogenic potential, and presents novel technologies that guide future research to study and target RyR channels within specific microdomains.
Collapse
Affiliation(s)
- Eef Dries
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Guillaume Gilbert
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Laboratoire ORPHY EA 4324, Université de Brest, Brest, France
| | - H Llewelyn Roderick
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Karin R Sipido
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Zhao YT, Liu YR, Yan YF, Tang ZS, Duan JA, Yang H, Song ZX, You XL, Wang MG. Fushenmu treatment ameliorates RyR2 with related metabolites in a zebrafish model of barium chloride induced arrhythmia. Chin Med 2023; 18:103. [PMID: 37598173 PMCID: PMC10439546 DOI: 10.1186/s13020-023-00812-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 07/27/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Fushenmu (Pini Radix in Poria, FSM) is a folk parasitic herb that has been mainly used for palpitation and amnesiain in traditional Chinese medicine (TCM). Recently, as an individual herb or a component of formulations, Fushenmu exhibits therapeutic potential for the treatment of cardiac arrhythmias. Yet, how specific targets or pathways of Fushenmu inhibit arrhythmia has not yet been reported. METHODS Here, based on clinical functional genomics, metabolomics and molecular biologic technologies, a network construction strategy was adopted to identify FSM therapeutic targets and biomarkers that might explore its functions. RESULTS In this study, it was found that FSM recovered arrhythmia-associated heart failure in barium chloride (BaCl2) induced arrhythmic zebrafish embryos, as was evidenced by the shortened cardiac sinus venosus-bulbus arteriosus (SV-BA) distance, smaller cardiovascular bleeding areas, and reduced cardiomyocyte apoptosis. Moreover, analysis via ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-QTOF-ESI-MS/MS) components identification and network pharmacology prediction showed that 11 main active components of FSM acted on 33 candidate therapeutic targets. Metabolomic analysis also suggested that FSM could rescue 242 abnormal metabolites from arrhythmic zebrafish embryos. Further analysis based on the combination of target prediction and metabolomic results illustrated that FSM down-regulated Ryanodine Receptor 2 (RyR2) expressions, inhibited adrenaline and 3',5'-Cyclic AMP (cAMP) levels in a dose-dependent manner, which was confirmed by metabolites quantification and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assay. CONCLUSION In summary, this study revealed that FSM mitigated BaCl2 induced cardiac damage caused by arrhythmia by suppressing RyR2 expressions, decreasing adrenaline and cAMP through the adrenergic signalling pathway.
Collapse
Affiliation(s)
- Yan-Ting Zhao
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China
| | - Yan-Ru Liu
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China.
| | - Ya-Feng Yan
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China
| | - Zhi-Shu Tang
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China.
- China Academy of Chinese Medical Sciences, No. 16, Nanxiao Street, Dongzhimen, Beijing, 100700, People's Republic of China.
| | - Jin-Ao Duan
- Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing, 210023, People's Republic of China
| | - Hui Yang
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China
| | - Zhong-Xing Song
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China
| | - Xue-Lian You
- Shaanxi Collaborative Innovation Center Medicinal Resource Industrialization, Shaanxi University of Chinese Medicine, No. 1 Weiyang Road, Qindu District, Xianyang, 712083, People's Republic of China
| | - Ming-Geng Wang
- Shandong Buchang Pharmaceutical Co. Ltd, Heze, 250000, Shandong, People's Republic of China
| |
Collapse
|
10
|
Nikolaienko R, Bovo E, Kahn D, Gracia R, Jamrozik T, Zima AV. Cysteines 1078 and 2991 cross-linking plays a critical role in redox regulation of cardiac ryanodine receptor (RyR). Nat Commun 2023; 14:4498. [PMID: 37495581 PMCID: PMC10372021 DOI: 10.1038/s41467-023-40268-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
The most common cardiac pathologies, such as myocardial infarction and heart failure, are associated with oxidative stress. Oxidation of the cardiac ryanodine receptor (RyR2) Ca2+ channel causes spontaneous oscillations of intracellular Ca2+, resulting in contractile dysfunction and arrhythmias. RyR2 oxidation promotes the formation of disulfide bonds between two cysteines on neighboring RyR2 subunits, known as intersubunit cross-linking. However, the large number of cysteines in RyR2 has been a major hurdle in identifying the specific cysteines involved in this pathology-linked post-translational modification of the channel. Through mutagenesis of human RyR2 and in-cell Ca2+ imaging, we identify that only two cysteines (out of 89) in each RyR2 subunit are responsible for half of the channel's functional response to oxidative stress. Our results identify cysteines 1078 and 2991 as a redox-sensitive pair that forms an intersubunit disulfide bond between neighboring RyR2 subunits during oxidative stress, resulting in a pathological "leaky" RyR2 Ca2+ channel.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Ryan Gracia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Thomas Jamrozik
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA.
| |
Collapse
|
11
|
Thomas NL, Dart C, Helassa N. Editorial: The role of calcium and calcium binding proteins in cell physiology and disease. Front Physiol 2023; 14:1228885. [PMID: 37362430 PMCID: PMC10289193 DOI: 10.3389/fphys.2023.1228885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Affiliation(s)
- N. Lowri Thomas
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - C. Dart
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, North West England, United Kingdom
| | - N. Helassa
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, North West England, United Kingdom
| |
Collapse
|
12
|
Nichtová Z, Fernandez-Sanz C, De La Fuente S, Yuan Y, Hurst S, Lanvermann S, Tsai HY, Weaver D, Baggett A, Thompson C, Bouchet-Marquis C, Várnai P, Seifert EL, Dorn GW, Sheu SS, Csordás G. Enhanced Mitochondria-SR Tethering Triggers Adaptive Cardiac Muscle Remodeling. Circ Res 2023; 132:e171-e187. [PMID: 37057625 PMCID: PMC10213149 DOI: 10.1161/circresaha.122.321833] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 03/29/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Cardiac contractile function requires high energy from mitochondria, and Ca2+ from the sarcoplasmic reticulum (SR). Via local Ca2+ transfer at close mitochondria-SR contacts, cardiac excitation feedforward regulates mitochondrial ATP production to match surges in demand (excitation-bioenergetics coupling). However, pathological stresses may cause mitochondrial Ca2+ overload, excessive reactive oxygen species production and permeability transition, risking homeostatic collapse and myocyte loss. Excitation-bioenergetics coupling involves mitochondria-SR tethers but the role of tethering in cardiac physiology/pathology is debated. Endogenous tether proteins are multifunctional; therefore, nonselective targets to scrutinize interorganelle linkage. Here, we assessed the physiological/pathological relevance of selective chronic enhancement of cardiac mitochondria-SR tethering. METHODS We introduced to mice a cardiac muscle-specific engineered tether (linker) transgene with a fluorescent protein core and deployed 2D/3D electron microscopy, biochemical approaches, fluorescence imaging, in vivo and ex vivo cardiac performance monitoring and stress challenges to characterize the linker phenotype. RESULTS Expressed in the mature cardiomyocytes, the linker expanded and tightened individual mitochondria-junctional SR contacts; but also evoked a marked remodeling with large dense mitochondrial clusters that excluded dyads. Yet, excitation-bioenergetics coupling remained well-preserved, likely due to more longitudinal mitochondria-dyad contacts and nanotunnelling between mitochondria exposed to junctional SR and those sealed away from junctional SR. Remarkably, the linker decreased female vulnerability to acute massive β-adrenergic stress. It also reduced myocyte death and mitochondrial calcium-overload-associated myocardial impairment in ex vivo ischemia/reperfusion injury. CONCLUSIONS We propose that mitochondria-SR/endoplasmic reticulum contacts operate at a structural optimum. Although acute changes in tethering may cause dysfunction, upon chronic enhancement of contacts from early life, adaptive remodeling of the organelles shifts the system to a new, stable structural optimum. This remodeling balances the individually enhanced mitochondrion-junctional SR crosstalk and excitation-bioenergetics coupling, by increasing the connected mitochondrial pool and, presumably, Ca2+/reactive oxygen species capacity, which then improves the resilience to stresses associated with dysregulated hyperactive Ca2+ signaling.
Collapse
Affiliation(s)
- Zuzana Nichtová
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Celia Fernandez-Sanz
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
- These authors contributed equally
| | - Sergio De La Fuente
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
- These authors contributed equally
| | - Yuexing Yuan
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - Stephen Hurst
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | | | - Hui-Ying Tsai
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - David Weaver
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Ariele Baggett
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | | | | | - Péter Várnai
- Department of Physiology, Faculty of Medicine, Semmelweis Univ., Budapest, Hungary
| | - Erin L Seifert
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Gerald W Dorn
- Center for Pharmacogenomics, John T. Milliken Dep. Med., WUSM, St Louis, MO, USA
| | - Shey-Shing Sheu
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - György Csordás
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| |
Collapse
|
13
|
Clements RT, Terentyeva R, Hamilton S, Janssen PML, Roder K, Martin BY, Perger F, Schneider T, Nichtova Z, Das AS, Veress R, Lee BS, Kim DG, Koren G, Stratton MS, Csordas G, Accornero F, Belevych AE, Gyorke S, Terentyev D. Sexual dimorphism in bidirectional SR-mitochondria crosstalk in ventricular cardiomyocytes. Basic Res Cardiol 2023; 118:15. [PMID: 37138037 PMCID: PMC10156626 DOI: 10.1007/s00395-023-00988-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Calcium transfer into the mitochondrial matrix during sarcoplasmic reticulum (SR) Ca2+ release is essential to boost energy production in ventricular cardiomyocytes (VCMs) and match increased metabolic demand. Mitochondria from female hearts exhibit lower mito-[Ca2+] and produce less reactive oxygen species (ROS) compared to males, without change in respiration capacity. We hypothesized that in female VCMs, more efficient electron transport chain (ETC) organization into supercomplexes offsets the deficit in mito-Ca2+ accumulation, thereby reducing ROS production and stress-induced intracellular Ca2+ mishandling. Experiments using mitochondria-targeted biosensors confirmed lower mito-ROS and mito-[Ca2+] in female rat VCMs challenged with β-adrenergic agonist isoproterenol compared to males. Biochemical studies revealed decreased mitochondria Ca2+ uniporter expression and increased supercomplex assembly in rat and human female ventricular tissues vs male. Importantly, western blot analysis showed higher expression levels of COX7RP, an estrogen-dependent supercomplex assembly factor in female heart tissues vs males. Furthermore, COX7RP was decreased in hearts from aged and ovariectomized female rats. COX7RP overexpression in male VCMs increased mitochondrial supercomplexes, reduced mito-ROS and spontaneous SR Ca2+ release in response to ISO. Conversely, shRNA-mediated knockdown of COX7RP in female VCMs reduced supercomplexes and increased mito-ROS, promoting intracellular Ca2+ mishandling. Compared to males, mitochondria in female VCMs exhibit higher ETC subunit incorporation into supercomplexes, supporting more efficient electron transport. Such organization coupled to lower levels of mito-[Ca2+] limits mito-ROS under stress conditions and lowers propensity to pro-arrhythmic spontaneous SR Ca2+ release. We conclude that sexual dimorphism in mito-Ca2+ handling and ETC organization may contribute to cardioprotection in healthy premenopausal females.
Collapse
Affiliation(s)
- Richard T Clements
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island College of Pharmacy, Kingston, RI, USA
- Department of Medicine, Providence VAMC and Brown University, Providence, RI, USA
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Shanna Hamilton
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
| | - Karim Roder
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Benjamin Y Martin
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Fruzsina Perger
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Timothy Schneider
- Department of Pathology, Anatomy and Cell Biology, MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zuzana Nichtova
- Department of Pathology, Anatomy and Cell Biology, MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Anindhya S Das
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Roland Veress
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Beth S Lee
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Gideon Koren
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Matthew S Stratton
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Gyorgy Csordas
- Department of Pathology, Anatomy and Cell Biology, MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sandor Gyorke
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA.
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
14
|
Guidarelli A, Spina A, Fiorani M, Zito E, Cantoni O. Arsenite enhances ERO1α expression via ryanodine receptor dependent and independent mechanisms. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104080. [PMID: 36781116 DOI: 10.1016/j.etap.2023.104080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/23/2022] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Arsenite is a potent carcinogen and toxic compound inducing an array of deleterious effects via different mechanisms, which include the Ca2+-dependent formation of reactive oxygen species. The mechanism whereby the metalloid affects Ca2+ homeostasis involves an initial stimulation of the inositol 1, 4, 5-triphosphate receptor, an event associated with an endoplasmic reticulum (ER) stress leading to increased ERO1α expression, and ERO1α dependent activation of the ryanodine receptor (RyR). Ca2+ release from the RyR is then critically connected with the mitochondrial accumulation of Ca2+. We now report that the resulting formation of mitochondrial superoxide triggers a second mechanism of ER stress dependent ERO1α expression, which however fails to impact on Ca2+ release from the RyR or, more generally, on Ca2+ homeostasis. Our results therefore demonstrate that arsenite stimulates two different and sequential mechanisms leading to increased ERO1α expression with different functions, possibly due to their different subcellular compartmentalization.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Spina
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy; Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
15
|
Keefe JA, Moore OM, Ho KS, Wehrens XHT. Role of Ca 2+ in healthy and pathologic cardiac function: from normal excitation-contraction coupling to mutations that cause inherited arrhythmia. Arch Toxicol 2023; 97:73-92. [PMID: 36214829 PMCID: PMC10122835 DOI: 10.1007/s00204-022-03385-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 01/19/2023]
Abstract
Calcium (Ca2+) ions are a key second messenger involved in the rhythmic excitation and contraction of cardiomyocytes throughout the heart. Proper function of Ca2+-handling proteins is required for healthy cardiac function, whereas disruption in any of these can cause cardiac arrhythmias. This comprehensive review provides a broad overview of the roles of Ca2+-handling proteins and their regulators in healthy cardiac function and the mechanisms by which mutations in these proteins contribute to inherited arrhythmias. Major Ca2+ channels and Ca2+-sensitive regulatory proteins involved in cardiac excitation-contraction coupling are discussed, with special emphasis on the function of the RyR2 macromolecular complex. Inherited arrhythmia disorders including catecholaminergic polymorphic ventricular tachycardia, long QT syndrome, Brugada syndrome, short QT syndrome, and arrhythmogenic right-ventricular cardiomyopathy are discussed with particular emphasis on subtypes caused by mutations in Ca2+-handling proteins.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Oliver M Moore
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin S Ho
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA. .,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Zhao J, Li J, Li G, Chen M. The role of mitochondria-associated membranes mediated ROS on NLRP3 inflammasome in cardiovascular diseases. Front Cardiovasc Med 2022; 9:1059576. [PMID: 36588561 PMCID: PMC9794868 DOI: 10.3389/fcvm.2022.1059576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/01/2022] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) metabolism is essential for the homeostasis of cells. Appropriate production of ROS is an important signaling molecule, but excessive ROS production can damage cells. ROS and ROS-associated proteins can act as damage associated molecular pattern molecules (DAMPs) to activate the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in cardiovascular diseases. Previous studies have shown that there are connected sites, termed mitochondria-associated membranes (MAMs), between mitochondria and the endoplasmic reticulum. In cardiovascular disease progression, MAMs play multiple roles, the most important of which is the ability to mediate ROS generation, which further activates the NLPR3 inflammasome, exacerbating the progression of disease. In this review, the following topics will be covered: 1. Molecular structures on MAMs that can mediate ROS generation; 2. Specific mechanisms of molecule-mediated ROS generation and the molecules' roles in cardiovascular disease, 3. The effects of MAMs-mediated ROS on the NLRP3 inflammasome in cardiovascular disease. The purpose of this review is to provide a basis for subsequent clinical treatment development.
Collapse
Affiliation(s)
- Jiahao Zhao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junli Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Guoyong Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Mao Chen
| |
Collapse
|
17
|
Loss-of-rescue of Ryr1 I4895T-related pathology by the genetic inhibition of the ER stress response mediator CHOP. Sci Rep 2022; 12:20632. [PMID: 36450915 PMCID: PMC9712496 DOI: 10.1038/s41598-022-25198-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/25/2022] [Indexed: 12/07/2022] Open
Abstract
RYR1 is the gene encoding the ryanodine receptor 1, a calcium release channel of the endo/sarcoplasmic reticulum. I4898T in RYR1 is one of the most common mutations that give rise to central core disease (CCD), with a variable phenotype ranging from mild to severe myopathy to lethal early-onset core-rod myopathy. Mice with the corresponding I4895T mutation in Ryr1 present mild myopathy when the mutation is heterozygous while I4895T homozygous is perinatal-lethal. Here we show that skeletal muscles of I4895T homozygous mice at birth present signs of stress of the endoplasmic reticulum (ER stress) and of the related unfolded protein response (UPR) with increased levels of the maladaptive mediators CHOP and ERO1. To gain information on the role of CHOP in the pathogenesis of RYR1I4895T-related myopathy, we generated compound Ryr1I4895T, Chop knock-out (-/-) mice. However, the genetic deletion of Chop, although it attenuates ER stress in the skeletal muscle of the newborns, does not rescue any phenotypic or functional features of Ryr1I4895T in mice: neither the perinatal-lethal phenotype nor the inability of Ryr1I4895T to respond to its agonist caffeine, but protects from ER stress-induced apoptosis. These findings suggest that genetic deletion of the ER stress response mediator CHOP is not sufficient to counteract the pathological Ryr1I4895T phenotype.
Collapse
|
18
|
Hamilton S, Terentyev D. ER stress and calcium-dependent arrhythmias. Front Physiol 2022; 13:1041940. [PMID: 36425292 PMCID: PMC9679650 DOI: 10.3389/fphys.2022.1041940] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
The sarcoplasmic reticulum (SR) plays the key role in cardiac function as the major source of Ca2+ that activates cardiomyocyte contractile machinery. Disturbances in finely-tuned SR Ca2+ release by SR Ca2+ channel ryanodine receptor (RyR2) and SR Ca2+ reuptake by SR Ca2+-ATPase (SERCa2a) not only impair contraction, but also contribute to cardiac arrhythmia trigger and reentry. Besides being the main Ca2+ storage organelle, SR in cardiomyocytes performs all the functions of endoplasmic reticulum (ER) in other cell types including protein synthesis, folding and degradation. In recent years ER stress has become recognized as an important contributing factor in many cardiac pathologies, including deadly ventricular arrhythmias. This brief review will therefore focus on ER stress mechanisms in the heart and how these changes can lead to pro-arrhythmic defects in SR Ca2+ handling machinery.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States,*Correspondence: Shanna Hamilton,
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
19
|
van Opbergen CJ, Pérez-Hernández M, Delmar M. Luminal Oxidative Regulation of the Ryanodine Receptor: More Sides to the Story? Circ Res 2022; 130:725-727. [PMID: 35239403 PMCID: PMC8909915 DOI: 10.1161/circresaha.122.320798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Chantal J.M. van Opbergen
- Leon H Charney Division of Cardiology, Department of Medicine, NYU-Grossman School of Medicine, New York, USA
| | - Marta Pérez-Hernández
- Leon H Charney Division of Cardiology, Department of Medicine, NYU-Grossman School of Medicine, New York, USA
| | - Mario Delmar
- Leon H Charney Division of Cardiology, Department of Medicine, NYU-Grossman School of Medicine, New York, USA
| |
Collapse
|