1
|
Kim SM, Park K, Yun HJ, Kim JM, Choi KH, Park KC. Identification of new small molecules for selective inhibition of SERCA 1 in patient-derived metastatic papillary thyroid cancer. Br J Pharmacol 2025; 182:2392-2408. [PMID: 39924143 DOI: 10.1111/bph.17442] [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: 06/29/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Papillary thyroid cancer (PTC) is a general thyroid cancer subtype; however, PTC is associated with metastasis or recurrence via anti-cancer drug resistance, rendering it practically incurable. Therefore, effective and reliable clinical approaches are urgently required. EXPERIMENTAL APPROACH We demonstrated the coordinated up-regulation of sarco/endoplasmic reticulum (ER) calcium ATPase 1 (SERCA1) in metastatic PTC under treatment with sorafenib or lenvatinib. We screened novel drug candidates in a patient-derived lymph node metastatic PTC and compared outcomes with those in non-metastatic and main mass PTC in an in vitro and in vivo model to propose a new clinical strategy. KEY RESULTS In the current study using patient-derived metastatic PTC cells, SERCA1 was considerably increased under sorafenib- or lenvatinib-treated conditions. SERCA is a critical component in cytosolic free calcium regulation and is regulated by calcium/calmodulin-dependent protein kinase 2 alpha (CaMK2α) via NFκB. However, cardiac dysfunction was inevitable in vivo because of non-specific inhibition of SERCA isoforms by conventional SERCA inhibitors. This study designed a therapeutic approach with decreased cardiac dysfunction via SERCA1 isoform-specific inhibition by novel small molecules, CKP1 and CKP2, under severe ER stress conditions in patient-derived metastatic PTC. These novel SERCA1-specific inhibitors remarkably increased tumour shrinkage in the patient-derived metastatic PTC xenograft tumour model without cardiac dysfunction when used in combination with sorafenib or lenvatinib. CONCLUSION AND IMPLICATIONS These outcomes suggest the potential efficacy of the novel combination strategy that uses targeted therapy to treat malignant cancer cells, such as sorafenib- or lenvatinib-resistant cancer cells.
Collapse
Affiliation(s)
- Seok-Mo Kim
- Department of Surgery, Thyroid Cancer Center, Gangnam Severance Hospital, Institute of Refractory Thyroid Cancer, Yonsei University College of Medicine, Seoul, South Korea
| | - Keunwan Park
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, South Korea
| | - Hyeok Jun Yun
- Department of Surgery, Thyroid Cancer Center, Gangnam Severance Hospital, Institute of Refractory Thyroid Cancer, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung Min Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Hwa Choi
- Department of Urology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Ki Cheong Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Yang L, Liu L, Zhang P, Pan W, Huang H, Qi Y, Wang Y, Zhang R, Zhou P. Rigid and soft substrates respectively promote the myocardial differentiation and maturation of human embryonic stem cells using elastic PDMS with thick synthetic coating. Colloids Surf B Biointerfaces 2025; 250:114540. [PMID: 39904141 DOI: 10.1016/j.colsurfb.2025.114540] [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: 10/23/2024] [Revised: 01/16/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Cardiovascular disease is the predominant cause of mortality and severe disability. Cardiomyocytes (CMs) derived from human embryonic stem cells (hESCs) have good application prospects for treating this disease. Unfortunately, CMs generated via current methods are relatively immature, as proven by defects such as sarcomer-like structures, calcium processing capacity and mitochondrial maturity. Therefore, in this study, tunable PDMS substrates that modified with sufficiently thick synthetic coatings were prepared to regulate both the myocardial differentiation of hESCs and subsequent maturation. Surprisingly, the effect of substrate elasticity on the critical attachment of hESCs and hESC-CMs vanished when common Matrigel coatings were used, but apparent differences were detected in the synthetic group. Rigid substrates promoted the adhesion of hESCs but not hESC-CMs. Moreover, the PDMS substrates with the highest hardness remarkably promoted the myocardial differentiation of hESCs, which was even better than that of the rigid plate group. The softest PDMS achieved the best performance among the groups in terms of the maturation of hESC-CMs, as confirmed by enhanced functional, metabolic, and ultrastructural maturation. This study reveals the real impact of an elastic substrate on the adhesion, differentiation, and maturation of hESC-CMs, which has value for accelerating the development of clinically applicable mature hESC-CMs with high induction efficiency.
Collapse
Affiliation(s)
- Ling Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Lu Liu
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China
| | - Pengxia Zhang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Wen Pan
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Hongxin Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Yingbin Wang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China
| | - Rongzhi Zhang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China.
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China.
| |
Collapse
|
3
|
de Oliveira MR. Pre-clinical evidence for mitochondria as a therapeutic target for luteolin: A mechanistic view. Chem Biol Interact 2025; 413:111492. [PMID: 40154935 DOI: 10.1016/j.cbi.2025.111492] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Pre-clinical evidence indicates that mitochondria may be a therapeutic target for luteolin (3',4',5,7-tetrahydroxyflavone; LUT) in different conditions. LUT modulates mitochondrial physiology in in vitro, ex vivo, and in vivo experimental models. This flavone exerted mitochondria-related antioxidant and anti-apoptotic effects, stimulated mitochondrial fusion and fission, induced mitophagy, and promoted mitochondrial biogenesis in human and animal cells and tissues. Moreover, LUT modulated the activity of components of the oxidative phosphorylation (OXPHOS) system, improving the ability of mitochondria to produce adenosine triphosphate (ATP) in certain circumstances. The mechanism of action by which LUT promoted mitochondrial benefits and protection are not completely clear yet. Nonetheless, LUT is a potential candidate to be utilized in mitochondrial therapy in the future. In this work, it is explored the mechanisms of action by which LUT modulates mitochondrial physiology in different pre-clinical experimental models.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), CEP 78060-900, Cuiaba, Mato Grosso, Brazil; Grupo de Estudos em Terapia Mitocondrial, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
Viskupicova J, Michel Espinoza-Fonseca L. Allosteric modulation of SERCA pumps in health and disease: structural dynamics, posttranslational modifications, and therapeutic potential. J Mol Biol 2025:169200. [PMID: 40349954 DOI: 10.1016/j.jmb.2025.169200] [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: 03/15/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Sarco/endoplasmic reticulum (SR/ER) Ca2+-ATPase (SERCA) pumps are ubiquitous membrane proteins in all eukaryotic cells, playing a central role in maintaining intracellular calcium homeostasis by re-sequestering Ca2+ ions from the cytosol into the SR/ER at the expense of ATP hydrolysis. SERCA pumps are well-characterized components of the calcium transport machinery in the cell, playing a role in various physiological processes, including muscle contraction, energy metabolism, secretion exocytosis, gene expression, synaptic transmission, cell survival, and fertilization. Allosteric regulation of SERCA pumps plays a key role in health and disease, and modulation of the SERCA pumps has emerged as a therapeutic approach for the treatment of cardiovascular, muscular, metabolic, and neurodegenerative disorders. In this review, we provide a comprehensive overview of the structural dynamics underlying allosteric modulation of SERCA, focusing on the effects of endogenous regulatory proteins, Ca2+ ions, ATP, and small-molecule effectors on the dynamics and function of the pump. We also examine in detail the role of posttranslational modifications as allosteric modulators of SERCA function, focusing on the oxidative modifications S-glutathionylation, S-nitrosylation, tyrosine nitration, and carbonylation, and non-oxidative modifications that include SUMOylation, acetylation, O-GlcNAcylation, phosphorylation, and ubiquitination. Finally, we discuss the therapeutic potential and challenges of allosteric modulation of SERCA pumps, including the design of small-molecule effectors, microRNA-based interventions, and targeted strategies that modulate SERCA posttranslational regulation. Overall, this review aims to bridge the gap between the mechanisms underlying allosteric modulation of SERCA and the translation of basic science discoveries into effective therapies targeting SERCA pumps.
Collapse
Affiliation(s)
- Jana Viskupicova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology & Toxicology, Slovak Academy of Sciences, 84104 Bratislava, Slovakia.
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Gan L, Li R, Wang Q, Zhou J, Zhang M, Leng M, Zhao J, Yang F, Jia S, Huang W, Ye J, Zheng M, Wang T. PAD2 disturbs cardiomyocyte calcium homeostasis by citrullinating SERCA2a protein in hemorrhagic shock induced arrhythmia. J Trauma Acute Care Surg 2025:01586154-990000000-00974. [PMID: 40307970 DOI: 10.1097/ta.0000000000004644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
BACKGROUND Malignant arrhythmia induced by traumatic hemorrhage is a leading cause of early mortality in hemorrhagic shock. Understanding the mechanisms driving these arrhythmias and identifying therapeutic targets are critical for improving early survival in patients with traumatic hemorrhagic shock. METHODS Peripheral blood samples from patients with hemorrhagic shock were collected and analyzed for peptidylarginine deiminase 2 (PAD2) protein levels using ELISA. Pad2 knockout mice (Pad2-/-, Pad2 KO) were generated, and the hemorrhagic shock model was constructed via femoral artery cannulation and bloodletting. Cardiomyocytes were isolated and contractility and calcium content were measured by confocal microscopy. PAD2 subcellular localization was assessed through immunofluorescence and Western blotting. Proteins interacting with PAD2 in cardiomyocytes were identified using co-immunoprecipitation followed by mass spectrometry (CoIP-MS). The effect of PAD2 on sarcoplasmic reticulum calcium-ATPase 2a (SERCA2a) activity and citrullination was evaluated through enzyme activity assays and protein citrullination detection. AAV9-PAD2 was injected into mice via tail vein to induce in vivo overexpression of PAD2 in the myocardium. The effects of PAD2 enzymatic activity mutations and a PAD2-specific inhibitor on survival rate and arrhythmia following hemorrhagic shock were assessed through intraperitoneal injection. RESULTS PAD2 protein levels were significantly elevated in the peripheral blood of patients with hemorrhagic shock. Pad2 knockout improved calcium homeostasis in the sarcoplasmic reticulum of cardiomyocytes and alleviated post-shock arrhythmia in mice. Following hypoxia, PAD2 exhibited increased colocalization with the sarcoplasmic reticulum. During hypoxia, PAD2 inhibited SERCA2a activity through citrullination. AAV9-mediated overexpression of PAD2 in cardiomyocytes worsened both survival rates and the incidence of ventricular arrhythmia following hemorrhagic shock in mice. Conversely, PAD2 enzymatic activity mutations and a PAD2-specific inhibitor improved survival rates and reduced arrhythmia after hemorrhagic shock. CONCLUSION During myocardial hypoxia occurs in hemorrhagic shock, PAD2 reduces SERCA2a enzyme activity by citrullination, disrupting myocardial calcium homeostasis. Peptidylarginine deiminase 2 gene deficiency or inhibition improves ventricular arrhythmias and increases survival following hemorrhagic shock. LEVEL OF EVIDENCE Original Research-basic sciences research; not applicable.
Collapse
Affiliation(s)
- Lebin Gan
- From the Trauma Treatment Center (L.G., R.L., Q.W., J.Z., M.Z., W.H., T.W.), Peking University People's Hospital; Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education (L.G., R.L., Q.W., J.Z., M.Z., W.H., T.W.); National Center for Trauma Medicine (L.G., R.L., Q.W., J.Z., M.Z., W.H., T.W.); and Department of Physiology and Pathophysiology, School of Basic Medical Sciences (M.L., J.Z., F.Y., S.J., M.Z.), Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Fei X, Song C, Cui J, Li Y, Lei X, Tang H. The role of deubiquitinases in cardiovascular diseases: mechanisms and therapeutic implications. Front Cardiovasc Med 2025; 12:1582049. [PMID: 40376148 PMCID: PMC12078317 DOI: 10.3389/fcvm.2025.1582049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
Cardiovascular diseases (CVDs) have become the leading cause of death globally, surpassing infectious diseases and other chronic illnesses. The incidence and mortality rates of CVDs are rising worldwide, posing a key challenge in public health. The ubiquitination system is a vast and complex. It is an important post-translational modification that plays a crucial role in various cellular processes. Deubiquitination is catalyzed by deubiquitinases (DUBs), which remove ubiquitin (Ub) from ubiquitinated proteins, thereby reversing the ubiquitination process. DUBs play an important role in many biological processes, such as DNA repair, cell metabolism, differentiation, epigenetic regulation, and protein stability control. They also participate in the regulation of many signaling pathways associated with the development and progression of CVDs. In this review, we primarily focus on the role of DUBs in various key pathological mechanisms of atherosclerosis (AS), such as foam cell formation, vascular remodeling (VR), endothelial-to-mesenchymal transition (End-MT), and clonal hematopoiesis (CH). In the heart, we summarize the involvement of DUBs in diseases and pathological processes, including heart failure (HF), myocardial infarction (MI), myocardial hypertrophy (MH) and ischemia/reperfusion (I/R) injury. Additionally, we also explore the diabetic cardiomyopathy (DCM) and the use of doxorubicin-induced cardiotoxicity in clinical settings. A comprehensive understanding of deubiquitination may provide new insights for the treatment and drug design of CVDs.
Collapse
Affiliation(s)
- Xiangyu Fei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Chao Song
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jian Cui
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yuqing Li
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xiaoyong Lei
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Huifang Tang
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China
- Department of Cardiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Myocardial Injury in Hunan Province, The First Affiliated Hospital, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Shooshtarian AK, O'Gallagher K, Shah AM, Zhang M. SERCA2a dysfunction in the pathophysiology of heart failure with preserved ejection fraction: a direct role is yet to be established. Heart Fail Rev 2025; 30:545-564. [PMID: 39843817 PMCID: PMC11991975 DOI: 10.1007/s10741-025-10487-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
With rising incidence, mortality and limited therapeutic options, heart failure with preserved ejection fraction (HFpEF) remains one of the most important topics in cardiovascular medicine today. Characterised by left ventricular diastolic dysfunction partially due to impaired Ca2+ homeostasis, one ion channel in particular, SarcoEndoplasmic Reticulum Ca2+-ATPase (SERCA2a), may play a significant role in its pathophysiology. A better understanding of the complex mechanisms interplaying to contribute to SERCA2a dysfunction will help develop treatments targeting it and thus address the growing clinical challenge HFpEF poses. This review examines the conflicting evidence present for changes in SERCA2a expression and activity in HFpEF, explores potential underlying mechanisms, and finally evaluates the drug and gene therapy trials targeting SERCA2a in heart failure. Recent positive results from trials involving widely used anti-diabetic agents such as sodium-glucose co-transporter protein 2 inhibitors (SGLT2i) and glucagon-like peptide-1 (GLP-1) agonists offer advancement in HFpEF management. The potential interplay between these agents and SERCA2a regulation presents a novel angle that could open new avenues for modulating diastolic function; however, the mechanistic research in this emerging field is limited. Overall, the direct role of SERCA2a dysfunction in HFpEF remains undetermined, highlighting the need for well-designed pre-clinical studies and robust clinical trials.
Collapse
Affiliation(s)
- Adam Kia Shooshtarian
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Kevin O'Gallagher
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Min Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK.
| |
Collapse
|
8
|
Ye X, Wang M, Qiu S, Pan Y, Cui Y, Yu S. Estradiol Alleviates Elevated Temperature-Induced Damage in Yak Oviductal Epithelial Cells by Maintaining Endoplasmic Reticulum Calcium Homeostasis. Animals (Basel) 2025; 15:1305. [PMID: 40362120 PMCID: PMC12070912 DOI: 10.3390/ani15091305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The oviduct is an organ that participates in multiple critical reproductive processes and provides essential nutritional support while maintaining a specialized microenvironment. It is particularly vulnerable to damage following heat stress-induced hyperthermia. Therefore, mitigating heat-induced damage to oviduct epithelial cells while preserving their physiological integrity under hyperthermia represents a critical therapeutic goal. OBJECTIVE This study aims to simulate the cellular damage state in yak oviduct epithelial cells (YOECs) under thermal challenge by increasing the incubation temperature of cultured cells, while observing changes in cellular injury upon supplementation with 17β-estradiol (E2), in order to explore the underlying cellular regulatory mechanisms involved. RESULTS After 48 h of exposure to 41 °C, YOECs exhibited elevated HSP70 and HSP90 protein expression levels, reduced OVGP1 protein expression, and increased apoptotic cells. Compared to the 41 °C group, the E2 + 41 °C group displayed decreased HSP70 protein levels, increased OVGP1 protein expression, and reduced apoptotic cell numbers. Additionally, changes in endoplasmic reticulum calcium ion (ER-Ca2+) distribution and fluorescence intensity variations in ER-Ca2+ regulatory proteins SERCA and IP3R3 were analyzed in the 37 °C, 41 °C, and E2 + 41 °C groups. The ER-Ca2+ distribution pattern in the E2 + 41 °C group remained similar to that of the 37 °C group. However, the fluorescence intensity levels of SERCA and IP3R3 proteins in the E2 + 41 °C group did not recover to levels comparable to the 37 °C group. CONCLUSION These findings suggest that E2 may mitigate thermal challenge-induced cellular damage in YOECs by maintaining ER-Ca2+ homeostasis, thereby preserving cellular functionality under elevated temperatures.
Collapse
Affiliation(s)
- Xiaolin Ye
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Shantong Qiu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (X.Y.); (M.W.); (S.Q.); (Y.P.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
9
|
Zhu JX, Pan ZN, Li D. Intracellular calcium channels: Potential targets for type 2 diabetes mellitus? World J Diabetes 2025; 16:98995. [PMID: 40236861 PMCID: PMC11947915 DOI: 10.4239/wjd.v16.i4.98995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent metabolic disorder. Despite the availability of numerous pharmacotherapies, a range of adverse reactions, including hypoglycemia, gastrointestinal discomfort, and lactic acidosis, limits their patient applicability and long-term application. Therefore, it is necessary to screen novel therapeutic drugs for T2DM treatment that have high efficacy but few adverse effects. AMP-activated protein kinase (AMPK) stands out as one of the most powerful targets for T2DM treatment. It can be activated through energy-sensing or calcium signaling. Medications that activate AMPK through the energy-sensing mechanism exhibit remarkable potency, but they are accompanied by lactic acidosis, carrying an alarmingly high mortality rate. Interestingly, medications that activate AMPK through calcium signaling, such as gliclazide, seldom induce lactic acidosis. However, the efficacy of gliclazide is much lower than metformin. Therefore, it is necessary to explore targets that activate AMPK via calcium signaling to avoid lactic acidosis while maintaining high potency. Ion channels are the main controller of intracellular calcium flow. Specific agonists and inhibitors targeting ion channels have been reported to activate AMPK. In this review, we will summarize the structure and function of calcium-permeable ion channels and discuss the potential of targeting these calcium channels for T2DM treatment.
Collapse
Affiliation(s)
- Jia-Xuan Zhu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, Zhejiang Province, China
| | - Zhao-Nan Pan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, Zhejiang Province, China
| | - Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, Zhejiang Province, China
| |
Collapse
|
10
|
Lei PJ, Ruscic KJ, Roh K, Rajotte JJ, O'Melia MJ, Bouta EM, Marquez M, Pereira ER, Kumar AS, Razavi MS, Zhou H, Menzel L, Huang L, Kumra H, Duquette M, Huang P, Baish JW, Munn LL, Kurpios NA, Ubellacker JM, Padera TP. Aging-induced changes in lymphatic muscle cell transcriptomes are associated with reduced pumping of peripheral collecting lymphatic vessels in mice. Dev Cell 2025; 60:1118-1133.e5. [PMID: 39731913 PMCID: PMC11981864 DOI: 10.1016/j.devcel.2024.12.010] [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: 03/27/2024] [Revised: 08/23/2024] [Accepted: 12/04/2024] [Indexed: 12/30/2024]
Abstract
Lymphatic muscle cells (LMCs) within the wall of collecting lymphatic vessels exhibit tonic and autonomous phasic contractions, which drive active lymph transport to maintain tissue-fluid homeostasis and support immune surveillance. Damage to LMCs disrupts lymphatic function and is related to various diseases. Despite their importance, knowledge of the gene transcriptional signatures in LMCs and how they relate to lymphatic function in normal and disease contexts is largely missing. We have generated a comprehensive transcriptional single-cell atlas-including LMCs-of peripheral collecting lymphatic vessels from mice across the lifespan. We identified genes that distinguish LMCs from other types of muscle cells, characterized the phenotypical and transcriptomic changes in LMCs in aged vessels, and identified a proinflammatory microenvironment that suppresses the contractile apparatus in LMCs from advanced-aged mice. Our findings provide a valuable resource to accelerate future research for the identification of potential drug targets on LMCs to improve lymphatic vessel function.
Collapse
Affiliation(s)
- Pin-Ji Lei
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Katarina J Ruscic
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kangsan Roh
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Corrigan-Minehan Heart Center and Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Johanna J Rajotte
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Roswell Park Cancer Institute, Buffalo, NY 14203, USA
| | - Meghan J O'Melia
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Echoe M Bouta
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Marla Marquez
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ethel R Pereira
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mohammad S Razavi
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hengbo Zhou
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lutz Menzel
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Liqing Huang
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Heena Kumra
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mark Duquette
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Peigen Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - James W Baish
- Department of Biomedical Engineering, Bucknell University, Lewisburg, PA 17837, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
11
|
Liu X, Wang S, Liang C, Zhang S, Liu S, Hu Y. Ano5 deficiency disturbed bone formation by inducing osteoclast apoptosis in Gnathodiaphyseal dysplasia. Exp Cell Res 2025; 447:114493. [PMID: 40049314 DOI: 10.1016/j.yexcr.2025.114493] [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: 11/01/2024] [Revised: 01/16/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Gnathodiaphyseal dysplasia (GDD) is a rare genetic syndrome characterized by cemento-ossifying fibroma lesions in the mandible and sclerosis of tubular bones. Currently, the clinical treatment of GDD is limited to surgical resection; therefore, novel treatment strategies developed through exploration of the related mechanisms are needed. Mutations in the TMEM16E/ANO5 gene are considered the main pathogenic factor of GDD, and the Ano5 knockout mouse model (Ano5-/-) established previously, which presented GDD-like characteristics, exhibited decreased osteoclastogenesis. ANO5, a calcium-activated chloride channel (CaCC), plays an important role in the maintenance of intracellular calcium homeostasis, which is crucial for osteoclast differentiation. In this study, our data indicated that the intracellular calcium concentration ([Ca2+]i) and calcium transients were significantly decreased in Ano5-/- osteoclasts accompanied by abnormally altered expression of calcium transporters, resulting in calcium dyshomeostasis. In addition, the endoplasmic reticulum stress (ERS) response was significantly enhanced in Ano5-/- osteoclasts, possibly because of calcium dyshomeostasis, which leading to the increased proportion of apoptotic osteoclasts via the activation of the C/EBP homologous protein (CHOP) signalling pathway, accompanied by abnormal changes in the expression of apoptosis-related factors. In summary, Ano5 deficiency impairs the function of osteoclasts by increasing osteoclast apoptosis, which is induced by an overactivated ERS response via calcium dyshomeostasis.
Collapse
Affiliation(s)
- Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China; Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Shengnan Wang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Chao Liang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China; Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China.
| | - Shuai Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Sirui Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Ying Hu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
12
|
Yang N, Li L, Shi XL, Liu YP, Wen R, Yang YH, Zhang T, Yang XR, Xu YF, Liu CF, Ning W, Zhang TN. Succinylation of SERCA2a at K352 Promotes Its Ubiquitinoylation and Degradation by Proteasomes in Sepsis-Induced Heart Dysfunction. Circ Heart Fail 2025; 18:e012180. [PMID: 39996319 DOI: 10.1161/circheartfailure.124.012180] [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: 07/09/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Intracellular Ca2+ cycling governs effective myocardial systolic contraction and diastolic relaxation. SERCA2a (sarco/endoplasmic reticulum Ca2+ ATPase type 2a), which plays a crucial role in controlling intracellular Ca2+ signaling and myocardial cell function, is downregulated and inactivated during sepsis-induced heart dysfunction. However, the cause of this dysregulation remains unclear. In this study, we investigated the effect of lysine succinylation in lipopolysaccharide-induced septic heart dysfunction through global succinylome analysis of myocardial tissues from septic rats. METHODS We conducted a succinylome profiling and developed a protein language model-based framework to prioritize succinylation at a functionally important site, and further analysis revealed crosstalk between ubiquitination and succinylation of SERCA2a. The succinylation of SERCA2a in septic rats or lipopolysaccharide-treated cells were detected by co-immunoprecipitation. Thereafter, a desuccinylated SERCA2aK352R was introduced and its function and stability were determined by Ca2+ transient and Western blot, respectively. Meanwhile, the effect on SERCA2aK352R on heart function was assessed in vivo by echocardiography and hemodynamics. RESULTS We identified 10 324 succinylated lysine sites in heart tissues, including 1042 differentially succinylated lysine sites, in response to lipopolysaccharide. SERCA2a was hypersuccinylated in the myocardial tissues of septic rats and lipopolysaccharide-treated cardiomyocytes. Increased ubiquitination level, reduced protein level, and activity of SERCA2a were observed, along with increased succinylation of SERCA2a in vivo and in vitro. K352 was essential for SERCA2a succinylation, which reduced SERCA2a protein level by promoting formation of the K48 ubiquitin chain on SERCA2a and its degradation by proteasomes. Co-immunoprecipitation combined with liquid chromatography-tandem mass spectrometry identified that SIRT2 (sirtuin2), a deacylase, exhibited interaction with SERCA2a. Furthermore, SIRT2 decreased K352 succinylation of SERCA2a, suggesting that SIRT2 may function as a desuccinylase for SERCA2a. CONCLUSIONS Succinylation of SERCA2a at K352, which was controlled by SIRT2, promotes its ubiquitinoylation and degradation by proteasomes in sepsis-induced heart dysfunction.
Collapse
Affiliation(s)
- Ni Yang
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Linus Li
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China (L.L., W.N.)
| | - Xiao-Lu Shi
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing (X.-L.S.)
| | - Yong-Ping Liu
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Ri Wen
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Yu-Hang Yang
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Tao Zhang
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Xin-Ru Yang
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Yang-Fan Xu
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Chun-Feng Liu
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
| | - Wanshan Ning
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China (L.L., W.N.)
| | - Tie-Ning Zhang
- Department of Pediatrics, Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang (N.Y., Y.-P.L., R.W., Y.-H.Y., T.Z., X.-R.Y., Y.-F.X., C.-F.L., T.-N.Z.)
- Institute for Clinical Medical Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China (L.L., W.N.)
| |
Collapse
|
13
|
Ramadan MM, Alshawi AL, Mostafa YA, Al-Obeid MT, Elmahal M. Omecamtiv Mecarbil in Systolic Heart Failure: Clinical Efficacy and Future Directions of a Novel Myosin-Activating Inotropic Agent. Cureus 2025; 17:e82128. [PMID: 40357100 PMCID: PMC12067359 DOI: 10.7759/cureus.82128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) remains a global health challenge, associated with high morbidity, mortality, and rising healthcare costs. Despite advances in guideline-directed therapy, many patients, especially those with severely reduced ejection fraction, remain symptomatic. Traditional inotropes, including β-agonists and phosphodiesterase inhibitors, are limited in chronic HFrEF due to risks of arrhythmias, calcium overload, and increased myocardial oxygen demand. Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that enhances systolic contraction by increasing the efficiency of actin-myosin interactions. It prolongs systolic ejection time and improves stroke volume without elevating intracellular calcium or energy consumption. Although theoretical concerns exist regarding impaired diastolic filling, clinical trials have not confirmed such adverse effects in most patients. This narrative review discusses OM's pharmacologic profile, clinical trial data, and its potential as an adjunct therapy in advanced HFrEF. While not yet guideline-recommended, OM may benefit patients who remain symptomatic despite optimal treatment.
Collapse
Affiliation(s)
- Mahmoud M Ramadan
- Cardiology, Faculty of Medicine, Mansoura University, Mansoura, EGY
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | - Abdullah L Alshawi
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | - Yasmeen A Mostafa
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | | | - Mohammed Elmahal
- Diabetes and Endocrinology, Khartoum North Teaching Hospital, Khartoum, SDN
| |
Collapse
|
14
|
Liu W, Xu S, Wang J, Li X, Liu R, Zhao L, Li Y, Shi R, Zhang J. Allicin ameliorates acute myocardial infarction in rats by modulating calcium homeostasis in cardiomyocytes through the induction of hydrogen sulfide production. Front Pharmacol 2025; 16:1557685. [PMID: 40206075 PMCID: PMC11979285 DOI: 10.3389/fphar.2025.1557685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a common cardiovascular disease with high morbidity and mortality rates. Allicin, the primary active component of traditional Chinese herbs garlic, has multiple cardiovascular effects. However, the protective effect of allicin on AMI is rare. This study aimed to identify the pathways through which allicin stimulates hydrogen sulfide (H2S) production to regulate calcium ion (Ca2+) homeostasis in cardiomyocytes, thereby contributing to AMI protection. METHODS In this study, we established an AMI rat model by ligating the left anterior descending branch of the coronary artery to assess the therapeutic effect of allicin. We also investigated its influence on cardiomyocyte Ca2+ homeostasis. To determine the role of H2S production in the effects of allicin, we identified the H2S synthase in healthy rat myocardial tissue and serum and then applied H2S synthase inhibitors to block H2S production. RESULTS The results indicate that allicin significantly enhanced cardiac function, raised H2S levels in myocardial tissue and serum, reduced necrosis tissue size, decreased myocardial enzyme levels, and improved myocardial pathological changes. Surprisingly, allicin also notably increased H2S synthase levels. These findings suggest that allicin shields AMI rats by stimulating H2S production, acting both as a direct H2S donor and indirectly boosting H2S synthase expression. Furthermore, allicin enhanced Ca2+ homeostasis in cardiomyocytes by improving cardiomyocyte contraction kinetics and regulating the function and expression of key proteins related to Ca2+ transport in cardiomyocytes. The effect of allicin on Ca2+ homeostasis was partially decreased but not entirely abolished when H2S production was inhibited using H2S synthase inhibitors PAG and AOAA. This suggests that while the impact of allicin is strongly associated with H2S, additional independent mechanisms are also involved. CONCLUSION Our study presents novel evidence demonstrating that allicin modulates Ca2+ homeostasis in cardiomyocytes by stimulating H2S production, thereby conferring protection against AMI. Furthermore, the protective effects of allicin are partly mediated by, but not solely reliant on, the generation of H2S. These findings not only provide mechanistic insights into the anti-AMI effects of allicin but also underscore its therapeutic promise.
Collapse
Affiliation(s)
- Weiyu Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medic, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaojun Xu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medic, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juan Wang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medic, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinxia Li
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Ruiting Liu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Garlic Medicinal Research, Urumqi, China
| | - Le Zhao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medic, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yikui Li
- Health Prevention Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rongmei Shi
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Garlic Medicinal Research, Urumqi, China
| | - Jinyan Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medic, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Li D, Ma Q. Ubiquitin-specific protease: an emerging key player in cardiomyopathy. Cell Commun Signal 2025; 23:143. [PMID: 40102846 PMCID: PMC11921692 DOI: 10.1186/s12964-025-02123-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Protein quality control (PQC) plays a vital role in maintaining normal heart function, as cardiomyocytes are relatively sensitive to misfolded or damaged proteins, which tend to accumulate under pathological conditions. Ubiquitin-specific protease (USP) is the largest deubiquitinating enzyme family and a key component of the ubiquitin proteasome system (UPS), which is a non-lysosomal protein degradation machinery to mediate PQC in cells. USPs regulate the stability or activity of the target proteins that involve intracellular signaling, transcriptional control of inflammation, antioxidation, and cell growth. Recent studies demonstrate that the USPs can regulate fibrosis, lipid metabolism, glucose homeostasis, hypertrophic response, post-ischemic recovery and cell death such as apoptosis and ferroptosis in cardiomyocytes. Since myocardial cell loss is an important component of cardiomyopathy, therefore, these findings suggest that the UPSs play emerging roles in cardiomyopathy. This review briefly summarizes recent literature on the regulatory roles of USPs in the occurrence and development of cardiomyopathy, giving us new insights into the molecular mechanisms of USPs in different cardiomyopathy and potential preventive strategies for cardiomyopathy.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Qilin Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China.
| |
Collapse
|
16
|
Ning S, Li J, He M, Yu Y, Guo Z. Androgen receptor alleviates doxorubicin-induced endoplasmic reticulum stress and myocardial injury by interacting with SERCA2a. Free Radic Biol Med 2025; 230:127-137. [PMID: 39947494 DOI: 10.1016/j.freeradbiomed.2025.02.010] [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: 01/07/2025] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
The clinical use of the anticancer drug doxorubicin (DOX) is limited due to its time- and dose-dependent cardiotoxicity. Therefore, there is an urgent need to explore the molecular mechanism and coping strategies for alleviating DOX-induced cardiotoxicity (DIC) and solve the difficulties in clinical application. The role and mechanism of androgen receptor (AR), which is the target of androgen, in DIC remain unclear. Here, we elucidated the molecular mechanisms of AR in DOX-induced cardiotoxicity. Inhibition of AR aggravated the DOX-induced cardiac function impairment, while the activation of AR showed obvious therapeutic effect and rescued cardiac function of rats. AR can physically interact with SERCA2a. Activation of AR participates in the regulation of DOX-induced myocardial injury by modulating SERCA2a, attenuating DOX-induced endoplasmic reticulum stress, improving calcium (Ca2+) cycling homeostasis, and inhibiting ROS levels and apoptosis, thereby participating in the regulation of DOX induced myocardial injury. Altogether, these findings reveal for the first time the relationship and role between AR and SERCA2a in regulating the progression of DIC, suggesting that AR may play a therapeutic role as a new target against DIC.
Collapse
Affiliation(s)
- Shuwei Ning
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Jianhui Li
- Department of Pathology, Xuchang Central Hospital, Xuchang, 461000, China
| | - Mei He
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Yuexin Yu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
17
|
Kook T, Lee MY, Kwak TH, Jeong D, Sim DS, Jeong MH, Ahn Y, Kook H, Park WJ, Jang SP. Intratracheal Delivery of a Phospholamban Decoy Peptide Attenuates Cardiac Damage Following Myocardial Infarction. Int J Mol Sci 2025; 26:2649. [PMID: 40141290 PMCID: PMC11942360 DOI: 10.3390/ijms26062649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Heart failure (HF) remains a major cause of mortality worldwide. While novel approaches, including gene and cell therapies, show promise, efficient delivery methods for such biologics to the heart are critically needed. One emerging strategy is lung-to-heart delivery using nanoparticle (NP)-encapsulated biologics. This study examines the efficiency of delivering a therapeutic peptide conjugated to a cell-penetrating peptide (CPP) to the heart via the lung-to-heart route through intratracheal (IT) injection in mice. The CPP, a tandem repeat of NP2 (dNP2) derived from the human novel LZAP-binding protein (NLBP), facilitates intracellular delivery of the therapeutic payload. The therapeutic peptide, SE, is a decoy peptide designed to inhibit protein phosphatase 1 (PP1)-mediated dephosphorylation of phospholamban (PLN). Our results demonstrated that IT injection of dNP2-SE facilitated efficient delivery to the heart, with peak accumulation at 3 h post-injection. The administration of dNP2-SE significantly ameliorated morphological and functional deterioration of the heart under myocardial infarction. At the molecular level, dNP2-SE effectively prevented PLN dephosphorylation in the heart. Immunoprecipitation experiments further revealed that dNP2-SE binds strongly to PP1 and disrupts its interaction with PLN. Collectively, our findings suggest that lung-to-heart delivery of a CPP-conjugated therapeutic peptide, dNP2-SE, represents a promising approach for the treatment of HF.
Collapse
Affiliation(s)
- Taewon Kook
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mi-Young Lee
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Tae Hwan Kwak
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan 15588, Republic of Korea;
| | - Doo Sun Sim
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Woo Jin Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- BethphaGen, S3-203, Gwangju 61005, Republic of Korea
| | - Seung Pil Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Center for Gene and Cell Therapy, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
18
|
Fan Q, Wang Y, An Q, Ling Y. Right ventricular dysfunction following surgical repair of tetralogy of Fallot: Molecular pathways and therapeutic prospects. Biomed Pharmacother 2025; 184:117924. [PMID: 39983432 DOI: 10.1016/j.biopha.2025.117924] [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: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease (CHD). Although surgical correction of TOF is possible, patients often face challenges related to right ventricle dysfunction (RVD) post-surgery, which can significantly impact their long-term survival. The causes of RVD in TOF patients are complex, involving both the unique structural characteristics of the TOF heart and damage resulting from surgical interventions. Residual anatomical issues following TOF repair are often unavoidable, placing the RV under stress and leading to the activation of multiple molecular pathways. This review comprehensively outlines the causes of RVD in patients after TOF surgery, particularly focusing the molecular pathways that contribute to RVD, including established signaling pathways as well as emerging pathways identified through transcriptomic analysis of RV myocardium in TOF patients. We also highlight the features of these molecular pathways concerning RVD, as well as the influence of gender disparities on these molecular pathways. By interpreting the causes and molecular mechanisms underlying RVD after TOF surgery, this review provides new insights for managing RVD in repaired TOF, potentially paving the way for targeted therapies aimed at improving long-term outcomes for those affected by RVD.
Collapse
Affiliation(s)
- Qiang Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yabo Wang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yunfei Ling
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
19
|
Jiang P, Huang F, Chen L, Zhou H, Deng Y, Li L, Chen M, Huang Y. Intercellular NETwork-facilitated sarcoplasmic reticulum targeting for myocardial ischemia-reperfusion injury treatment. SCIENCE ADVANCES 2025; 11:eadr4333. [PMID: 39937916 PMCID: PMC11818016 DOI: 10.1126/sciadv.adr4333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) often leads to irreversible myocardium dysfunction, while existing therapies are palliatives that transiently alleviate the disease symptoms. Repairing sarcoplasmic reticulum Ca2+-ATPase (SERCA) could reverse MIRI, which, however, requires precise drug delivery to the sarcoplasmic reticulum (SR). To this end, we leverage cell-cell "NETwork" of neutrophils to deliver SERCA activator-loaded SR-localized nanoparticles (L-P-NPs) to the damaged myocardial cells, following a hierarchical targeting process: (i) chemotactic neutrophils deliver L-P-NPs to ischemia-reperfused heart, achieving tissue level targeting; (ii) neutrophils produce neutrophil extracellular traps (NETs) to transport L-P-NPs to injured myocardial cell, achieving cellular level targeting; (iii) L-P-NPs escort therapeutic payloads to the SR, achieving subcellular targeting. We showed that this platform profoundly restored SERCA activity, augmented cardiac function, and ameliorated adverse heart remodeling. Our study provides insight into the direct restoration of SR for the effective treatment of MIRI and other muscle diseases.
Collapse
Affiliation(s)
- Peihang Jiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fangyang Huang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases and Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases and Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yudi Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases and Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
20
|
Song W, Zhang Q, Cao Z, Jing G, Zhan T, Yuan Y, Kang N, Zhang Q. Targeting SERCA2 in Anti-Tumor Drug Discovery. Curr Drug Targets 2025; 26:1-16. [PMID: 39323343 DOI: 10.2174/0113894501325497240918042654] [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: 04/23/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
SERCA2, a P-type ATPase located on the endoplasmic reticulum of cells, plays an important role in maintaining calcium balance within cells by transporting calcium from the cytoplasm to the endoplasmic reticulum against its concentration gradient. A multitude of studies have demonstrated that the expression of SERCA2 is abnormal in a wide variety of tumor cells. Consequently, research exploring compounds that target SERCA2 may offer a promising avenue for the development of novel anti-tumor drugs. This review has summarized the anti-tumor compounds targeting SERCA2, including thapsigargin, dihydroartemisinin, curcumin, galangin, etc. These compounds interact with SERCA2 on the endoplasmic reticulum membrane, disrupting intracellular calcium ion homeostasis, leading to tumor cell apoptosis, autophagy and cell cycle arrest, ultimately producing anti-tumor effects. Additionally, several potential research directions for compounds targeting SERCA2 as clinical anti-cancer drugs have been proposed in the review. In summary, SERCA2 is a promising anti-tumor target for drug discovery and development.
Collapse
Affiliation(s)
- Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhiyong Cao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
21
|
Liu J, Xu S, Gao B, Yuan M, Zhong L, Guo R. Protective effect of SERCA2a-SUMOylation by SUMO-1 on diabetes-induced atherosclerosis and aortic vascular injury. Mol Cell Biochem 2025; 480:279-293. [PMID: 38438822 DOI: 10.1007/s11010-024-04953-x] [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: 10/09/2023] [Accepted: 01/26/2024] [Indexed: 03/06/2024]
Abstract
Diabetes is a major risk factor for cardiovascular disease. However, the exact mechanism by which diabetes contributes to vascular damage is not fully understood. The aim of this study was to investigate the role of SUMO-1 mediated SERCA2a SUMOylation in the development of atherosclerotic vascular injury associated with diabetes mellitus. ApoE-/- mice were treated with streptozotocin (STZ) injection combined with high-fat feeding to simulate diabetic atherosclerosis and vascular injury. Human aortic vascular smooth muscle cells (HAVSMCs) were treated with high glucose (HG, 33.3 mM) and palmitic acid (PA, 200 µM) for 24 h to mimic a model of diabetes-induced vascular injury in vitro. Aortic vascular function, phenotypic conversion, migration, proliferation, intracellular Ca2+ concentration, the levels of small ubiquitin-like modifier type 1 (SUMO1), SERCA2a and SUMOylated SERCA2a were detected. Diabetes-induced atherosclerotic mice presented obvious atherosclerotic plaques and vascular injury, companied by significantly lower levels of SUMO1 and SERCA2a in aorta. HG and PA treatment in HAVSMCs reduced the expressions of SUMO1, SERCA2a and SUMOylated SERCA2a, facilitated the HAVSMCs phenotypic transformation, proliferation and migration, attenuated the Ca2+ transport, and increased the resting intracellular Ca2+ concentration. We also confirmed that SUMO1 directly bound to SERCA2a in HAVSMCs. Overexpression of SUMO1 restored the function and phenotypic contractile ability of HAVSMCs by upregulating SERCA2a SUMOylation, thereby alleviating HG and PA-induced vascular injury. These observations suggest an essential role of SUMO1 to protect diabetes-induced atherosclerosis and aortic vascular injury by the regulation of SERCA2a-SUMOylation and calcium homeostasis.
Collapse
MESH Headings
- Animals
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- SUMO-1 Protein/metabolism
- Sumoylation
- Mice
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Humans
- Aorta/pathology
- Aorta/metabolism
- Male
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout, ApoE
Collapse
Affiliation(s)
- Jinlin Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Shifang Xu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Bin Gao
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Meng Yuan
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China.
- The Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, China.
| |
Collapse
|
22
|
Praveen J, Anusuyadevi M, Jayachandra KS. Unraveling the potential of Epicatechin gallate from crataegus oxyacantha in targeting aberrant cardiac Ca2+ signalling proteins: an in-depth in-silico investigation for heart failure therapy. J Biomol Struct Dyn 2024:1-15. [PMID: 39648361 DOI: 10.1080/07391102.2024.2435624] [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: 10/03/2023] [Accepted: 03/25/2024] [Indexed: 12/10/2024]
Abstract
The cardiovascular sarcoplasmic reticulum (SR) calcium (Ca2+) ATPase is an imperative determinant of cardiac functionality. In addition, anomalies in Ca2+ handling protein and atypical energy metabolism are inherent in heart failure (HF). Moreover, Ca2+ overload in SR leads to mitochondrial matrix Ca2+ overload, which can trigger the generation of Reactive Oxygen Species (ROS), culminating in the triggering of the Permeability Transition Pore (PTP) and Cytochrome C release, resulting in apoptosis that leads to arrhythmias and numerous disorders. Although proteins involved in the molecular mechanism of Ca2+ dysfunction regarding mitochondrial dysfunction remains elusive, this study aims to assess the major Ca2+ handling proteins which may be involved in the Ca2+ malfunction that causes mitochondrial dysfunction and predicting the most effective drug by targeting the analyzed Ca2+ handling proteins through various insilico analyses. Thirteen proteins absorbed from interaction analysis were docked with four optimal phytochemicals from Crataegus oxyacantha (COC). Furthermore, The ADME profile of tyramine, vitexin, Epicatechin, and Epicatechin gallate was acclimated to evaluate potential drugability utilizing QikProp. So, molecular docking evaluations were performed using Glide (Maestro), autodock, and vina. Based on the results of 156 dockings by Maestro, auto-dock, and auto-dock vina, PKAC-a with Epicatechin gallate exhibits good interaction. Therefore, a 2000 ns molecular dynamics (MD) simulation was utilized to assess the feasible phytochemical Epicatechin gallate - PKAC-a complex binding stability utilizing Desmond and this study confirmed that Epicatechin gallate from COC has high possibilities to inhibit the aberrant cardiac Ca2+ signaling proteins due to its conformational rigidity.
Collapse
Affiliation(s)
- J Praveen
- Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - M Anusuyadevi
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - K S Jayachandra
- Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| |
Collapse
|
23
|
Ryu G, Ko M, Lee S, Park SI, Choi JW, Lee JY, Kim JY, Kwon HJ. Urolithin A Protects Hepatocytes from Palmitic Acid-Induced ER Stress by Regulating Calcium Homeostasis in the MAM. Biomolecules 2024; 14:1505. [PMID: 39766212 PMCID: PMC11673756 DOI: 10.3390/biom14121505] [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: 10/06/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
An ellagitannin-derived metabolite, Urolithin A (UA), has emerged as a potential therapeutic agent for metabolic disorders due to its antioxidant, anti-inflammatory, and mitochondrial function-improving properties, but its efficacy in protecting against ER stress remains underexplored. The endoplasmic reticulum (ER) is a cellular organelle involved in protein folding, lipid synthesis, and calcium regulation. Perturbations in these functions can lead to ER stress, which contributes to the development and progression of metabolic disorders such as metabolic-associated fatty liver disease (MAFLD). In this study, we identified a novel target protein of UA and elucidated its mechanism for alleviating palmitic acid (PA)-induced ER stress. Cellular thermal shift assay (CETSA)-LC-MS/MS analysis revealed that UA binds directly to the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA), an important regulator of calcium homeostasis in mitochondria-associated ER membranes (MAMs). As an agonist of SERCA, UA attenuates abnormal calcium fluctuations and ER stress in PA-treated liver cells, thereby contributing to cell survival. The lack of UA activity in SERCA knockdown cells suggests that UA regulates cellular homeostasis through its interaction with SERCA. Collectively, our results demonstrate that UA protects against PA-induced ER stress and enhances cell survival by regulating calcium homeostasis in MAMs through SERCA. This study highlights the potential of UA as a therapeutic agent for metabolic disorders associated with ER stress.
Collapse
Affiliation(s)
- Gayoung Ryu
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Minjeong Ko
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Sooyeon Lee
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Se In Park
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Jin-Woong Choi
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
| | - Ju Yeon Lee
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Ho Jeong Kwon
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| |
Collapse
|
24
|
Yang B, Wang SQ, Yang HQ. β-adrenergic regulation of Ca 2+ signaling in heart cells. BIOPHYSICS REPORTS 2024; 10:274-282. [PMID: 39539286 PMCID: PMC11554573 DOI: 10.52601/bpr.2024.240906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/15/2024] [Indexed: 11/16/2024] Open
Abstract
β-adrenergic receptors (βARs) play significant roles in regulating Ca2+ signaling in cardiac myocytes, thus holding a key function in modulating heart performance. βARs regulate the influx of extracellular Ca2+ and the release and uptake of Ca2+ from the sarcoplasmic reticulum (SR) by activating key components such as L-type calcium channels (LTCCs), ryanodine receptors (RyRs) and phospholamban (PLN), mediated by the phosphorylation actions by protein kinase A (PKA). In cardiac myocytes, the presence of β2AR provides a protective mechanism against potential overstimulation of β1AR, which may aid in the restoration of cardiac dysfunctions. Understanding the Ca2+ regulatory signaling pathways of βARs in cardiac myocytes and the differences among various βAR subtypes are crucial in cardiology and hold great potential for developing treatments for heart diseases.
Collapse
Affiliation(s)
- Bo Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shi-Qiang Wang
- State Key Lab of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hua-Qian Yang
- Cyrus Tang Medical Institute, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
25
|
Parichatikanond W, Duangrat R, Kurose H, Mangmool S. Regulation of β-Adrenergic Receptors in the Heart: A Review on Emerging Therapeutic Strategies for Heart Failure. Cells 2024; 13:1674. [PMID: 39451192 PMCID: PMC11506672 DOI: 10.3390/cells13201674] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
The prolonged overstimulation of β-adrenergic receptors (β-ARs), a member of the G protein-coupled receptor (GPCR) family, causes abnormalities in the density and functionality of the receptor and contributes to cardiac dysfunctions, leading to the development and progression of heart diseases, especially heart failure (HF). Despite recent advancements in HF therapy, mortality and morbidity rates continue to be high. Treatment with β-AR antagonists (β-blockers) has improved clinical outcomes and reduced overall hospitalization and mortality rates. However, several barriers in the management of HF remain, providing opportunities to develop new strategies that focus on the functions and signal transduction of β-ARs involved in the pathogenesis of HF. As β-AR can signal through multiple pathways influenced by different receptor subtypes, expression levels, and signaling components such as G proteins, G protein-coupled receptor kinases (GRKs), β-arrestins, and downstream effectors, it presents a complex mechanism that could be targeted in HF management. In this narrative review, we focus on the regulation of β-ARs at the receptor, G protein, and effector loci, as well as their signal transductions in the physiology and pathophysiology of the heart. The discovery of potential ligands for β-AR that activate cardioprotective pathways while limiting off-target signaling is promising for the treatment of HF. However, applying findings from preclinical animal models to human patients faces several challenges, including species differences, the genetic variability of β-ARs, and the complexity and heterogeneity of humans. In this review, we also summarize recent updates and future research on the regulation of β-ARs in the molecular basis of HF and highlight potential therapeutic strategies for HF.
Collapse
Affiliation(s)
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Hitoshi Kurose
- Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan;
- Pharmacology for Life Sciences, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
26
|
Redwanz C, Pires RH, Biedenweg D, Groß S, Otto O, Könemann S. Endothelin-1 influences mechanical properties and contractility of hiPSC derived cardiomyocytes resulting in diastolic dysfunction. J Mol Cell Cardiol 2024; 194:105-117. [PMID: 39019395 DOI: 10.1016/j.yjmcc.2024.07.004] [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: 01/15/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
A better understanding of the underlying pathomechanisms of diastolic dysfunction is crucial for the development of targeted therapeutic options with the aim to increase the patients' quality of life. In order to shed light on the processes involved, suitable models are required. Here, effects of endothelin-1 (ET-1) treatment on cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) were investigated. While it is well established, that ET-1 treatment induces hypertrophy in cardiomyocytes, resulting changes in cell mechanics and contractile behavior with focus on relaxation have not been examined before. Cardiomyocytes were treated with 10 nM of ET-1 for 24 h and 48 h, respectively. Hypertrophy was confirmed by real-time deformability cytometry (RT-DC) which was also used to assess the mechanical properties of cardiomyocytes. For investigation of the contractile behavior, 24 h phase contrast video microscopy was applied. To get a deeper insight into changes on the molecular biological level, gene expression analysis was performed using the NanoString nCounter® cardiovascular disease panel. Besides an increased cell size, ET-1 treated cardiomyocytes are stiffer and show an impaired relaxation. Gene expression patterns in ET-1 treated hiPSC derived cardiomyocytes showed that pathways associated with cardiovascular diseases, cardiac hypertrophy and extracellular matrix were upregulated while those associated with fatty acid metabolism were downregulated. We conclude that alterations in cardiomyocytes after ET-1 treatment go far beyond hypertrophy and represent a useful model for diastolic dysfunction.
Collapse
Affiliation(s)
- Caterina Redwanz
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| | - Ricardo H Pires
- Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany
| | - Doreen Biedenweg
- Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany.
| | - Stefan Groß
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| | - Oliver Otto
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; Institute of Physics, University of Greifswald, Felix-Hausdorff-Straße 6, 17489 Greifswald, Germany; Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Straße 15a, 17489 Greifswald, Germany.
| | - Stephanie Könemann
- Department for Internal Medicine B, Cardiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| |
Collapse
|
27
|
Sun X, Zhou Q, Xiao C, Mao C, Liu Y, Chen G, Song Y. Role of post-translational modifications of Sp1 in cardiovascular diseases. Front Cell Dev Biol 2024; 12:1453901. [PMID: 39252788 PMCID: PMC11381397 DOI: 10.3389/fcell.2024.1453901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Specific protein 1 (Sp1) is pivotal in sustaining baseline transcription as well as modulating cell signaling pathways and transcription factors activity. Through interactions with various proteins, especially transcription factors, Sp1 controls the expression of target genes, influencing numerous biological processes. Numerous studies have confirmed Sp1's significant regulatory role in the pathogenesis of cardiovascular disorders. Post-translational modifications (PTMs) of Sp1, such as phosphorylation, ubiquitination, acetylation, glycosylation, SUMOylation, and S-sulfhydration, can enhance or modify its transcriptional activity and DNA-binding stability. These modifications also regulate Sp1 expression across different cell types. Sp1 is crucial in regulating non-coding gene expression and the activity of proteins in response to pathophysiological stimuli. Understanding Sp1 PTMs advances our knowledge of cell signaling pathways in controlling Sp1 stability during cardiovascular disease onset and progression. It also aids in identifying novel pharmaceutical targets and biomarkers essential for preventing and managing cardiovascular diseases.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Synopsis of the Golden Chamber, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhou
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Typhoid, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caiyun Mao
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Liu
- The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Guozhen Chen
- Department of Pediatrics, Yantai Yuhuangding Hospital, Shandong, China
| | - Yunjia Song
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
28
|
Lagunas-Rangel FA. Aging insights from heterochronic parabiosis models. NPJ AGING 2024; 10:38. [PMID: 39154047 PMCID: PMC11330497 DOI: 10.1038/s41514-024-00166-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Heterochronic parabiosis consists of surgically connecting the circulatory systems of a young and an old animal. This technique serves as a model to study circulating factors that accelerate aging in young organisms exposed to old blood or induce rejuvenation in old organisms exposed to young blood. Despite the promising results, the exact cellular and molecular mechanisms remain unclear, so this study aims to explore and elucidate them in more detail.
Collapse
|
29
|
Kundra S, Kaur R, Pasricha C, Kumari P, Gurjeet Singh T, Singh R. Pathological insights into activin A: Molecular underpinnings and therapeutic prospects in various diseases. Int Immunopharmacol 2024; 139:112709. [PMID: 39032467 DOI: 10.1016/j.intimp.2024.112709] [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: 02/13/2024] [Revised: 05/14/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Activin A (Act A) is a member of the TGFβ (transforming growth factor β) superfamily. It communicates via the Suppressor of Mothers against Decapentaplegic Homolog (SMAD2/3) proteins which govern processes such as cell proliferation, wound healing, apoptosis, and metabolism. Act A produces its action by attaching to activin receptor type IIA (ActRIIA) or activin receptor type IIB (ActRIIB). Increasing circulating Act A increases ActRII signalling, which on phosphorylation initiates the ALK4 (activin receptor-like kinase 4) type 1 receptor which further turns on the SMAD pathway and hinders cell functioning. Once triggered, this route leads to gene transcription, differentiation, apoptosis, and extracellular matrix (ECM) formation. Act A also governs the immunological and inflammatory responses of the body, as well as cell death. Moreover, Act A levels have been observed to elevate in several disorders like renal fibrosis, CKD, asthma, NAFLD, cardiovascular diseases, cancer, inflammatory conditions etc. Here, we provide an update on the recent studies relevant to the role of Act A in the modulation of various pathological disorders, giving an overview of the biology of Act A and its signalling pathways, and discuss the possibility of incorporating activin-A targeting as a novel therapeutic approach for the control of various disorders. Pathways such as SMAD signaling, in which SMAD moves to the nucleus by making a complex and leads to tissue fibrosis in CKD, STAT3, which drives renal fibroblast activity and the production of ECM, Kidney injury molecule (KIM-1) in the synthesis, deposition of ECM proteins, SERCA2a (sarcoplasmic reticulum Ca2+ ATPase) in cardiac dysfunction, and NF-κB (Nuclear factor kappa-light-chain-enhancer of activated B cells) in inflammation are involved in Act A signaling, have also been discussed.
Collapse
Affiliation(s)
- Sejal Kundra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rupinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Chirag Pasricha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pratima Kumari
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Ravinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
30
|
Ding J, Fayyaz AI, Ding Y, Liang D, Luo M. Role of Specificity Protein 1 (SP1) in Cardiovascular Diseases: Pathological Mechanisms and Therapeutic Potentials. Biomolecules 2024; 14:807. [PMID: 39062521 PMCID: PMC11274404 DOI: 10.3390/biom14070807] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
In mammals, specificity protein 1 (SP1) was the first Cys2-His2 zinc finger transcription factor to be isolated within the specificity protein and Krüppel-like factor (Sp/KLF) gene family. SP1 regulates gene expression by binding to Guanine-Cytosine (GC)-rich sequences on promoter regions of target genes, affecting various cellular processes. Additionally, the activity of SP1 is markedly influenced by posttranslational modifications, such as phosphorylation, acetylation, glycosylation, and proteolysis. SP1 is implicated in the regulation of apoptosis, cell hypertrophy, inflammation, oxidative stress, lipid metabolism, plaque stabilization, endothelial dysfunction, fibrosis, calcification, and other pathological processes. These processes impact the onset and progression of numerous cardiovascular disorders, including coronary heart disease, ischemia-reperfusion injury, cardiomyopathy, arrhythmia, and vascular disease. SP1 emerges as a potential target for the prevention and therapeutic intervention of cardiac ailments. In this review, we delve into the biological functions, pathophysiological mechanisms, and potential clinical implications of SP1 in cardiac pathology to offer valuable insights into the regulatory functions of SP1 in heart diseases and unveil novel avenues for the prevention and treatment of cardiovascular conditions.
Collapse
Affiliation(s)
- Jie Ding
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Aminah I. Fayyaz
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Dandan Liang
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Ming Luo
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
| |
Collapse
|
31
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [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] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
32
|
Li C, Zhang L, Zhang L, Zhang G. Correlation between elevated HCLS1 levels and heart failure: A diagnostic biomarker. Medicine (Baltimore) 2024; 103:e38484. [PMID: 38847679 PMCID: PMC11155546 DOI: 10.1097/md.0000000000038484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/16/2024] [Indexed: 06/10/2024] Open
Abstract
The correlation between hematopoietic cell-specific lyn substrate 1 (HCLS1) expression levels and heart failure (HF) remains unclear. HF datasets GSE192886 and GSE196656 profiles were generated from GPL24676 and GPL20301 platforms in gene expression omnibus (GEO) database and differentially expressed genes (DEGs) were obtained, which was followed by weighted gene co-expression network analysis, protein-protein interaction (PPI) networks, functional enrichment analysis and comparative toxicogenomics database (CTD) analysis. Heatmaps of gene expression levels were plotted. TargetScan was used to screen miRNAs regulating central DEGs. A total of 500 DEGs were found and mainly concentrated in leukocyte activation, protein phosphorylation, and protein complexes involved in cell adhesion, PI3K Akt signaling pathway, Notch signaling pathway, and right ventricular cardiomyopathy. PPI network identified 15 core genes (HCLS1, FERMT3, CD53, CD34, ITGAL, EP300, LYN, VAV1, ITGAX, LEP, ITGB1, IGF1, MMP9, SMAD2, RAC2). Heatmap shows that 4 genes (EP300, CD53, HCLS1, LYN) are highly expressed in HF tissue samples. We found that 4 genes (EP300, CD53, HCLS1, LYN) were associated with heart diseases, cardiovascular diseases, edema, rheumatoid arthritis, necrosis, and inflammation. HCLS1 is highly expressed in HF and maybe its target.
Collapse
Affiliation(s)
- Chunguang Li
- Clinical Lab Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Li Zhang
- Blood Transfusion Department, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Long Zhang
- Geriatric Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Guang Zhang
- Clinical Lab Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Lou S, Zhu W, Yu T, Zhang Q, Wang M, Jin L, Xiong Y, Xu J, Wang Q, Chen G, Liang G, Hu X, Luo W. Compound SJ-12 attenuates streptozocin-induced diabetic cardiomyopathy by stabilizing SERCA2a. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167140. [PMID: 38548092 DOI: 10.1016/j.bbadis.2024.167140] [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: 10/04/2023] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024]
Abstract
Heart failure (HF) is one of the major causes of death among diabetic patients. Although studies have shown that curcumin analog C66 can remarkably relieve diabetes-associated cardiovascular and kidney complications, the role of SJ-12, SJ-12, a novel curcumin analog, in diabetic cardiomyopathy and its molecular targets are unknown. 7-week-old male C57BL/6 mice were intraperitoneally injected with single streptozotocin (STZ) (160 mg/kg) to develop diabetic cardiomyopathy (DCM). The diabetic mice were then treated with SJ-12 via gavage for two months. Body weight, fast blood glucose, cardiac utrasonography, myocardial injury markers, pathological morphology of the heart, hypertrophic and fibrotic markers were assessed. The potential target of SJ-12 was evaluated via RNA-sequencing analysis. The O-GlcNAcylation levels of SP1 were detected via immunoprecipitation. SJ-12 effectively suppressed myocardial hypertrophy and fibrosis, thereby preventing heart dysfunction in mice with STZ-induced heart failure. RNA-sequencing analysis revealed that SJ-12 exerted its therapeutic effects through the modulation of the calcium signaling pathway. Furthermore, SJ-12 reduced the O-GlcNAcylation levels of SP1 by inhibiting O-linked N-acetylglucosamine transferase (OGT). Also, SJ-12 stabilized Sarcoplasmic/Endoplasmic Reticulum Calcium ATPase 2a (SERCA2a), a crucial regulator of calcium homeostasis, thus reducing hypertrophy and fibrosis in mouse hearts and cultured cardiomyocytes. However, the anti-fibrotic effects of SJ-12 were not detected in SERCA2a or OGT-silenced cardiomyocytes, indicating that SJ-12 can prevent DCM by targeting OGT-dependent O-GlcNAcylation of SP1.These findings indicate that SJ-12 can exert cardioprotective effects in STZ-induced mice by reducing the O-GlcNAcylation levels of SP1, thus stabilizing SERCA2a and reducing myocardial fibrosis and hypertrophy. Therefore, SJ-12 can be used for the treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shuaijie Lou
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Weiwei Zhu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Tianxiang Yu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Qianhui Zhang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Minxiu Wang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Leiming Jin
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongqiang Xiong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiachen Xu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Qinyan Wang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Gaozhi Chen
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Guang Liang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| | - Xiang Hu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Wu Luo
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
34
|
Acharya P, Parkins S, Tranter M. RNA binding proteins as mediators of pathological cardiac remodeling. Front Cell Dev Biol 2024; 12:1368097. [PMID: 38818408 PMCID: PMC11137256 DOI: 10.3389/fcell.2024.1368097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
RNA binding proteins (RBPs) play a central in the post-transcriptional regulation of gene expression, which can account for up to 50% of all variations in protein expression within a cell. Following their binding to target RNAs, RBPs most typically confer changes in gene expression through modulation of alternative spicing, RNA stabilization/degradation, or ribosome loading/translation rate. All of these post-transcriptional regulatory processes have been shown to play a functional role in pathological cardiac remodeling, and a growing body of evidence is beginning to identify the mechanistic contribution of individual RBPs and their cardiac RNA targets. This review highlights the mechanisms of RBP-dependent post-transcriptional gene regulation in cardiomyocytes and fibroblasts and our current understanding of how RNA binding proteins functionally contribute to pathological cardiac remodeling.
Collapse
Affiliation(s)
- Pooja Acharya
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sharon Parkins
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michael Tranter
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
35
|
Yin D, Liu Y, Xue B, Ding R, Wang G, Xia S, Zhang D. IL-37 Modulates Myocardial Calcium Handling via the p-STAT3/SERCA2a Axis in HF-Related Engineered Human Heart Tissue. Adv Healthc Mater 2024; 13:e2303957. [PMID: 38339835 DOI: 10.1002/adhm.202303957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Interleukin-37 (IL-37) is a potent anti-inflammatory cytokine belonging to the IL-1 family. This study investigates the regulatory mechanism and reparative effects of IL-37 on HF-related human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) and engineered human heart tissue subjected to hypoxia and H2O2 treatment. The contractile force and Ca2+ conduction capacity of the tissue are assessed using a stretching platform and high-resolution fluorescence imaging system. This investigation reveals that IL-37 treatment significantly enhances cell viability, calcium transient levels, contractile force, and Ca2+ conduction capacity in HF-related hiPSC-CMs and engineered human heart tissue. Notably, IL-37 facilitates the upregulation of sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) through enhancing nuclear p-STAT3 levels. This effect is mediated by the binding of p-STAT3 to the SERCA2a promoter, providing a novel insight on the reparative potential of IL-37 in HF. IL-37 demonstrates its ability to enhance systolic function by modulating myocardial calcium handling via the p-STAT3/SERCA2a axis in HF-related engineered human heart tissue (as shown in schematic diagram).
Collapse
Affiliation(s)
- Dan Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Yong Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Bingqing Xue
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Rui Ding
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Gang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Shutao Xia
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| |
Collapse
|
36
|
Chen P, Wu S, Hu Z, Hao B, Huang Y, Chen X, Guo Y, Wang Z, Chen X, Su M, Chen W, Zhuo Y, Li J, Wei S, Xu B, Xu J. Serum SERCA2a levels in heart failure patients are associated with adverse events after discharge. Medicine (Baltimore) 2024; 103:e37761. [PMID: 38640274 PMCID: PMC11029989 DOI: 10.1097/md.0000000000037761] [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: 08/30/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 04/21/2024] Open
Abstract
Calcium homeostasis imbalance is one of the important pathological mechanisms in heart failure. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a), a calcium ATPase on the sarcoplasmic reticulum in cardiac myocytes, is a myocardial systolic-diastolic Ca2 + homeostasis regulating enzyme that is not only involved in cardiac diastole but also indirectly affects cardiac myocyte contraction. SERCA2a expression was found to be decreased in myocardial tissue in heart failure, however, there are few reports on serum SERCA2a expression in patients with heart failure, and this study was designed to investigate whether serum SERCA2a levels are associated with the occurrence of adverse events after discharge in patients hospitalized with heart failure. Patients with heart failure hospitalized in the cardiovascular department of the Second Affiliated Hospital of Guangdong Medical University, China, from July 2018 to July 2019 were included in this study, and serum SERCA2a concentrations were measured; each enrolled patient was followed up by telephone after 6 months (6 ± 1 months) for general post-discharge patient status. The correlation between serum SERCA2a levels and the occurrence of adverse events (death or readmission due to heart failure) after hospital discharge was assessed using multiple analysis and trend analysis. Seventy-one patients with heart failure were finally included in this study, of whom 38 (53.5%) were men and 33 (46.5%) were women (All were postmenopausal women). Multiple analysis revealed no correlation between serum SERCA2a levels and the occurrence of adverse events in the total study population and in male patients, but serum SERCA2a levels were associated with the occurrence of adverse outcome events after hospital discharge in female patients (OR = 1.02, P = .047). Further analysis using a trend analysis yielded a 4.0% increase in the risk of adverse outcomes after hospital discharge for each unit increase in SERCA2a in female patients (OR = 1.04; P = .02), while no significant difference was seen in men. This study suggests that serum SERCA2a levels at admission are associated with the occurrence of post-discharge adverse events in postmenopausal female patients hospitalized with heart failure.
Collapse
Affiliation(s)
- Panghe Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shudie Wu
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhihui Hu
- Guangdong Medical University, Zhanjiang, China
| | - Biao Hao
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuesheng Huang
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xu Chen
- Guangdong Medical University, Zhanjiang, China
| | - Yingjie Guo
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhiye Wang
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoxin Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Miaoling Su
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Weiwen Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yinan Zhuo
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiahao Li
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shaofeng Wei
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bilian Xu
- Guangdong Medical University, Zhanjiang, China
| | - Jinrong Xu
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
37
|
Li Z, Chen J, Huang H, Zhan Q, Wang F, Chen Z, Lu X, Sun G. Post-translational modifications in diabetic cardiomyopathy. J Cell Mol Med 2024; 28:e18158. [PMID: 38494853 PMCID: PMC10945092 DOI: 10.1111/jcmm.18158] [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: 07/30/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 03/19/2024] Open
Abstract
The increasing attention towards diabetic cardiomyopathy as a distinctive complication of diabetes mellitus has highlighted the need for standardized diagnostic criteria and targeted treatment approaches in clinical practice. Ongoing research is gradually unravelling the pathogenesis of diabetic cardiomyopathy, with a particular emphasis on investigating various post-translational modifications. These modifications dynamically regulate protein function in response to changes in the internal and external environment, and their disturbance of homeostasis holds significant relevance for the development of chronic ailments. This review provides a comprehensive overview of the common post-translational modifications involved in the initiation and progression of diabetic cardiomyopathy, including O-GlcNAcylation, phosphorylation, methylation, acetylation and ubiquitination. Additionally, the review discusses drug development strategies for targeting key post-translational modification targets, such as agonists, inhibitors and PROTAC (proteolysis targeting chimaera) technology that targets E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Zhi Li
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Jie Chen
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Hailong Huang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Qianru Zhan
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Fengzhi Wang
- Department of Neurology, People's Hospital of Liaoning ProvincePeople's Hospital of China Medical UniversityShenyangChina
| | - Zihan Chen
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Xinwei Lu
- Department of CardiologySiping Central People's HospitalSipingChina
| | - Guozhe Sun
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
38
|
He X, Cantrell AC, Williams QA, Gu W, Chen Y, Chen JX, Zeng H. p53 Acetylation Exerts Critical Roles in Pressure Overload-Induced Coronary Microvascular Dysfunction and Heart Failure in Mice. Arterioscler Thromb Vasc Biol 2024; 44:826-842. [PMID: 38328937 PMCID: PMC10978286 DOI: 10.1161/atvbaha.123.319601] [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: 05/10/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) has been shown to contribute to cardiac hypertrophy and heart failure (HF) with preserved ejection fraction. At this point, there are no proven treatments for CMD. METHODS We have shown that histone acetylation may play a critical role in the regulation of CMD. By using a mouse model that replaces lysine with arginine at residues K98, K117, K161, and K162R of p53 (p534KR), preventing acetylation at these sites, we test the hypothesis that acetylation-deficient p534KR could improve CMD and prevent the progression of hypertensive cardiac hypertrophy and HF. Wild-type and p534KR mice were subjected to pressure overload by transverse aortic constriction to induce cardiac hypertrophy and HF. RESULTS Echocardiography measurements revealed improved cardiac function together with a reduction of apoptosis and fibrosis in p534KR mice. Importantly, myocardial capillary density and coronary flow reserve were significantly improved in p534KR mice. Moreover, p534KR upregulated the expression of cardiac glycolytic enzymes and Gluts (glucose transporters), as well as the level of fructose-2,6-biphosphate; increased PFK-1 (phosphofructokinase 1) activity; and attenuated cardiac hypertrophy. These changes were accompanied by increased expression of HIF-1α (hypoxia-inducible factor-1α) and proangiogenic growth factors. Additionally, the levels of SERCA-2 were significantly upregulated in sham p534KR mice, as well as in p534KR mice after transverse aortic constriction. In vitro, p534KR significantly improved endothelial cell glycolytic function and mitochondrial respiration and enhanced endothelial cell proliferation and angiogenesis. Similarly, acetylation-deficient p534KR significantly improved coronary flow reserve and rescued cardiac dysfunction in SIRT3 (sirtuin 3) knockout mice. CONCLUSIONS Our data reveal the importance of p53 acetylation in coronary microvascular function, cardiac function, and remodeling and may provide a promising approach to improve hypertension-induced CMD and to prevent the transition of cardiac hypertrophy to HF.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Aubrey C Cantrell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Quinesha A Williams
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Wei Gu
- Department of Pathology & Cell Biology, Columbia University, Institute for Cancer Genetics, New York, NY 10032, USA
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Jian-Xiong Chen
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Heng Zeng
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| |
Collapse
|
39
|
Le Quilliec E, LeBlanc CA, Neuilly O, Xiao J, Younes R, Altuntas Y, Xiong F, Naud P, Villeneuve L, Sirois MG, Tanguay JF, Tardif JC, Hiram R. Atrial cardiomyocytes contribute to the inflammatory status associated with atrial fibrillation in right heart disease. Europace 2024; 26:euae082. [PMID: 38546222 PMCID: PMC11000822 DOI: 10.1093/europace/euae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
AIMS Right heart disease (RHD), characterized by right ventricular (RV) and atrial (RA) hypertrophy, and cardiomyocytes' (CM) dysfunctions have been described to be associated with the incidence of atrial fibrillation (AF). Right heart disease and AF have in common, an inflammatory status, but the mechanisms relating RHD, inflammation, and AF remain unclear. We hypothesized that right heart disease generates electrophysiological and morphological remodelling affecting the CM, leading to atrial inflammation and increased AF susceptibility. METHODS AND RESULTS Pulmonary artery banding (PAB) was surgically performed (except for sham) on male Wistar rats (225-275 g) to provoke an RHD. Twenty-one days (D21) post-surgery, all rats underwent echocardiography and electrophysiological studies (EPS). Optical mapping was performed in situ, on Langendorff-perfused hearts. The contractility of freshly isolated CM was evaluated and recorded during 1 Hz pacing in vitro. Histological analyses were performed on formalin-fixed RA to assess myocardial fibrosis, connexin-43 levels, and CM morphology. Right atrial levels of selected genes and proteins were obtained by qPCR and Western blot, respectively. Pulmonary artery banding induced severe RHD identified by RV and RA hypertrophy. Pulmonary artery banding rats were significantly more susceptible to AF than sham. Compared to sham RA CM from PAB rats were significantly elongated and hypercontractile. Right atrial CM from PAB animals showed significant augmentation of mRNA and protein levels of pro-inflammatory interleukin (IL)-6 and IL1β. Sarcoplasmic-endoplasmic reticulum Ca2+-ATPase-2a (SERCA2a) and junctophilin-2 were decreased in RA CM from PAB compared to sham rats. CONCLUSIONS Right heart disease-induced arrhythmogenicity may occur due to dysfunctional SERCA2a and inflammatory signalling generated from injured RA CM, which leads to an increased risk of AF.
Collapse
Affiliation(s)
- Ewen Le Quilliec
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Charles-Alexandre LeBlanc
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Orlane Neuilly
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Jiening Xiao
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Rim Younes
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Yasemin Altuntas
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Feng Xiong
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Patrice Naud
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Louis Villeneuve
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Martin G Sirois
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Jean-François Tanguay
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| | - Roddy Hiram
- Department of Medicine, Montreal Heart Institute, University of Montreal, 5000 Belanger Street, Montreal, QC HIT 1C8, Canada
| |
Collapse
|
40
|
Zhan X, Yang Y, Li Q, He F. The role of deubiquitinases in cardiac disease. Expert Rev Mol Med 2024; 26:e3. [PMID: 38525836 PMCID: PMC11062144 DOI: 10.1017/erm.2024.2] [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: 08/08/2023] [Revised: 12/10/2023] [Accepted: 12/28/2023] [Indexed: 03/26/2024]
Abstract
Deubiquitinases are a group of proteins that identify and digest monoubiquitin chains or polyubiquitin chains attached to substrate proteins, preventing the substrate protein from being degraded by the ubiquitin-proteasome system. Deubiquitinases regulate cellular autophagy, metabolism and oxidative stress by acting on different substrate proteins. Recent studies have revealed that deubiquitinases act as a critical regulator in various cardiac diseases, and control the onset and progression of cardiac disease through a board range of mechanism. This review summarizes the function of different deubiquitinases in cardiac disease, including cardiac hypertrophy, myocardial infarction and diabetes mellitus-related cardiac disease. Besides, this review briefly recapitulates the role of deubiquitinases modulators in cardiac disease, providing the potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Xiaona Zhan
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Qing Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Fan He
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
41
|
Wang X, Yang J, Lu C, Hu Y, Xu Z, Wan Q, Zhang M, Shi T, Liu Z, Liu Y. Qifu Yixin Formula Improves Heart Failure by Enhancing β-Arrestin2 Mediated the SUMOylation of SERCA2a. Drug Des Devel Ther 2024; 18:781-799. [PMID: 38500692 PMCID: PMC10946281 DOI: 10.2147/dddt.s446324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Purpose This study aimed to elucidate the protective mechanism of Traditional Chinese Medicine (TCM) Qifu Yixin formula (QFYXF) to improve heart failure (HF) by promoting β-arrestin2 (β-arr2)-mediated SERCA2a SUMOylation. Materials and Methods The transverse aortic constriction (TAC)-induced HF mice were treated with QFYXF or carvedilol for 8 weeks. β-arr2-KO mice and their littermate wild-type (WT) mice were used as controls. Neonatal rat cardiomyocytes (NRCMs) were used in vitro. Cardiac function was evaluated by echocardiography and serum NT-proBNP. Myocardial hypertrophy and myocardial fibrosis were assessed by histological staining. β-arr2, SERCA2a, SUMO1, PLB and p-PLB expressions were detected by Western blotting, immunofluorescence and immunohistochemistry. SERCA2a SUMOylation was detected by Co-IP. The molecular docking method was used to predict the binding ability of the main active components of QFYXF to β-arr2, SERCA2a, and SUMO1, and the binding degree of SERCA2a to SUMO1 protein. Results The HF model was constructed 8 weeks after TAC. QFYXF ameliorated cardiac function, inhibiting myocardial hypertrophy and fibrosis. QFYXF promoted SERCA2a expression and SERCA2a SUMOylation. Further investigation showed that QFYXF promoted β-arr2 expression, whereas Barbadin (β-arr2 inhibitor) or β-arr2-KO reduced SERCA2a SUMOylation and attenuated the protective effect of QFYXF improved HF. Molecular docking showed that the main active components of QFYXF had good binding activities with β-arr2, SERCA2a, and SUMO1, and SERCA2a had a high binding degree with SUMO1 protein. Conclusion QFYXF improves HF by promoting β-arr2 mediated SERCA2a SUMOylation and increasing SERCA2a expression.
Collapse
Affiliation(s)
- Xinting Wang
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Jiahui Yang
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Cheng Lu
- Department of Cardiology, Seventh People’s Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, People’s Republic of China
| | - Yinqin Hu
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Zhaohui Xu
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Qiqi Wan
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Meng Zhang
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Tianyun Shi
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Zhirui Liu
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| | - Yongming Liu
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, People’s Republic of China
| |
Collapse
|
42
|
Fairuz S, Ang CW, Mraiche F, Goh JK. Current Targets and Future Directions of Positive Inotropes for Heart Failure. Curr Med Chem 2024; 31:6971-6991. [PMID: 37909442 DOI: 10.2174/0109298673262360231018193823] [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: 05/23/2023] [Revised: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/03/2023]
Abstract
While a congestive heart failure patient will ultimately need an assist device or even a replacement heart as the disease progresses, not every patient is qualified for such advanced therapy. Such patients awaiting better circulatory support benefit from positive inotropes in the meantime as palliative care. These agents are often prescribed in patients with acute decompensated heart failure, with reduced left ventricular ejection fraction and symptoms of organ dysfunction. Although positive inotropes, for example, digoxin, dobutamine, milrinone, levosimendan, etc., are successfully marketed and in use, a lot of their adverse effects, like arrhythmias, hypotension, and even sudden cardiac death, are rather encouraging further research on the development of novel positive inotropes. This review has investigated the molecular mechanisms of some of these adverse effects in terms of the proteins they target, followed by research on newer targets. Studies from 2013-2023 that have reported new small molecules with positive inotropic effects have been revisited in order to determine the progress made so far in drug discovery.
Collapse
Affiliation(s)
- Shadreen Fairuz
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Chee Wei Ang
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Fatima Mraiche
- Department of Pharmacology, University of Alberta, 116 St & 85 Ave, Edmonton, ABT6G 2R3, Canada
| | - Joo Kheng Goh
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
43
|
Chiorescu RM, Lazar RD, Ruda A, Buda AP, Chiorescu S, Mocan M, Blendea D. Current Insights and Future Directions in the Treatment of Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2023; 25:440. [PMID: 38203612 PMCID: PMC10778923 DOI: 10.3390/ijms25010440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Heart failure is a clinical syndrome associated with poor quality of life, substantial healthcare resource utilization, and premature mortality, in large part related to high rates of hospitalizations. The clinical manifestations of heart failure are similar regardless of the ejection fraction. Unlike heart failure with reduced ejection fraction, there are few therapeutic options for treating heart failure with preserved ejection fraction. Molecular therapies that have shown reduced mortality and morbidity in heart failure with reduced ejection have not been proven to be effective for patients with heart failure and preserved ejection fraction. The study of pathophysiological processes involved in the production of heart failure with preserved ejection fraction is the basis for identifying new therapeutic means. In this narrative review, we intend to synthesize the existing therapeutic means, but also those under research (metabolic and microRNA therapy) for the treatment of heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Roxana-Daiana Lazar
- Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania; (A.R.); (A.P.B.); (D.B.)
| | - Alexandru Ruda
- Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania; (A.R.); (A.P.B.); (D.B.)
| | - Andreea Paula Buda
- Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania; (A.R.); (A.P.B.); (D.B.)
| | - Stefan Chiorescu
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania;
| | - Mihaela Mocan
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Dan Blendea
- Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania; (A.R.); (A.P.B.); (D.B.)
- Department of Cardiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| |
Collapse
|
44
|
Ramos TAR, Urquiza-Zurich S, Kim SY, Gillette TG, Hill JA, Lavandero S, do Rêgo TG, Maracaja-Coutinho V. Single-cell transcriptional landscape of long non-coding RNAs orchestrating mouse heart development. Cell Death Dis 2023; 14:841. [PMID: 38110334 PMCID: PMC10728149 DOI: 10.1038/s41419-023-06296-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 10/18/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
Long non-coding RNAs (lncRNAs) comprise the most representative transcriptional units of the mammalian genome. They are associated with organ development linked with the emergence of cardiovascular diseases. We used bioinformatic approaches, machine learning algorithms, systems biology analyses, and statistical techniques to define co-expression modules linked to heart development and cardiovascular diseases. We also uncovered differentially expressed transcripts in subpopulations of cardiomyocytes. Finally, from this work, we were able to identify eight cardiac cell-types; several new coding, lncRNA, and pcRNA markers; two cardiomyocyte subpopulations at four different time points (ventricle E9.5, left ventricle E11.5, right ventricle E14.5 and left atrium P0) that harbored co-expressed gene modules enriched in mitochondrial, heart development and cardiovascular diseases. Our results evidence the role of particular lncRNAs in heart development and highlight the usage of co-expression modular approaches in the cell-type functional definition.
Collapse
Grants
- R01 HL155765 NHLBI NIH HHS
- R01 HL126012 NHLBI NIH HHS
- R01 HL147933 NHLBI NIH HHS
- R01 HL128215 NHLBI NIH HHS
- R01 HL120732 NHLBI NIH HHS
- Agencia Nacional de Investigacion y Desarrollo (ANID, Chile), FONDAP 15130011 (SL), FONDECYT 1200490 (SL)
- the NIH: HL-120732 (JAH), HL-128215 (JAH), HL-126012 (JAH), HL-147933, (JAH), HL-155765 (JAH), 14SFRN20510023 (JAH), 14SFRN20670003 (JAH), Leducq grant number 11CVD04 (JAH), Cancer Prevention and Research Institute of Texas grant RP110486P3 (JAH)
- Agencia Nacional de Investigacion y Desarrollo (ANID, Chile), FONDAP 15130011 (VMC) and FONDECYT 1211731 (VMC).
Collapse
Affiliation(s)
- Thaís A R Ramos
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil
- Departamento de Informática, Centro de Informática, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Sebastián Urquiza-Zurich
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago, Chile.
| | - Thaís G do Rêgo
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil.
- Departamento de Informática, Centro de Informática, Universidade Federal da Paraíba, João Pessoa, Brazil.
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil.
| |
Collapse
|
45
|
Sahgal A, Uversky V, Davé V. Microproteins transitioning into a new Phase: Defining the undefined. Methods 2023; 220:38-54. [PMID: 37890707 DOI: 10.1016/j.ymeth.2023.10.009] [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: 09/01/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Recent advancements in omics technologies have unveiled a hitherto unknown group of short polypeptides called microproteins (miPs). Despite their size, accumulating evidence has demonstrated that miPs exert varied and potent biological functions. They act in paracrine, juxtracrine, and endocrine fashion, maintaining cellular physiology and driving diseases. The present study focuses on biochemical and biophysical analysis and characterization of twenty-four human miPs using distinct computational methods, including RIDAO, AlphaFold2, D2P2, FuzDrop, STRING, and Emboss Pep wheel. miPs often lack well-defined tertiary structures and may harbor intrinsically disordered regions (IDRs) that play pivotal roles in cellular functions. Our analyses define the physicochemical properties of an essential subset of miPs, elucidating their structural characteristics and demonstrating their propensity for driving or participating in liquid-liquid phase separation (LLPS) and intracellular condensate formation. Notably, miPs such as NoBody and pTUNAR revealed a high propensity for LLPS, implicating their potential involvement in forming membrane-less organelles (MLOs) during intracellular LLPS and condensate formation. The results of our study indicate that miPs have functionally profound implications in cellular compartmentalization and signaling processes essential for regulating normal cellular functions. Taken together, our methodological approach explains and highlights the biological importance of these miPs, providing a deeper understanding of the unusual structural landscape and functionality of these newly defined small proteins. Understanding their functions and biological behavior will aid in developing targeted therapies for diseases that involve miPs.
Collapse
Affiliation(s)
- Aayushi Sahgal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Biotechnology Graduate Program, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Vladimir Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Vrushank Davé
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Biotechnology Graduate Program, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States.
| |
Collapse
|
46
|
Wang Y, Liu Z, Bian X, Zhao C, Zhang X, Liu X, Wang N. Function and regulation of ubiquitin-like SUMO system in heart. Front Cell Dev Biol 2023; 11:1294717. [PMID: 38033852 PMCID: PMC10687153 DOI: 10.3389/fcell.2023.1294717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
The small ubiquitin-related modifier (SUMOylation) system is a conserved, reversible, post-translational protein modification pathway covalently attached to the lysine residues of proteins in eukaryotic cells, and SUMOylation is catalyzed by SUMO-specific activating enzyme (E1), binding enzyme (E2) and ligase (E3). Sentrin-specific proteases (SENPs) can cleave the isopeptide bond of a SUMO conjugate and catalyze the deSUMOylation reaction. SUMOylation can regulate the activity of proteins in many important cellular processes, including transcriptional regulation, cell cycle progression, signal transduction, DNA damage repair and protein stability. Biological experiments in vivo and in vitro have confirmed the key role of the SUMO conjugation/deconjugation system in energy metabolism, Ca2+ cycle homeostasis and protein quality control in cardiomyocytes. In this review, we summarized the research progress of the SUMO conjugation/deconjugation system and SUMOylation-mediated cardiac actions based on related studies published in recent years, and highlighted the further research areas to clarify the role of the SUMO system in the heart by using emerging technologies.
Collapse
Affiliation(s)
- Ying Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiyun Bian
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Chenxu Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xin Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaozhi Liu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
47
|
Luo Y, Zhou S, Xu T, Wu W, Shang P, Wang S, Pan D, Li D. SENP2-mediated SERCA2a deSUMOylation increases calcium overload in cardiomyocytes to aggravate myocardial ischemia/reperfusion injury. Chin Med J (Engl) 2023; 136:2496-2507. [PMID: 37462038 PMCID: PMC10586866 DOI: 10.1097/cm9.0000000000002757] [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: 10/01/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) is a key protein that maintains myocardial Ca 2+ homeostasis. The present study aimed to investigate the mechanism underlying the SERCA2a-SUMOylation (small ubiquitin-like modifier) process after ischemia/reperfusion injury (I/RI) in vitro and in vivo . METHODS Calcium transient and systolic/diastolic function of cardiomyocytes isolated from Serca2a knockout (KO) and wild-type mice with I/RI were compared. SUMO-relevant protein expression and localization were detected by quantitative real-time PCR (RT-qPCR), Western blotting, and immunofluorescence in vitro and in vivo . Serca2a-SUMOylation, infarct size, and cardiac function of Senp1 or Senp2 overexpressed/suppressed adenovirus infected cardiomyocytes, were detected by immunoprecipitation, triphenyltetrazolium chloride (TTC)-Evans blue staining, and echocardiography respectively. RESULTS The results showed that the changes of Fura-2 fluorescence intensity and contraction amplitude of cardiomyocytes decreased in the I/RI groups and were further reduced in the Serca2a KO + I/RI groups. Senp1 and Senp2 messenger ribose nucleic acid (mRNA) and protein expression levels in vivo and in cardiomyocytes were highest at 6 h and declined at 12 h after I/RI. However, the highest levels in HL-1 cells were recorded at 12 h. Senp2 expression increased in the cytoplasm, unlike that of Senp1. Inhibition of Senp2 protein reversed the I/RI-induced Serca2a-SUMOylation decline, reduced the infarction area, and improved cardiac function, while inhibition of Senp1 protein could not restore the above indicators. CONCLUSION I/RI activated Senp1 and Senp2 protein expression, which promoted Serca2a-deSUMOylation, while inhibition of Senp2 expression reversed Serca2a-SUMOylation and improved cardiac function.
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, China
| | - Shuaishuai Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Tao Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, China
| | - Pingping Shang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Shuai Wang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, China
| | - Dongye Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
48
|
Yang Q, Gao J, Wang TY, Ding JC, Hu PF. Integration of RNA-Seq and Machine Learning Identifies Hub Genes for Empagliflozin Benefitable Heart Failure with Reduced Ejection Fraction. J Inflamm Res 2023; 16:4733-4749. [PMID: 37872956 PMCID: PMC10590560 DOI: 10.2147/jir.s429096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023] Open
Abstract
Purpose This study aimed to analyze the hub genes of heart failure with reduced ejection fraction (HFrEF) treated with Empagliflozin using RNA sequencing (RNA-seq) and bioinformatics methods, including machine learning. Methods From February 2021 to February 2023, nine patients with HFrEF were enrolled from our hospital's cardiovascular department. In addition to routine drug treatment, these patients received 10 mg of Empagliflozin once daily for two months. Efficacy was assessed and RNA-seq was performed on peripheral blood before and after treatment with empagliflozin. HFrEF-related hub genes were identified through bioinformatics analyses including differential gene expression analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, immune infiltration analysis, machine learning, immune cell correlation analysis and clinical indicator correlation analysis. Results The nine patients included in this study completed a two-month treatment regimen, with an average age of 62.11 ± 6.36 years. By performing bioinformatics analysis on the transcriptome from the treatment groups, 42 differentially expressed genes were identified, with six being up-regulated and 36 being down-regulated (|log2FC|>1 and adj.pvalue<0.05). Immune infiltration analysis of these genes demonstrated a significant difference in the proportion of plasma between the pre-treatment and post-treatment groups (p<0.05). Two hub genes, GTF2IP14 and MTLN, were finally identified through machine learning. Further analysis of the correlation between the hub genes and immune cells suggested a negative correlation between GTF2IP14 and naive B cells, and a positive correlation between MTLN and regulatory T cells and resting memory CD4+ T cells (p<0.05). Conclusion Through RNA-seq and bioinformatics analysis, this study identified GTF2IP14 and MTLN as the hub genes of HFrEF, and their mechanisms may be related to immune inflammatory responses and various immune cells.
Collapse
Affiliation(s)
- Qiang Yang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, Zhejiang Province, 310018, People’s Republic of China
| | - Tian-Yu Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Jun-Can Ding
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Peng-Fei Hu
- Department of Cardiology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310005, People’s Republic of China
| |
Collapse
|
49
|
Rabkin SW, Wong CN. Epigenetics in Heart Failure: Role of DNA Methylation in Potential Pathways Leading to Heart Failure with Preserved Ejection Fraction. Biomedicines 2023; 11:2815. [PMID: 37893188 PMCID: PMC10604152 DOI: 10.3390/biomedicines11102815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
This review will focus on epigenetic modifications utilizing the DNA methylation mechanism, which is potentially involved in the pathogenesis of heart failure with preserved ejection fraction (HFpEF). The putative pathways of HFpEF will be discussed, specifically myocardial fibrosis, myocardial inflammation, sarcoplasmic reticulum Ca2+-ATPase, oxidative-nitrosative stress, mitochondrial and metabolic defects, as well as obesity. The relationship of HFpEF to aging and atrial fibrillation will be examined from the perspective of DNA methylation.
Collapse
Affiliation(s)
- Simon W. Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Division of Cardiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Chenille N. Wong
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
50
|
Ahmed F, Kahlon T, Mohamed TMA, Ghafghazi S, Settles D. Literature Review: Pathophysiology of Heart Failure with Preserved Ejection Fraction. Curr Probl Cardiol 2023; 48:101745. [PMID: 37087081 DOI: 10.1016/j.cpcardiol.2023.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
Heart failure with preserved ejection fraction is a growing public health concern, a disease with poor health outcomes, and is showing increased prevalence globally. This review paper explores the literature with a focus on the pathophysiology and microbiology of preserved ejection fraction heart failure while drawing connections between preserved and reduced ejection fraction states. The discussion teases out the cellular level changes that affect the overall dysfunction of the cardiac tissue, including the clinical manifestations, microbiological changes (endothelial cells, fibroblasts, cardiomyocytes, and excitation-contraction coupling), and the burden of structural diastolic dysfunction. The goal of this review is to summarize the pathophysiological disease state of heart failure with preserved ejection fraction to enhance understanding, knowledge, current treatment models of this pathology.
Collapse
Affiliation(s)
- Faizan Ahmed
- Department of Anesthesiology, University of Louisville School of Medicine, Louisville, Kentucky, USA.
| | - Tani Kahlon
- Department of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tamer M A Mohamed
- Department of Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shahab Ghafghazi
- Department of Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Dana Settles
- Department of Cardiothoracic Anesthesia, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|