1
|
Zaccolo M, Kovanich D. Nanodomain cAMP signaling in cardiac pathophysiology: potential for developing targeted therapeutic interventions. Physiol Rev 2025; 105:541-591. [PMID: 39115424 PMCID: PMC7617275 DOI: 10.1152/physrev.00013.2024] [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: 03/29/2024] [Revised: 07/22/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
The 3',5'-cyclic adenosine monophosphate (cAMP) mediates the effects of sympathetic stimulation on the rate and strength of cardiac contraction. Beyond this pivotal role, in cardiac myocytes cAMP also orchestrates a diverse array of reactions to various stimuli. To ensure specificity of response, the cAMP signaling pathway is intricately organized into multiple, spatially confined, subcellular domains, each governing a distinct cellular function. In this review, we describe the molecular components of the cAMP signaling pathway with a specific focus on adenylyl cyclases, A-kinase anchoring proteins, and phosphodiesterases. We discuss how they are organized inside the intracellular space and how they achieve exquisite regulation of signaling within nanometer-size domains. We delineate the key experimental findings that lead to the current model of compartmentalized cAMP signaling, and we offer an overview of our present understanding of how cAMP nanodomains are structured and regulated within cardiac myocytes. Furthermore, we discuss how compartmentalized cAMP signaling is affected in cardiac disease and consider the potential therapeutic opportunities arising from understanding such organization. By exploiting the nuances of compartmentalized cAMP signaling, novel and more effective therapeutic strategies for managing cardiac conditions may emerge. Finally, we highlight the unresolved questions and hurdles that must be addressed to translate these insights into interventions that may benefit patients.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Duangnapa Kovanich
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
2
|
Verçosa BLA, Muniz-Junqueira MI, Mineiro ALBB, Melo MN, Vasconcelos AC. Enhanced apoptosis and inflammation allied with autophagic and apoptotic Leishmania amastigotes in the seemingly undamaged ear skin of clinically affected dogs with canine visceral Leishmaniasis. Cell Immunol 2025; 408:104909. [PMID: 39701006 DOI: 10.1016/j.cellimm.2024.104909] [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: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Programmed cell death plays a relevant role in the pathogenesis of visceral Leishmaniasis. Apoptosis selects suitable parasites, regulating parasite density, whereas autophagy eliminates pathogens. This study aimed to assess the inflammation and apoptosis in inflammatory cells and presents a unique description of the presence of autophagic and apoptotic Leishmania amastigotes in naturally Leishmania-infected dogs. Fragments from seemingly undamaged ear skin of sixteen Leishmania-infected dogs and seven uninfected dogs were evaluated through histomorphometry, ultrastructural, immunohistochemical and transmission electron microscopy (TEM) analyses. Leishmania amastigotes were present on seemingly undamaged ear skin only in clinically affected dogs. Parasite load, morphometrical parameters of inflammation and apoptotic index of inflammatory cells were higher in clinically affected animals and were related to clinical manifestations. Apoptotic index and morphometric parameters of the inflammatory infiltrate in undamaged ear skin were positively correlated with parasite load. Apoptotic and non-apoptotic Leishmania amastigotes were observed within neutrophils and macrophages. Leishmania amastigotes were positive for Bax, a marker for apoptosis, by immunohistochemistry. Morphological characteristics of apoptosis and autophagy in Leishmania amastigotes were observed only in phagocytes of clinically affected dogs. Positive correlations were found between histomorphometry and clinical manifestations. Our results showed that apoptosis and autophagy in Leishmania amastigotes may be related to both the increase in parasite load and apoptotic index in inflammatory cells, and with the intensity of the inflammatory response in clinically affected dogs. Thus, our study suggests that apoptotic and autophagy Leishmania within phagocytes may have facilitate the survival of the parasite and it appears to play an important role in the process of Leishmania infection.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil; Faculdade de Ciências da Saúde Pitágoras, Campus Codó, Codó, Maranhão, Brazil.
| | | | - Ana Lys Bezerra Barradas Mineiro
- Departamento de Clínica e Cirurgia Veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Jiang T, Jin X, Gao Y, Zhou W, Yu J, Li Y, Xu J, Cai B. CardioAtlas: deciphering the single-cell transcriptome landscape in cardiovascular tissues and diseases. Biomark Res 2024; 12:149. [PMID: 39609860 PMCID: PMC11606023 DOI: 10.1186/s40364-024-00696-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Increasing scRNA-seq data in cardiovascular research have substantially improved our knowledge on the development of the cardiovascular system and the mechanisms underlying cardiovascular diseases. However, the single-cell transcriptome datasets were dispersed in literature and no resource for cardiovascular systems and diseases. Here, we constructed an organized resource CardioAtlas, which provides comprehensive analysis results for > 1,929,000 cells in 27 human data sets and > 1,088,000 cells in 39 mouse data sets. Through large-scale literature retrieval and manual annotation, we constructed 12 and 15 scRNA-seq reference atlas for common human and mouse cardiovascular systems and diseases, covering 43 and 39 cell types. In particular, CardioAtlas provides five analytic modules, including cell-type prediction, identification of marker genes, functional enrichment analysis, identification of cell-type-specific transcription regulons, and cell-cell communication analysis. In addition, users can upload scRNA-seq data for personalized analysis. CardioAtlas is available at http://bio-bigdata.hrbmu.edu.cn/CardioAtlas . CardioAtlas provides the first comprehensive and well-crafted reference atlas of cardiovascular systems and diseases and describes in detail previously unrecognized cell populations across a large number of humans and mice.
Collapse
Affiliation(s)
- Tiantongfei Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaoyan Jin
- Department of Pharmacy, The Second Affiliated Hospital, Department of Pharmacology, College of Pharmacy, (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Yueying Gao
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Weiwei Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jinyang Yu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation, College of Biomedical Information and Engineering, Hainan Medical University, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, 571199, China
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China.
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation, College of Biomedical Information and Engineering, Hainan Medical University, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, 571199, China.
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Benzhi Cai
- Department of Pharmacy, The Second Affiliated Hospital, Department of Pharmacology, College of Pharmacy, (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
4
|
Liu Q, Li J, Li Y, Cheng M, Zhang H, Ma B. Estrogen Regulates Ca 2+ to Promote Mitochondrial Function Through G-Protein-Coupled Estrogen Receptors During Oocyte Maturation. Biomolecules 2024; 14:1430. [PMID: 39595606 PMCID: PMC11591592 DOI: 10.3390/biom14111430] [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: 09/04/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Estrogen is a steroid hormone that plays a key role in regulating many physiological processes, such as follicle activation and development and oocyte maturation in mammals. Ca2+ is crucial in oogenesis, oocyte maturation, ovulation, and fertilization. However, the mechanism by which estrogen regulates Ca2+ during oocyte maturation in mice has not been reported. This study revealed that Ca2+ levels in oocytes significantly increase during the 4-12 h period in vitro. Oocytes treated with 0.1 µM estrogen and 1 µM G1, a G-protein-coupled estrogen receptor (GPER) agonist, showed significantly increased Ca2+ levels, while treatment with 1 µM G15, an antagonist of GPER, significantly decreased Ca2+ levels. Notably, estrogen regulates Ca2+ in oocytes through the GPER pathway and promotes the expression of the Ca2+-producing protein EPAC1. In addition, estrogen alleviates the inhibitory effect of the Ca2+ chelator BAPTA-AM during oocyte maturation by promoting Ca2+ production. Furthermore, estrogen can promote the expression of the mitochondrial generation-associated protein SIRT1 through the GPER pathway, alleviate mitochondrial oxidative damage caused by BAPTA-AM, and restore the mitochondrial membrane potential level. Collectively, this study demonstrates that estrogen can regulate Ca2+ through the GPER-EPAC1 pathway and promote the expression of SIRT1, which promotes oocyte mitochondrial function during oocyte maturation.
Collapse
Affiliation(s)
- Qingyang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Jingmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Yanxue Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Ming Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| |
Collapse
|
5
|
Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: focus on mitochondrial dysfunction. Angiogenesis 2024; 27:623-639. [PMID: 39060773 PMCID: PMC11564294 DOI: 10.1007/s10456-024-09938-4] [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: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
As a vital component of blood vessels, endothelial cells play a key role in maintaining overall physiological function by residing between circulating blood and semi-solid tissue. Various stress stimuli can induce endothelial injury, leading to the onset of corresponding diseases in the body. In recent years, the importance of mitochondria in vascular endothelial injury has become increasingly apparent. Mitochondria, as the primary site of cellular aerobic respiration and the organelle for "energy information transfer," can detect endothelial cell damage by integrating and receiving various external stress signals. The generation of reactive oxygen species (ROS) and mitochondrial dysfunction often determine the evolution of endothelial cell injury towards necrosis or apoptosis. Therefore, mitochondria are closely associated with endothelial cell function, helping to determine the progression of clinical diseases. This article comprehensively reviews the interconnection and pathogenesis of mitochondrial-induced vascular endothelial cell injury in cardiovascular diseases, renal diseases, pulmonary-related diseases, cerebrovascular diseases, and microvascular diseases associated with diabetes. Corresponding therapeutic approaches are also provided. Additionally, strategies for using clinical drugs to treat vascular endothelial injury-based diseases are discussed, aiming to offer new insights and treatment options for the clinical diagnosis of related vascular injuries.
Collapse
Affiliation(s)
- Boxian Pang
- Beijing University of Chinese Medicine, Beijing, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | | | - Tieliang Pang
- Beijing Anding hospital, Capital Medical University, Beijing, China
| | - Xinyao Sun
- Beijing University of Chinese Medicine, Beijing, China
| | - Xin Liu
- Bioscience Department, University of Nottingham, Nottingham, UK
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiagge, Xicheng District, Beijing, China.
| |
Collapse
|
6
|
Wan H, Yang X, Zhang Y, Liu X, Li Y, Qin Y, Yan H, Gui L, Li K, Zhang L, Yang L, Zhang B, Wang Y. Polyphenol-Reinforced Glycocalyx-Like Hydrogel Coating Induced Myocardial Regeneration and Immunomodulation. ACS NANO 2024; 18:21512-21522. [PMID: 39096486 DOI: 10.1021/acsnano.4c06332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Although minimally invasive interventional occluders can effectively seal heart defect tissue, they still have some limitations, including poor endothelial healing, intense inflammatory response, and thrombosis formation. Herein, a polyphenol-reinforced medicine/peptide glycocalyx-like coating was prepared on cardiac occluders. A coating consisting of carboxylated chitosan, epigallocatechin-3-gallate (EGCG), tanshinone IIA sulfonic sodium (TSS), and hyaluronic acid grafted with 3-aminophenylboronic acid was prepared. Subsequently, the mercaptopropionic acid-GGGGG-Arg-Glu-Asp-Val peptide was grafted by the thiol-ene "click" reaction. The coating showed good hydrophilicity and free radical-scavenging ability and could release EGCG-TSS. The results of biological experiments suggested that the coating could reduce thrombosis by promoting endothelialization, and promote myocardial repair by regulating the inflammatory response. The functions of regulating cardiomyocyte apoptosis and metabolism were confirmed, and the inflammatory regulatory functions of the coating were mainly dependent on the NF-kappa B and TNF signaling pathway.
Collapse
Affiliation(s)
- Huining Wan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaohui Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yutong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiyu Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yanyan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hui Yan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Lan Gui
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ke Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Longjian Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
- Bioengineering Department, University of California, Los Angeles, California 90095, United States
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
7
|
Lecchi G, Mocchetti C, Tunesi D, Berto A, Balasubramanian HB, Biswas S, Bagchi A, Pollastro F, Fresu LG, Talmon M. Single-Nucleotide Polymorphisms of TAS2R46 Affect the Receptor Downstream Calcium Regulation in Histamine-Challenged Cells. Cells 2024; 13:1204. [PMID: 39056786 PMCID: PMC11275237 DOI: 10.3390/cells13141204] [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: 05/08/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Bitter taste receptors (TAS2Rs) expressed in extraoral tissues represent a whole-body sensory system, whose role and mechanisms could be of interest for the identification of new therapeutic targets. It is known that TAS2R46s in pre-contracted airway smooth muscle cells increase mitochondrial calcium uptake, leading to bronchodilation, and that several SNPs have been identified in its gene sequence. There are very few reports on the structure-function analysis of TAS2Rs. Thus, we delved into the subject by using mutagenesis and in silico studies. We generated a cellular model that expresses native TAS2R46 to evaluate the influence of the four most common SNPs on calcium fluxes following the activation of the receptor by its specific ligand absinthin. Then, docking studies were conducted to correlate the calcium flux results to the structural mutation. The analysed SNPs differently modulate the TAS2R46 signal cascade according to the altered protein domain. In particular, the SNP in the sixth transmembrane domain of the receptors did not modulate calcium homeostasis, while the SNPs in the sequence coding for the fourth transmembrane domain completely abolished the mitochondrial calcium uptake. In conclusion, these results indicate the fourth transmembrane domain of TAS2R46 is critical for the intrinsic receptor activity.
Collapse
Affiliation(s)
- Giulia Lecchi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Chiara Mocchetti
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Davide Tunesi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Arianna Berto
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Hari Baskar Balasubramanian
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Luigia Grazia Fresu
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy
| | - Maria Talmon
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| |
Collapse
|
8
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
9
|
Zhang W, Zhang M, Sun M, Hu M, Yu M, Sun J, Zhang X, Du B. Metabolomics-transcriptomics joint analysis: unveiling the dysregulated cell death network and developing a diagnostic model for high-grade neuroblastoma. Front Immunol 2024; 14:1345734. [PMID: 38239355 PMCID: PMC10794662 DOI: 10.3389/fimmu.2023.1345734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024] Open
Abstract
High-grade neuroblastoma (HG-NB) exhibits a significantly diminished survival rate in comparison to low-grade neuroblastoma (LG-NB), primarily attributed to the mechanism of HG-NB is unclear and the lacking effective therapeutic targets and diagnostic model. Therefore, the current investigation aims to study the dysregulated network between HG-NB and LG-NB based on transcriptomics and metabolomics joint analysis. Meanwhile, a risk diagnostic model to distinguish HG-NB and LG-NB was also developed. Metabolomics analysis was conducted using plasma samples obtained from 48 HG-NB patients and 36 LG-NB patients. A total of 39 metabolites exhibited alterations, with 20 showing an increase and 19 displaying a decrease in HG-NB. Additionally, transcriptomics analysis was performed on NB tissue samples collected from 31 HG-NB patients and 20 LG-NB patients. Results showed that a significant alteration was observed in a total of 1,199 mRNAs in HG-NB, among which 893 were upregulated while the remaining 306 were downregulated. In particular, the joint analysis of both omics data revealed three aberrant pathways, namely the cAMP signaling pathway, PI3K-Akt signaling pathway, and TNF signaling pathway, which were found to be associated with cell death. Notably, a diagnostic model for HG-NB risk classification was developed based on the genes MGST1, SERPINE1, and ERBB3 with an area under the receiver operating characteristic curve of 0.915. In the validation set, the sensitivity and specificity were determined to be 75.0% and 80.0%, respectively.
Collapse
Affiliation(s)
- Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Mengxin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Meng Sun
- Henan Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Minghui Hu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Muchun Yu
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jushan Sun
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xianwei Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Bang Du
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Talmon M, Massara E, Quaregna M, De Battisti M, Boccafoschi F, Lecchi G, Puppo F, Bettega Cajandab MA, Salamone S, Bovio E, Boldorini R, Riva B, Pollastro F, Fresu LG. Bitter taste receptor (TAS2R) 46 in human skeletal muscle: expression and activity. Front Pharmacol 2023; 14:1205651. [PMID: 37771728 PMCID: PMC10522851 DOI: 10.3389/fphar.2023.1205651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/27/2023] [Indexed: 09/30/2023] Open
Abstract
Bitter taste receptors are involved not only in taste perception but in various physiological functions as their anatomical location is not restricted to the gustatory system. We previously demonstrated expression and activity of the subtype hTAS2R46 in human airway smooth muscle and broncho-epithelial cells, and here we show its expression and functionality in human skeletal muscle cells. Three different cellular models were used: micro-dissected human skeletal tissues, human myoblasts/myotubes and human skeletal muscle cells differentiated from urine stem cells of healthy donors. We used qPCR, immunohistochemistry and immunofluorescence analysis to evaluate gene and protein hTAS2R46 expression. In order to explore receptor activity, cells were incubated with the specific bitter ligands absinthin and 3ß-hydroxydihydrocostunolide, and calcium oscillation and relaxation were evaluated by calcium imaging and collagen assay, respectively, after a cholinergic stimulus. We show, for the first time, experimentally the presence and functionality of a type 2 bitter receptor in human skeletal muscle cells. Given the tendentially protective role of the bitter receptors starting from the oral cavity and following also in the other ectopic sites, and given its expression already at the myoblast level, we hypothesize that the bitter receptor can play an important role in the development, maintenance and in the protection of muscle tissue functions.
Collapse
Affiliation(s)
- Maria Talmon
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Erika Massara
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Martina Quaregna
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Marta De Battisti
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Francesca Boccafoschi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Giulia Lecchi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Federico Puppo
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | | | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Enrica Bovio
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Renzo Boldorini
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Beatrice Riva
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Luigia G. Fresu
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
11
|
Shen S, Shao Y, Li C. Different types of cell death and their shift in shaping disease. Cell Death Discov 2023; 9:284. [PMID: 37542066 PMCID: PMC10403589 DOI: 10.1038/s41420-023-01581-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/16/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
Cell death is the irreversible stop of life. It is also the basic physiological process of all organisms which involved in the embryonic development, organ maintenance and autoimmunity of the body. In recent years, we have gained more comprehension of the mechanism in cell death and have basically clarified the different types of "programmed cell death", such as apoptosis, necroptosis, autophagy, and pyroptosis, and identified some key genes in these processes. However, in these previous studies, the conversion between different cell death modes and their application in diseases are rarely explored. To sum up, although many valued discoveries have been discovered in the field of cell death in recent years, there are still many unknown problems to be solved in this field. Facts have proved that cell death is a very complex game, and a series of core players have the ability to destroy the delicate balance of the cell environment, from survival to death, from anti-inflammatory to pro-inflammatory. With the thorough research of the complex regulatory mechanism of cell death, there will certainly be exciting new research in this field in the next few years. The sake of this paper is to emphasize the complex mechanism of overturning the balance between different cell fates and provide relevant theoretical basis for the connection between cell death transformation and disease treatment in the future.
Collapse
Affiliation(s)
- Sikou Shen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
12
|
Sartre C, Peurois F, Ley M, Kryszke MH, Zhang W, Courilleau D, Fischmeister R, Ambroise Y, Zeghouf M, Cianferani S, Ferrandez Y, Cherfils J. Membranes prime the RapGEF EPAC1 to transduce cAMP signaling. Nat Commun 2023; 14:4157. [PMID: 37438343 DOI: 10.1038/s41467-023-39894-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023] Open
Abstract
EPAC1, a cAMP-activated GEF for Rap GTPases, is a major transducer of cAMP signaling and a therapeutic target in cardiac diseases. The recent discovery that cAMP is compartmentalized in membrane-proximal nanodomains challenged the current model of EPAC1 activation in the cytosol. Here, we discover that anionic membranes are a major component of EPAC1 activation. We find that anionic membranes activate EPAC1 independently of cAMP, increase its affinity for cAMP by two orders of magnitude, and synergize with cAMP to yield maximal GEF activity. In the cell cytosol, where cAMP concentration is low, EPAC1 must thus be primed by membranes to bind cAMP. Examination of the cell-active chemical CE3F4 in this framework further reveals that it targets only fully activated EPAC1. Together, our findings reformulate previous concepts of cAMP signaling through EPAC proteins, with important implications for drug discovery.
Collapse
Affiliation(s)
- Candice Sartre
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - François Peurois
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Marie Ley
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, IPHC, CNRS UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Marie-Hélène Kryszke
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Wenhua Zhang
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Delphine Courilleau
- Université Paris-Saclay, IPSIT-CIBLOT, Inserm US31, CNRS UAR3679, 91400, Orsay, France
| | | | - Yves Ambroise
- Université Paris-Saclay, CEA, Service de Chimie Bioorganique et de Marquage, 91191, Gif-sur-Yvette, France
| | - Mahel Zeghouf
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, IPHC, CNRS UMR 7178, Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Yann Ferrandez
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France
| | - Jacqueline Cherfils
- Université Paris-Saclay, Ecole Normale Supérieure Paris-Saclay, CNRS, 91190, Gif-sur-Yvette, France.
| |
Collapse
|
13
|
Greiser M, Karbowski M, Kaplan AD, Coleman AK, Verhoeven N, Mannella CA, Lederer WJ, Boyman L. Calcium and bicarbonate signaling pathways have pivotal, resonating roles in matching ATP production to demand. eLife 2023; 12:e84204. [PMID: 37272417 PMCID: PMC10284600 DOI: 10.7554/elife.84204] [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: 10/14/2022] [Accepted: 06/01/2023] [Indexed: 06/06/2023] Open
Abstract
Mitochondrial ATP production in ventricular cardiomyocytes must be continually adjusted to rapidly replenish the ATP consumed by the working heart. Two systems are known to be critical in this regulation: mitochondrial matrix Ca2+ ([Ca2+]m) and blood flow that is tuned by local cardiomyocyte metabolic signaling. However, these two regulatory systems do not fully account for the physiological range of ATP consumption observed. We report here on the identity, location, and signaling cascade of a third regulatory system -- CO2/bicarbonate. CO2 is generated in the mitochondrial matrix as a metabolic waste product of the oxidation of nutrients. It is a lipid soluble gas that rapidly permeates the inner mitochondrial membrane and produces bicarbonate in a reaction accelerated by carbonic anhydrase. The bicarbonate level is tracked physiologically by a bicarbonate-activated soluble adenylyl cyclase (sAC). Using structural Airyscan super-resolution imaging and functional measurements we find that sAC is primarily inside the mitochondria of ventricular cardiomyocytes where it generates cAMP when activated by bicarbonate. Our data strongly suggest that ATP production in these mitochondria is regulated by this cAMP signaling cascade operating within the inter-membrane space by activating local EPAC1 (Exchange Protein directly Activated by cAMP) which turns on Rap1 (Ras-related protein-1). Thus, mitochondrial ATP production is increased by bicarbonate-triggered sAC-signaling through Rap1. Additional evidence is presented indicating that the cAMP signaling itself does not occur directly in the matrix. We also show that this third signaling process involving bicarbonate and sAC activates the mitochondrial ATP production machinery by working independently of, yet in conjunction with, [Ca2+]m-dependent ATP production to meet the energy needs of cellular activity in both health and disease. We propose that the bicarbonate and calcium signaling arms function in a resonant or complementary manner to match mitochondrial ATP production to the full range of energy consumption in ventricular cardiomyocytes.
Collapse
Affiliation(s)
- Maura Greiser
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Physiology, University of Marylan School of MedicineBaltimoreUnited States
- Claude D. Pepper Older Americans Independence Center, University of Maryland School of MedicineBaltimoreUnited States
| | - Mariusz Karbowski
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimoreUnited States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of MedicineBaltimoreUnited States
| | - Aaron David Kaplan
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of MedicineBaltimoreUnited States
| | - Andrew Kyle Coleman
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Physiology, University of Marylan School of MedicineBaltimoreUnited States
| | - Nicolas Verhoeven
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimoreUnited States
| | - Carmen A Mannella
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Physiology, University of Marylan School of MedicineBaltimoreUnited States
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Physiology, University of Marylan School of MedicineBaltimoreUnited States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of MedicineBaltimoreUnited States
| | - Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Physiology, University of Marylan School of MedicineBaltimoreUnited States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of MedicineBaltimoreUnited States
| |
Collapse
|
14
|
Fu D, Luo J, Wu Y, Zhang L, Li L, Chen H, Wen T, Fu Y, Xiong W. Angiotensin II-induced calcium overload affects mitochondrial functions in cardiac hypertrophy by targeting the USP2/MFN2 axis. Mol Cell Endocrinol 2023; 571:111938. [PMID: 37100191 DOI: 10.1016/j.mce.2023.111938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/12/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023]
Abstract
Ubiquitination, a common type of post-translational modification, is known to affect various diseases, including cardiac hypertrophy. Ubiquitin-specific peptidase 2 (USP2) plays a crucial role in regulating cell functions, but its role in cardiac functions remains elusive. The present study aims to investigate the mechanism of USP2 in cardiac hypertrophy. Animal and cell models of cardiac hypertrophy were established using Angiotensin II (Ang II) induction. Our experiments revealed that Ang II induced USP2 downregulation in the in vitro and in vivo models. USP2 overexpression suppressed the degree of cardiac hypertrophy (decreased ANP, BNP, and β-MHC mRNA levels, cell surface area, and ratio of protein/DNA), calcium overload (decreased Ca2+ concentration and t-CaMKⅡ and p-CaMKⅡ, and increased SERCA2), and mitochondrial dysfunction (decreased MDA and ROS and increased MFN1, ATP, MMP, and complex Ⅰ and II) both in vitro and in vivo. Mechanically, USP2 interacted with MFN2 and improved the protein level of MFN2 through deubiquitination. Rescue experiments confirmed that MFN2 downregulation neutralized the protective role of USP2 overexpression in cardiac hypertrophy. Overall, our findings suggested that USP2 overexpression mediated deubiquitination to upregulate MFN2, thus alleviating calcium overload-induced mitochondrial dysfunction and cardiac hypertrophy.
Collapse
Affiliation(s)
- Daoyao Fu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Jing Luo
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Yanze Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Liuping Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Lei Li
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Hui Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Tong Wen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Yongnan Fu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Wenjun Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China; Hypertension Research Institute of Jiangxi Province, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
15
|
Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiac hypertrophy and heart failure. Nat Rev Cardiol 2023; 20:90-108. [PMID: 36050457 DOI: 10.1038/s41569-022-00756-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate the neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple PDE isozymes with different enzymatic properties and subcellular localization regulate local pools of cyclic nucleotides and specific functions. This organization is heavily perturbed during cardiac hypertrophy and heart failure (HF), which can contribute to disease progression. Clinically, PDE inhibition has been considered a promising approach to compensate for the catecholamine desensitization that accompanies HF. Although PDE3 inhibitors, such as milrinone or enoximone, have been used clinically to improve systolic function and alleviate the symptoms of acute HF, their chronic use has proved to be detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as new potential targets to treat HF, each having a unique role in local cyclic nucleotide signalling pathways. In this Review, we describe cAMP and cGMP signalling in cardiomyocytes and present the various PDE families expressed in the heart as well as their modifications in pathological cardiac hypertrophy and HF. We also appraise the evidence from preclinical models as well as clinical data pointing to the use of inhibitors or activators of specific PDEs that could have therapeutic potential in HF.
Collapse
|
16
|
Farag MA, Hariri MLM, Ehab A, Homsi MN, Zhao C, von Bergen M. Cocoa seeds and chocolate products interaction with gut microbiota; mining microbial and functional biomarkers from mechanistic studies, clinical trials and 16S rRNA amplicon sequencing. Crit Rev Food Sci Nutr 2022; 64:3122-3138. [PMID: 36190306 DOI: 10.1080/10408398.2022.2130159] [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] [Indexed: 11/03/2022]
Abstract
In recent years, gut microbiome has evolved as a focal point of interest with growing recognition that a well-balanced gut microbiota is highly relevant to an individual's health status. The present review provides a mechanistic insight on the effects of cocoa chemicals on the gut microbiome and further reveals in silico biomarkers, taxonomic and functional features that distinguish gut microbiome of cocoa consumers and controls by using 16S rRNA gene sequencing data. The polyphenols in cocoa can change the gut microbiota either by inhibiting the growth of pathogenic bacteria in the gut such as Clostridium perfringens or by increasing the growth of beneficial microbiota in the gut such as Lactobacillus and Bifidobacterium. This paper demonstrates the holistic effect of gut microbiota on cocoa chemicals and how it impacts human health. We present herein the first comprehensive review and analysis of how raw and roasted cocoa and its products can specifically influence gut homeostasis, and likewise, how microbiota metabolizes cocoa chemicals. In addition to that, our 16S rRNA amplicon sequencing analysis revealed that the flavone and flavonols metabolism, aminobenzoate degradation and fatty acid elongation pathways represent the three most important signatures of microbial functions associated with cocoa consumption.
Collapse
Affiliation(s)
- Mohamed A Farag
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamad Louai M Hariri
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, New Cairo, Egypt
| | - Aya Ehab
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, New Cairo, Egypt
| | - Masun Nabhan Homsi
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Chao Zhao
- College of Marine Sciences, Fujian Agricultural and Forestry University, Fuzhou, China
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
- Institute of Biochemistry, Life Science Faculty, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Corporeau C, Petton S, Vilaça R, Delisle L, Quéré C, Le Roy V, Dubreuil C, Lacas-Gervais S, Guitton Y, Artigaud S, Bernay B, Pichereau V, Huvet A, Petton B, Pernet F, Fleury E, Madec S, Brigaudeau C, Brenner C, Mazure NM. Harsh intertidal environment enhances metabolism and immunity in oyster (Crassostrea gigas) spat. MARINE ENVIRONMENTAL RESEARCH 2022; 180:105709. [PMID: 35988349 DOI: 10.1016/j.marenvres.2022.105709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
The Pacific oyster Crassostrea gigas is established in the marine intertidal zone, experiencing rapid and highly dynamic environmental changes throughout the tidal cycle. Depending on the bathymetry, oysters face oxygen deprivation, lack of nutrients, and high changes in temperature during alternation of the cycles of emersion/immersion. Here we showed that intertidal oysters at a bathymetry level of 3 and 5 m delayed by ten days the onset of mortality associated with Pacific Oyster Mortality Syndrome (POMS) as compared to subtidal oysters. Intertidal oysters presented a lower growth but similar energetic reserves to subtidal oysters but induced proteomic changes indicative of a boost in metabolism, inflammation, and innate immunity that may have improved their resistance during infection with the Ostreid herpes virus. Our work highlights that intertidal harsh environmental conditions modify host-pathogen interaction and improve oyster health. This study opens new perspectives on oyster farming for mitigation strategies based on tidal height.
Collapse
Affiliation(s)
- Charlotte Corporeau
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France.
| | - Sébastien Petton
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Romain Vilaça
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Lizenn Delisle
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Claudie Quéré
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Valérian Le Roy
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Christine Dubreuil
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, CCMA, Nice, France
| | - Yann Guitton
- Laboratoire d'étude des Résidus et Contaminants dans les Aliments, Oniris, INRA, F-44307, Nantes, France
| | - Sébastien Artigaud
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Benoît Bernay
- Plateforme Proteogen, SFR ICORE 4206, Univ. Caen Basse-Normandie, 14000, Caen, France
| | - Vianney Pichereau
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Arnaud Huvet
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Bruno Petton
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Fabrice Pernet
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Elodie Fleury
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | - Stéphanie Madec
- Ifremer, Univ. Bretagne Occidentale, CNRS, IRD, Équipe soutenue par la fondation ARC, UMR 6539, LEMAR, F, 29280, Plouzané, France
| | | | - Catherine Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches Thérapeutiques, 94805, Villejuif, France
| | - Nathalie M Mazure
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire, 151 route St Antoine de Ginestière, 06204, Nice, France
| |
Collapse
|
18
|
Chen K, Wang S, Sun Z. In Vivo Cardiac-specific Expression of Adenylyl Cyclase 4 Gene Protects against Klotho Deficiency-induced Heart Failure. Transl Res 2022; 244:101-113. [PMID: 35114419 PMCID: PMC9119924 DOI: 10.1016/j.trsl.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/10/2023]
Abstract
Klotho is an aging-suppressor gene. Klotho gene deficiency causes heart failure in Klotho-hypomorphic mutant (KL (-/-)) mice. RNA-seq and western blot analysis showed that adenylyl cyclase type IV (AC4) mRNA and protein expression was largely decreased in cardiomyocytes of KL (-/-) mice. The objective of this study was to investigate whether in vivo cardiac-specific expression of AC4 gene protects against Klotho deficiency-induced heart failure. Interestingly, in vivo AAV-based cardiac-specific AC4 gene expression increased left ventricular fractional shortening, ejection fraction, stroke volume, and left ventricular end-diastolic volume in KL (-/-) mice, suggesting that cardiac-specific AC4 gene expression improves Klotho deficiency-induced heart dysfunction. Cardiac-specific AC4 gene expression also decreased Klotho deficiency-induced cardiac hypertrophy. Cardiac-specific AC4 gene expression alleviated Klotho deficiency-induced cardiac fibrosis and calcification. Furthermore, cardiac-specific AC4 gene expression attenuated mitochondrial dysfunction, superoxide accumulation and cardiomyocyte apoptotic cell death. Thus, downregulation of AC4 may contribute to Klotho deficiency-induced heart failure. Mechanistically, AAV2/9-αMHC-AC4 increased cardiomyocytic cAMP levels and thus regulated the PKA-PLN-SERCA2 signal pathway, which is critical in modulating calcium flux and mitochondrial function. In conclusion, cardiac-specific AC4 gene expression protects against Klotho deficiency-induced heart failure through increasing cardiomyocytic cAMP levels, which alleviates cAMP-dependent mitochondrial dysfunction, superoxide accumulation and apoptotic cell death. AC4 regulates superoxide levels via the cAMP-PKA pathway. AC4 could be a potential therapeutic target for heart failure associated with Klotho deficiency. Heart failure is the major cause of mortality in patients with chronic kidney disease (CKD). A decrease in Klotho levels is linked to CKD.
Collapse
Affiliation(s)
- Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America.
| |
Collapse
|
19
|
Musheshe N, Oun A, Sabogal-Guáqueta AM, Trombetta-Lima M, Mitchel SC, Adzemovic A, Speek O, Morra F, van der Veen CHJT, Lezoualc’h F, Cheng X, Schmidt M, Dolga AM. Pharmacological Inhibition of Epac1 Averts Ferroptosis Cell Death by Preserving Mitochondrial Integrity. Antioxidants (Basel) 2022; 11:antiox11020314. [PMID: 35204198 PMCID: PMC8868285 DOI: 10.3390/antiox11020314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 01/27/2023] Open
Abstract
Exchange proteins directly activated by cAMP (Epac) proteins are implicated in a wide range of cellular functions including oxidative stress and cell survival. Mitochondrial-dependent oxidative stress has been associated with progressive neuronal death underlying the pathology of many neurodegenerative diseases. The role of Epac modulation in neuronal cells in relation to cell survival and death, as well as its potential effect on mitochondrial function, is not well established. In immortalized hippocampal (HT-22) neuronal cells, we examined mitochondria function in the presence of various Epac pharmacological modulators in response to oxidative stress due to ferroptosis. Our study revealed that selective pharmacological modulation of Epac1 or Epac2 isoforms, exerted differential effects in erastin-induced ferroptosis conditions in HT-22 cells. Epac1 inhibition prevented cell death and loss of mitochondrial integrity induced by ferroptosis, while Epac2 inhibition had limited effects. Our data suggest Epac1 as a plausible therapeutic target for preventing ferroptosis cell death associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Nshunge Musheshe
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Correspondence: (N.M.); (A.M.D.)
| | - Asmaa Oun
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Angélica María Sabogal-Guáqueta
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Sarah C. Mitchel
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Ahmed Adzemovic
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Oliver Speek
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Francesca Morra
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Christina H. J. T. van der Veen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
| | - Frank Lezoualc’h
- Inserm UMR-1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse Paul Sabatier, 31400 Toulouse, France;
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX 7000, USA;
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Groningen Research Institute of Asthma and COPD (GRIAC), Groningen Research Institute of Pharmacy (GRIP), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (A.O.); (A.M.S.-G.); (M.T.-L.); (S.C.M.); (A.A.); (O.S.); (F.M.); (C.H.J.T.v.d.V.); (M.S.)
- Correspondence: (N.M.); (A.M.D.)
| |
Collapse
|
20
|
Liu D, Peyre F, Loissell-Baltazar YA, Courilleau D, Lacas-Gervais S, Nicolas V, Jacquet E, Dokudovskaya S, Taran F, Cintrat JC, Brenner C. Identification of Small Molecules Inhibiting Cardiomyocyte Necrosis and Apoptosis by Autophagy Induction and Metabolism Reprogramming. Cells 2022; 11:cells11030474. [PMID: 35159285 PMCID: PMC8834338 DOI: 10.3390/cells11030474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022] Open
Abstract
Improvement of anticancer treatments is associated with increased survival of cancer patients at risk of cardiac disease. Therefore, there is an urgent need for new therapeutic molecules capable of preventing acute and long-term cardiotoxicity. Here, using commercial and home-made chemolibraries, we performed a robust phenotypic high-throughput screening in rat cardiomyoblast cell line H9c2, searching for small molecules capable of inhibiting cell death. A screen of 1600 compounds identified six molecules effective in preventing necrosis and apoptosis induced by H2O2 and camptothecin in H9c2 cells and in rat neonatal ventricular myocytes. In cells treated with these molecules, we systematically evaluated the expression of BCL-2 family members, autophagy progression, mitochondrial network structure, regulation of mitochondrial fusion/fission, reactive oxygen species, and ATP production. We found that these compounds affect autophagy induction to prevent cardiac cell death and can be promising cardioprotective drugs during chemotherapy.
Collapse
Affiliation(s)
- Dawei Liu
- Centre National de Recherche Scientifique (CNRS), Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (D.L.); (F.P.); (Y.A.L.-B.); (S.D.)
| | - Félix Peyre
- Centre National de Recherche Scientifique (CNRS), Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (D.L.); (F.P.); (Y.A.L.-B.); (S.D.)
| | - Yahir Alberto Loissell-Baltazar
- Centre National de Recherche Scientifique (CNRS), Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (D.L.); (F.P.); (Y.A.L.-B.); (S.D.)
| | - Delphine Courilleau
- Inserm, Centre National de Recherche Scientifique (CNRS), Ingénierie et Plateformes au Service de l’Innovation Thérapeutique, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (D.C.); (V.N.)
| | - Sandra Lacas-Gervais
- Centre Commun de Microscopie Appliquée, CCMA, Université Côte d’Azur, 06103 Nice, France;
| | - Valérie Nicolas
- Inserm, Centre National de Recherche Scientifique (CNRS), Ingénierie et Plateformes au Service de l’Innovation Thérapeutique, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (D.C.); (V.N.)
| | - Eric Jacquet
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, 91190 Gif-sur-Yvette, France;
| | - Svetlana Dokudovskaya
- Centre National de Recherche Scientifique (CNRS), Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (D.L.); (F.P.); (Y.A.L.-B.); (S.D.)
| | - Frédéric Taran
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, CEA, INRAE, SCBM, 91191 Gif-sur-Yvette, France; (F.T.); (J.-C.C.)
| | - Jean-Christophe Cintrat
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, CEA, INRAE, SCBM, 91191 Gif-sur-Yvette, France; (F.T.); (J.-C.C.)
| | - Catherine Brenner
- Centre National de Recherche Scientifique (CNRS), Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (D.L.); (F.P.); (Y.A.L.-B.); (S.D.)
- Correspondence:
| |
Collapse
|
21
|
Figarola-Centurión I, Escoto-Delgadillo M, González-Enríquez GV, Gutiérrez-Sevilla JE, Vázquez-Valls E, Torres-Mendoza BM. Sirtuins Modulation: A Promising Strategy for HIV-Associated Neurocognitive Impairments. Int J Mol Sci 2022; 23:643. [PMID: 35054829 PMCID: PMC8775450 DOI: 10.3390/ijms23020643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 02/01/2023] Open
Abstract
HIV-Associated neurocognitive disorder (HAND) is one of the major concerns since it persists in 40% of this population. Nowadays, HAND neuropathogenesis is considered to be caused by the infected cells that cross the brain-blood barrier and produce viral proteins that can be secreted and internalized into neurons leading to disruption of cellular processes. The evidence points to viral proteins such as Tat as the causal agent for neuronal alteration and thus HAND. The hallmarks in Tat-induced neurodegeneration are endoplasmic reticulum stress and mitochondrial dysfunction. Sirtuins (SIRTs) are NAD+-dependent deacetylases involved in mitochondria biogenesis, unfolded protein response, and intrinsic apoptosis pathway. Tat interaction with these deacetylases causes inhibition of SIRT1 and SIRT3. Studies revealed that SIRTs activation promotes neuroprotection in neurodegenerative diseases such Alzheimer's and Parkinson's disease. Therefore, this review focuses on Tat-induced neurotoxicity mechanisms that involve SIRTs as key regulators and their modulation as a therapeutic strategy for tackling HAND and thereby improving the quality of life of people living with HIV.
Collapse
Affiliation(s)
- Izchel Figarola-Centurión
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
| | - Martha Escoto-Delgadillo
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Guadalajara 44600, Mexico
| | - Gracia Viviana González-Enríquez
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Juan Ernesto Gutiérrez-Sevilla
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Microbiología Médica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Eduardo Vázquez-Valls
- Generación de Recursos Profesionales, Investigación y Desarrollo, Secretaria de Salud, Jalisco, Guadalajara 44100, Mexico;
| | - Blanca Miriam Torres-Mendoza
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| |
Collapse
|
22
|
Wang H, Liu Y, Guo Z, Wu K, Zhang Y, Tian Y, Zhao B, Lu H. Aconitine induces cell apoptosis via mitochondria and death receptor signaling pathways in hippocampus cell line. Res Vet Sci 2022; 143:124-133. [PMID: 35026629 DOI: 10.1016/j.rvsc.2022.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/20/2021] [Accepted: 01/03/2022] [Indexed: 01/10/2023]
Abstract
Aconitine is a plant toxin derived from aconitum genus and well known for its neurological and vascular toxicity. However, the mechanism of toxicity on the growth and apoptosis of the neurological cells has not been well investigated. In this study, we used HT22 cell lines derived from hippocampus to explore the mechanism. We began with examination of the viability and DA (dopamine) contents of cells treated with different dose of aconitine. In this study, we investigated the role of apoptosis in AC-induced HT22 cells. Our results showed that aconitine inhibited HT22 cells growth and increased DA contents in a dose dependent manner. Aconitine treatment induced apoptosis in HT22 cells and we found aconitine induced apoptosis by upregulating the expression of Bax, Cyto c, Apaf-1, Caspase9, Fas, Fas-L, Fadd, Caspase8, Caspase3 with concomitant decreasing of Bcl-2 and Bid expression. Collectively, results suggest that aconitine induce apoptosis through mitochondrial-mediated and death receptor signaling pathways in HT22 cells.
Collapse
Affiliation(s)
- Hui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanbing Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ziyu Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Kexin Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yunhao Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hao Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
23
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
24
|
Mora FAA, Musheshe N, Arroyave Ospina JC, Geng Y, Soto JM, Rodrigo JA, Alieva T, Buist-Homan M, Lezoualc'h F, Cheng X, Schmidt M, Moshage H. Metformin protects against diclofenac-induced toxicity in primary rat hepatocytes by preserving mitochondrial integrity via a pathway involving EPAC. Biomed Pharmacother 2021; 143:112072. [PMID: 34464747 DOI: 10.1016/j.biopha.2021.112072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/23/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE It has been shown that the antidiabetic drug metformin protects hepatocytes against toxicity by various stressors. Chronic or excessive consumption of diclofenac (DF) - a pain-relieving drug, leads to drug-induced liver injury via a mechanism involving mitochondrial damage and ultimately apoptotic death of hepatocytes. However, whether metformin protects against DF-induced toxicity is unknown. Recently, it was also shown that cAMP elevation is protective against DF-induced apoptotic death in hepatocytes, a protective effect primarily involving the downstream cAMP effector EPAC and preservation of mitochondrial function. This study therefore aimed at investigating whether metformin protects against DF-induced toxicity via cAMP-EPACs. EXPERIMENTAL APPROACH Primary rat hepatocytes were exposed to 400 µmol/L DF. CE3F4 or ESI-O5 were used as EPAC-1 or 2 inhibitors respectively. Apoptosis was measured by caspase-3 activity and necrosis by Sytox green staining. Seahorse X96 assay was used to determine mitochondrial function. Mitochondrial reactive oxygen species (ROS) production was measured using MitoSox, mitochondrial MnSOD expression was determined by immunostaining and mitochondrial morphology (fusion and fission ratio) by 3D refractive index imaging. KEY RESULTS Metformin (1 mmol/L) was protective against DF-induced apoptosis in hepatocytes. This protective effect was EPAC-dependent (mainly EPAC-2). Metformin restored mitochondrial morphology in an EPAC-independent manner. DF-induced mitochondrial dysfunction which was demonstrated by decreased oxygen consumption rate, an increased ROS production and a reduced MnSOD level, were all reversed by metformin in an EPAC-dependent manner. CONCLUSION AND IMPLICATIONS Metformin protects hepatocytes against DF-induced toxicity via cAMP-dependent EPAC-2.
Collapse
Affiliation(s)
- Fabio Alejandro Aguilar Mora
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Nshunge Musheshe
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Johanna C Arroyave Ospina
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Yana Geng
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Juan M Soto
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - José A Rodrigo
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Tatiana Alieva
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Metaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France.
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Martina Schmidt
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
25
|
Adzika GK, Hou H, Adekunle AO, Rizvi R, Adzraku SY, Li K, Deng QM, Mprah R, Ndzie Noah ML, Adu-Amankwaah J, Machuki JO, Shang W, Ma T, Koda S, Ma X, Sun H. Amlexanox and Forskolin Prevents Isoproterenol-Induced Cardiomyopathy by Subduing Cardiomyocyte Hypertrophy and Maladaptive Inflammatory Responses. Front Cell Dev Biol 2021; 9:719351. [PMID: 34631707 PMCID: PMC8497899 DOI: 10.3389/fcell.2021.719351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic catecholamine stress (CCS) induces the occurrence of cardiomyopathy-pathological cardiac hypertrophy (PCH), which is characterized by left ventricular systolic dysfunction (LVSD). Recently, mounting evidence has implicated myocardial inflammation in the exacerbation of pathological cardiac remodeling. However, there are currently no well-defined treatment interventions or regimes targeted at both the attenuation of maladaptive myocardial hypertrophy and inflammation during CCS to prevent PCH. G protein-coupled receptor kinase 5 (GRK5) and adenylyl cyclases (ACs)-cAMP mediates both cardiac and inflammatory responses. Also, GRK5 and ACs are implicated in stress-induced LVSD. Herein, we aimed at preventing PCH during CCS via modulating adaptive cardiac and inflammatory responses by inhibiting GRK5 and/or stimulating ACs. Isoproterenol-induced cardiomyopathy (ICM) was modeled using 0.5 mg/100 g/day isoproterenol injections for 40 days. Alterations in cardiac and inflammatory responses were assessed from the myocardia. Similarities in the immunogenicity of cardiac troponin I (cTnI) and lipopolysaccharide under CCS were assessed, and Amlexanox (35 μM/ml) and/or Forskolin (10 μM/ml) were then employed in vitro to modulate adaptive inflammatory responses by inhibiting GRK5 or activating ACs-cAMP, respectively. Subsequently, Amlexanox (2.5 mg/100 g/day) and/or Forskolin (0.5 mg/100 g/day) were then translated into in vivo during CCS to modulate adaptive cardiac and inflammatory responses. The effects of Amlexanox and Forskolin on regulating myocardial systolic functions and inflammatory responses during CCS were ascertained afterward. PCH mice had excessive myocardial hypertrophy, fibrosis, and aggravated LVSD, which were accompanied by massive CD68+ inflammatory cell infiltrations. In vitro, Forskolin-AC/cAMP was effective than Amlexanox-GRK5 at downregulating proinflammatory responses during stress; nonetheless, Amlexanox and Forskolin combination demonstrated the most efficacy in modulating adaptive inflammatory responses. Individually, the translated Amlexanox and Forskolin treatment interventions were ineffective at subduing the pathological remodeling and sustaining cardiac function during CCS. However, their combination was potent at preventing LVSD during CCS by attenuating maladaptive myocardial hypertrophy, fibrosis, and inflammatory responses. The treatment intervention attained its potency mainly via Forskolin-ACs/cAMP-mediated modulation of cardiac and inflammatory responses, coupled with Amlexanox inhibition of GRK5 mediated maladaptive cascades. Taken together, our findings highlight the Amlexanox and Forskolin combination as a potential therapeutic intervention for preventing the occurrence of pathological cardiac hypertrophy during chronic stress.
Collapse
Affiliation(s)
| | - Hongjian Hou
- Department of Physiology, Xuzhou Medical University, Xuzhou, China.,The College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | | | | | - Seyram Yao Adzraku
- Key Laboratory of Bone Marrow Stem Cell, Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kexue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Qi-Ming Deng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Department of Cardiology, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | | | | | - Wenkang Shang
- Faculty of Biology, Institute of Biochemistry and Molecular Biology, ZBMZ, Albert-Ludwigs University of Freiburg, Freiburg, Germany
| | - Tongtong Ma
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Stephane Koda
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Xianluo Ma
- Internal Medicine-Cardiovascular Department, People's Hospital of Jiawang District, Xuzhou, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
26
|
Laudette M, Sainte-Marie Y, Cousin G, Bergonnier D, Belhabib I, Brun S, Formoso K, Laib L, Tortosa F, Bergoglio C, Marcheix B, Borén J, Lairez O, Fauconnier J, Lucas A, Mialet-Perez J, Moro C, Lezoualc'h F. Cyclic AMP-binding protein Epac1 acts as a metabolic sensor to promote cardiomyocyte lipotoxicity. Cell Death Dis 2021; 12:824. [PMID: 34471096 PMCID: PMC8410846 DOI: 10.1038/s41419-021-04113-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/28/2021] [Accepted: 08/16/2021] [Indexed: 01/21/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) is a master regulator of mitochondrial metabolism but its precise mechanism of action yet remains unclear. Here, we found that a dietary saturated fatty acid (FA), palmitate increased intracellular cAMP synthesis through the palmitoylation of soluble adenylyl cyclase in cardiomyocytes. cAMP further induced exchange protein directly activated by cyclic AMP 1 (Epac1) activation, which was upregulated in the myocardium of obese patients. Epac1 enhanced the activity of a key enzyme regulating mitochondrial FA uptake, carnitine palmitoyltransferase 1. Consistently, pharmacological or genetic Epac1 inhibition prevented lipid overload, increased FA oxidation (FAO), and protected against mitochondrial dysfunction in cardiomyocytes. In addition, analysis of Epac1 phosphoproteome led us to identify two key mitochondrial enzymes of the the β-oxidation cycle as targets of Epac1, the long-chain FA acyl-CoA dehydrogenase (ACADL) and the 3-ketoacyl-CoA thiolase (3-KAT). Epac1 formed molecular complexes with the Ca2+/calmodulin-dependent protein kinase II (CaMKII), which phosphorylated ACADL and 3-KAT at specific amino acid residues to decrease lipid oxidation. The Epac1-CaMKII axis also interacted with the α subunit of ATP synthase, thereby further impairing mitochondrial energetics. Altogether, these findings indicate that Epac1 disrupts the balance between mitochondrial FA uptake and oxidation leading to lipid accumulation and mitochondrial dysfunction, and ultimately cardiomyocyte death.
Collapse
Affiliation(s)
- Marion Laudette
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yannis Sainte-Marie
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Grégoire Cousin
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse Rangueil, Toulouse, France
| | - Dorian Bergonnier
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Ismahane Belhabib
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Stéphanie Brun
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse Rangueil, Toulouse, France
| | - Karina Formoso
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Loubna Laib
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Florence Tortosa
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Camille Bergoglio
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Bertrand Marcheix
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse Rangueil, Toulouse, France
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Olivier Lairez
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse Rangueil, Toulouse, France
| | - Jérémy Fauconnier
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Alexandre Lucas
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Jeanne Mialet-Perez
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Cédric Moro
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Frank Lezoualc'h
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France.
| |
Collapse
|
27
|
Vega Hissi EG, De Costa Guardamagna AB, Garro AD, Falcon CR, Anderluh M, Tomašič T, Kikelj D, Yaneff A, Davio CA, Enriz RD, Zurita AR. A Potent N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide Inhibitor of Adenylyl Cyclase of G. lamblia: Biological Evaluation and Molecular Modelling Studies. ChemMedChem 2021; 16:2094-2105. [PMID: 33783977 DOI: 10.1002/cmdc.202100037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Indexed: 11/06/2022]
Abstract
In this work, we report a derivative of N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide as a new inhibitor for adenylyl cyclase of Giardia lamblia which was obtained from a study using structural data of the nucleotidyl cyclase 1 (gNC1) of this parasite. For such a study, we developed a model for this specific enzyme by using homology techniques, which is the first model reported for gNC1 of G. lamblia. Our studies show that the new inhibitor has a competitive mechanism of action against this enzyme. 2-Hydroxyestradiol was used as the reference compound for comparative studies. Results in this work are important from two points of view. on the one hand, an experimentally corroborated model for gNC1 of G. lamblia obtained by molecular modelling is presented; on the other hand, the new inhibitor obtained is an undoubtedly excellent starting structure for the development of new metabolic inhibitors for G. lamblia.
Collapse
Affiliation(s)
- Esteban G Vega Hissi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Antonella B De Costa Guardamagna
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adriana D Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Cristian R Falcon
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Marko Anderluh
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Tihomir Tomašič
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Danijel Kikelj
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Carlos A Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adolfo R Zurita
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| |
Collapse
|
28
|
Aguilar Mora FA, Musheshe N, Oun A, Buist-Homan M, Lezoualc'h F, Cheng X, Schmidt M, Moshage H. Elevated cAMP Protects against Diclofenac-Induced Toxicity in Primary Rat Hepatocytes: A Protective Effect Mediated by the Exchange Protein Directly Activated by cAMP/cAMP-Regulated Guanine Nucleotide Exchange Factors. Mol Pharmacol 2021; 99:294-307. [PMID: 33574047 PMCID: PMC11033960 DOI: 10.1124/molpharm.120.000217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic consumption of the nonsteroidal anti-inflammatory drug diclofenac may induce drug-induced liver injury (DILI). The mechanism of diclofenac-induced liver injury is partially elucidated and involves mitochondrial damage. Elevated cAMP protects hepatocytes against bile acid-induced injury. However, it is unknown whether cAMP protects against DILI and, if so, which downstream targets of cAMP are implicated in the protective mechanism, including the classic protein kinase A (PKA) pathway or alternative pathways like the exchange protein directly activated by cAMP (EPAC). The aim of this study was to investigate whether cAMP and/or its downstream targets protect against diclofenac-induced injury in hepatocytes. Rat hepatocytes were exposed to 400 µmol/l diclofenac. Apoptosis and necrosis were measured by caspase-3 activity assay and Sytox green staining, respectively. Mitochondrial membrane potential (MMP) was measured by JC-10 staining. mRNA and protein expression were assessed by quantitative polymerase chain reaction (qPCR) and Western blot, respectively. The cAMP-elevating agent 7β-acetoxy-8,13-epoxy-1α,6β,9α-trihydroxylabd-14-en-11-one (forskolin), the pan-phosphodiesterase inhibitor IBMX, and EPAC inhibitors 5,7-dibromo-6-fluoro-3,4-dihydro-2-methyl-1(2H)-quinoline carboxaldehyde (CE3F4) and ESI-O5 were used to assess the role of cAMP and its effectors, PKA or EPAC. Diclofenac exposure induced apoptotic cell death and loss of MMP in hepatocytes. Both forskolin and IBMX prevented diclofenac-induced apoptosis. EPAC inhibition but not PKA inhibition abolished the protective effect of forskolin and IBMX. Forskolin and IBMX preserved the MMP, whereas both EPAC inhibitors diminished this effect. Both EPAC1 and EPAC2 were expressed in hepatocytes and localized in mitochondria. cAMP elevation protects hepatocytes against diclofenac-induced cell death, a process primarily involving EPACs. The cAMP/EPAC pathway may be a novel target for treatment of DILI. SIGNIFICANCE STATEMENT: This study shows two main highlights. First, elevated cAMP levels protect against diclofenac-induced apoptosis in primary hepatocytes via maintenance of mitochondrial integrity. In addition, this study proposes the existence of mitochondrial cAMP-EPAC microdomains in rat hepatocytes, opening new avenues for targeted therapy in drug-induced liver injury (DILI). Both EPAC1 and EPAC2, but not protein kinase A, are responsible for this protective effect. Our findings present cAMP-EPAC as a potential target for the treatment of DILI and liver injury involving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fabio Alejandro Aguilar Mora
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Nshunge Musheshe
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Asmaa Oun
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Manon Buist-Homan
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Frank Lezoualc'h
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Xiaodong Cheng
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Martina Schmidt
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| | - Han Moshage
- Dept. Gastroenterology and Hepatology (F.A.A.M., M.B.-H., H.M.), Dept. Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC (N.M., A.O., M.S.), Dept. Laboratory Medicine (M.B.-H., H.M.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France (F.L.); and Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, Texas (X.C.)
| |
Collapse
|
29
|
Liu D, Lai HT, Peyre F, Brenner C. Multiple analysis of mitochondrial metabolism, autophagy and cell death. Methods Cell Biol 2021; 164:95-112. [PMID: 34225921 DOI: 10.1016/bs.mcb.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In the perspective to evaluate the toxicity of drug candidates or the exploration of intracellular signaling pathways of cell stress response and pathophysiological conditions, we propose to evaluate cell death, autophagy, mitochondrial network and energetic metabolism by a series of optimized joint protocols for neonatal primary rat cardiomyocytes or H9c2 cardiac cell line in 96 well microtiter plates. We used Digitoxigenin and Digoxin, two cardiac glycosides, and Rapamycin as control drugs, for inhibition of oxidative stress-induced cell death and autophagy induction, respectively.
Collapse
Affiliation(s)
- D Liu
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France
| | - H T Lai
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France
| | - F Peyre
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France
| | - C Brenner
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France.
| |
Collapse
|
30
|
Regulation of Mitochondrial Homeostasis by sAC-Derived cAMP Pool: Basic and Translational Aspects. Cells 2021; 10:cells10020473. [PMID: 33671810 PMCID: PMC7926680 DOI: 10.3390/cells10020473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 01/21/2023] Open
Abstract
In contrast to the traditional view of mitochondria being solely a source of cellular energy, e.g., the "powerhouse" of the cell, mitochondria are now known to be key regulators of numerous cellular processes. Accordingly, disturbance of mitochondrial homeostasis is a basic mechanism in several pathologies. Emerging data demonstrate that 3'-5'-cyclic adenosine monophosphate (cAMP) signalling plays a key role in mitochondrial biology and homeostasis. Mitochondria are equipped with an endogenous cAMP synthesis system involving soluble adenylyl cyclase (sAC), which localizes in the mitochondrial matrix and regulates mitochondrial function. Furthermore, sAC localized at the outer mitochondrial membrane contributes significantly to mitochondrial biology. Disturbance of the sAC-dependent cAMP pools within mitochondria leads to mitochondrial dysfunction and pathology. In this review, we discuss the available data concerning the role of sAC in regulating mitochondrial biology in relation to diseases.
Collapse
|
31
|
Soluble adenylyl cyclase regulates the cytosolic NADH/NAD + redox state and the bioenergetic switch between glycolysis and oxidative phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148367. [PMID: 33412125 DOI: 10.1016/j.bbabio.2020.148367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/11/2020] [Accepted: 12/19/2020] [Indexed: 12/22/2022]
Abstract
The evolutionarily conserved soluble adenylyl cyclase (sAC, ADCY10) mediates cAMP signaling exclusively in intracellular compartments. Because sAC activity is sensitive to local concentrations of ATP, bicarbonate, and free Ca2+, sAC is potentially an important metabolic sensor. Nonetheless, little is known about how sAC regulates energy metabolism in intact cells. In this study, we demonstrated that both pharmacological and genetic suppression of sAC resulted in increased lactate secretion and decreased pyruvate secretion in multiple cell lines and primary cultures of mouse hepatocytes and cholangiocytes. The increased extracellular lactate-to-pyruvate ratio upon sAC suppression reflected an increased cytosolic free [NADH]/[NAD+] ratio, which was corroborated by using the NADH/NAD+ redox biosensor Peredox-mCherry. Mechanistic studies in permeabilized HepG2 cells showed that sAC inhibition specifically suppressed complex I of the mitochondrial respiratory chain. A survey of cAMP effectors revealed that only selective inhibition of exchange protein activated by cAMP 1 (Epac1), but not protein kinase A (PKA) or Epac2, suppressed complex I-dependent respiration and significantly increased the cytosolic NADH/NAD+ redox state. Analysis of the ATP production rate and the adenylate energy charge showed that inhibiting sAC reciprocally affects ATP production by glycolysis and oxidative phosphorylation while maintaining cellular energy homeostasis. In conclusion, our study shows that, via the regulation of complex I-dependent mitochondrial respiration, sAC-Epac1 signaling regulates the cytosolic NADH/NAD+ redox state, and coordinates oxidative phosphorylation and glycolysis to maintain cellular energy homeostasis. As such, sAC is effectively a bioenergetic switch between aerobic glycolysis and oxidative phosphorylation at the post-translational level.
Collapse
|
32
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Identification of Featured Metabolism-Related Genes in Patients with Acute Myocardial Infarction. DISEASE MARKERS 2020; 2020:8880004. [PMID: 33354250 PMCID: PMC7737445 DOI: 10.1155/2020/8880004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022]
Abstract
Objective A growing body of emerging evidence indicates that metabolic processes play a pivotal role in the biological processes underlying acute myocardial infarction (AMI). The aim of the current study was to identify featured metabolism-related genes in patients with AMI using a support vector machine (SVM) and to further explore the value of these genes in the diagnosis of AMI. Methods Gene microarray expression data related to AMI were downloaded from the GSE66360 dataset in the Gene Expression Omnibus (GEO) database. This data set consisted of 50 AMI samples and 49 normal controls that were randomly classified into a discovery cohort (21 AMI samples and 22 normal controls) and a validation cohort (28 AMI and 28 normal controls). We applied a machine learning method that combined SVM with recursive feature elimination (RFE) to discriminate AMI patients from normal controls. Based on this, an SVM classifier was constructed. Receiver operating characteristic (ROC) analysis was used to investigate the predictive value for the early diagnosis of AMI in the two cohorts and was then further verified in an independent external cohort. Results Three metabolism-related genes were identified based on SVM-RFE (AKR1C3, GLUL, and PDE4B). The SVM classifier based on the three genes allowed for excellent discrimination between AMI and healthy samples in both the discovery cohort (AUC = 0.989) and the validation cohort (AUC = 0.964), and this was further confirmed in the GSE68060 dataset (AUC = 0.839). Additionally, the SVM classifier allowed for perfect discrimination between recurrent AMI events and nonrecurrent events in the GSE68060 cohort (AUC = 0.992). GO and KEGG pathway enrichment analysis of the identified featured genes revealed significant enrichment of specific metabolic pathways. Conclusion The identified metabolism-related genes may play important roles in the development of AMI and may represent diagnostic and therapeutic biomarkers of AMI.
Collapse
|
34
|
Beiersdorf J, Hevesi Z, Calvigioni D, Pyszkowski J, Romanov R, Szodorai E, Lubec G, Shirran S, Botting CH, Kasper S, Guy GW, Gray R, Di Marzo V, Harkany T, Keimpema E. Adverse effects of Δ9-tetrahydrocannabinol on neuronal bioenergetics during postnatal development. JCI Insight 2020; 5:135418. [PMID: 33141759 PMCID: PMC7714410 DOI: 10.1172/jci.insight.135418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Ongoing societal changes in views on the medical and recreational roles of cannabis increased the use of concentrated plant extracts with a Δ9-tetrahydrocannabinol (THC) content of more than 90%. Even though prenatal THC exposure is widely considered adverse for neuronal development, equivalent experimental data for young age cohorts are largely lacking. Here, we administered plant-derived THC (1 or 5 mg/kg) to mice daily during P5–P16 and P5–P35 and monitored its effects on hippocampal neuronal survival and specification by high-resolution imaging and iTRAQ proteomics, respectively. We found that THC indiscriminately affects pyramidal cells and both cannabinoid receptor 1+ (CB1R)+ and CB1R– interneurons by P16. THC particularly disrupted the expression of mitochondrial proteins (complexes I–IV), a change that had persisted even 4 months after the end of drug exposure. This was reflected by a THC-induced loss of membrane integrity occluding mitochondrial respiration and could be partially or completely rescued by pH stabilization, antioxidants, bypassed glycolysis, and targeting either mitochondrial soluble adenylyl cyclase or the mitochondrial voltage-dependent anion channel. Overall, THC exposure during infancy induces significant and long-lasting reorganization of neuronal circuits through mechanisms that, in large part, render cellular bioenergetics insufficient to sustain key developmental processes in otherwise healthy neurons. Repeated THC exposure in juvenile mice compromises the limbic circuitry, with life-long impairment to the respiration of neurons.
Collapse
Affiliation(s)
- Johannes Beiersdorf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela Calvigioni
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Roman Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Paracelsus Private Medical University, Salzburg, Austria
| | - Sally Shirran
- School of Chemistry, University of St. Andrews, St. Andrews, United Kingdom
| | | | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | | | - Roy Gray
- GW Phamaceuticals, Salisbury, Wiltshire, United Kingdom
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Canada Excellence Research Chair, Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, Québec, Canada
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Department of Neuroscience, Biomedikum D7, Karolinska Institutet, Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
36
|
Formoso K, Lezoualc'h F, Mialet-Perez J. Role of EPAC1 Signalosomes in Cell Fate: Friends or Foes? Cells 2020; 9:E1954. [PMID: 32854274 PMCID: PMC7563956 DOI: 10.3390/cells9091954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
The compartmentation of signaling processes is accomplished by the assembly of protein complexes called signalosomes. These signaling platforms colocalize enzymes, substrates, and anchoring proteins into specific subcellular compartments. Exchange protein directly activated by cAMP 1 (EPAC1) is an effector of the second messenger, 3',5'-cyclic adenosine monophosphate (cAMP) that is associated with multiple roles in several pathologies including cardiac diseases. Both EPAC1 intracellular localization and molecular partners are key players in the regulation of cell fate, which may have important therapeutic potential. In this review, we summarize the recent findings on EPAC1 structure, regulation, and pharmacology. We describe the importance of EPAC1 subcellular distribution in its biological action, paying special attention to its nuclear localization and mechanism of action leading to cardiomyocyte hypertrophy. In addition, we discuss the role of mitochondrial EPAC1 in the regulation of cell death. Depending on the cell type and stress condition, we present evidence that supports either a protective or detrimental role of EPAC1 activation.
Collapse
Affiliation(s)
- Karina Formoso
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| | - Frank Lezoualc'h
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| | - Jeanne Mialet-Perez
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| |
Collapse
|
37
|
Khailova LS, Vygodina TV, Lomakina GY, Kotova EA, Antonenko YN. Bicarbonate suppresses mitochondrial membrane depolarization induced by conventional uncouplers. Biochem Biophys Res Commun 2020; 530:29-34. [PMID: 32828301 DOI: 10.1016/j.bbrc.2020.06.131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
Abstract
Bicarbonate has been known to modulate activities of various mitochondrial enzymes such as ATPase and soluble adenylyl cyclase. Here, we found that the ability of conventional protonophoric uncouplers, such as 2,4-dinitrophenol (DNP), carbonylcyanide p-trifluoromethoxyphenylhydrazone (FCCP) and carbonyl cyanide m-chlorophenyl hydrazone (CCCP), but not that of the new popular uncoupler BAM15, to decrease mitochondrial membrane potential was significantly diminished in the presence of millimolar concentrations of bicarbonate. Thus, the depolarizing activity of DNP and FCCP in mitochondria could be sensitive to the local concentration of bicarbonate in cells and tissues. However, bicarbonate could not restore the ATP synthesis suppressed by DNP or CCCP in mitochondria. Bicarbonate neither altered the depolarizing action of DNP and FCCP on proteoliposomes with reconstituted cytochrome c oxidase, nor affected the protonophoric activity of DNP and FCCP in artificial lipid membranes as measured with pyranine-loaded liposomes, thereby showing that the bicarbonate-induced reversal of the depolarizing action of DNP and FCCP on mitochondria did not result from direct interaction of bicarbonate with the uncouplers.
Collapse
Affiliation(s)
- Ljudmila S Khailova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, Moscow, 119991, Russia
| | - Tatyana V Vygodina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, Moscow, 119991, Russia
| | - Galina Y Lomakina
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow, 119991, Russia; Bauman Moscow State Technical University, Baumanskaya 2-ya, 5/1, Moscow, 105005, Russia
| | - Elena A Kotova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, Moscow, 119991, Russia
| | - Yuri N Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, Moscow, 119991, Russia.
| |
Collapse
|
38
|
The Soluble Adenylyl Cyclase Inhibitor LRE1 Prevents Hepatic Ischemia/Reperfusion Damage Through Improvement of Mitochondrial Function. Int J Mol Sci 2020; 21:ijms21144896. [PMID: 32664470 PMCID: PMC7402335 DOI: 10.3390/ijms21144896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury is a leading cause of organ dysfunction and failure in numerous pathological and surgical settings. At the core of this issue lies mitochondrial dysfunction. Hence, strategies that prime mitochondria towards damage resilience might prove applicable in a clinical setting. A promising approach has been to induce a mitohormetic response, removing less capable organelles, and replacing them with more competent ones, in preparation for an insult. Recently, a soluble form of adenylyl cyclase (sAC) has been shown to exist within mitochondria, the activation of which improved mitochondrial function. Here, we sought to understand if inhibiting mitochondrial sAC would elicit mitohormesis and protect the liver from I/R injury. Wistar male rats were pretreated with LRE1, a specific sAC inhibitor, prior to the induction of hepatic I/R injury, after which mitochondria were collected and their metabolic function was assessed. We find LRE1 to be an effective inducer of a mitohormetic response based on all parameters tested, a phenomenon that appears to require the activity of the NAD+-dependent sirtuin deacylase (SirT3) and the subsequent deacetylation of mitochondrial proteins. We conclude that LRE1 pretreatment leads to a mitohormetic response that protects mitochondrial function during I/R injury.
Collapse
|
39
|
Nuamnaichati N, Mangmool S, Chattipakorn N, Parichatikanond W. Stimulation of GLP-1 Receptor Inhibits Methylglyoxal-Induced Mitochondrial Dysfunctions in H9c2 Cardiomyoblasts: Potential Role of Epac/PI3K/Akt Pathway. Front Pharmacol 2020; 11:805. [PMID: 32547400 PMCID: PMC7274035 DOI: 10.3389/fphar.2020.00805] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Accumulation of methylglyoxal (MG) contributes to oxidative stress, apoptosis, and mitochondrial dysfunction, leading to the development of type 2 diabetes and cardiovascular diseases. Inhibition of mitochondrial abnormalities induced by MG in the heart may improve and delay the progression of heart failure. Although glucagon-like peptide-1 receptor (GLP-1R) agonists have been used as anti-diabetic drugs and GLP-1R has been detected in the heart, the cardioprotective effects of GLP-1R agonists on the inhibition of MG-induced oxidative stress and mitochondrial abnormalities have not been elucidated. Stimulation of GLP-1Rs leads to cAMP elevation and subsequently activates PKA- and/or Epac-dependent signaling pathway. However, the signaling pathway involved in the prevention of MG-induced mitochondrial dysfunctions in the heart has not been clarified so far. In the present study, we demonstrated that stimulation of GLP-1Rs with exendin-4 inhibited MG-induced intracellular and mitochondrial reactive oxygen species (ROS) production and apoptosis in H9c2 cardiomyoblasts. GLP-1R stimulation also improved the alterations of mitochondrial membrane potential (MMP) and expressions of genes related to mitochondrial functions and dynamics induced by MG. In addition, stimulation of GLP-1R exhibits antioxidant and antiapoptotic effects as well as the improvement of mitochondrial functions through cAMP/Epac/PI3K/Akt signaling pathway in H9c2 cells. Our study is the first work demonstrating a novel signaling pathway for cardioprotective effects of GLP-1R agonist on inhibition of oxidative stress and prevention of mitochondrial dysfunction. Thus, GLP-1R agonist represents a potential therapeutic target for inhibition of oxidative stress and modulation of mitochondrial functions in the heart.
Collapse
Affiliation(s)
- Narawat Nuamnaichati
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
40
|
Gao S, Zhang Q, Tian C, Li C, Lin Y, Gao W, Wu D, Jiao N, Zhu L, Li W, Zhu R, Wang W, Wang Y. The roles of Qishen granules recipes, Qingre Jiedu, Wenyang Yiqi and Huo Xue, in the treatment of heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112372. [PMID: 31683036 DOI: 10.1016/j.jep.2019.112372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/23/2019] [Accepted: 10/31/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Recipes (Qingre Jiedu (QJ), Wenyang Yiqi (WYYQ) and Huo Xue (HX)) in Qishen granules (QSG) are believed to synergistically exert cardio-protective effects. However, the underlying pattern of each decomposed recipe in QSG and their synergistic effects in the treatment of heart failure (HF) are not clear. OBJECTIVE The purpose of this study is to explore the biological contributions of decomposed recipes to therapeutic effects of QSG and reveal the pharmacological mechanism of QSG in treating HF. MATERIALS AND METHODS The therapeutic effects of QSG or its recipes on heart failure were examined in wet-lab at both transcription and phenotypic level using HF Sprague-Dawley rats. Sequencing and transcriptome analyses were performed using in silico approaches including identification of differentially expressed genes, pathway enrichment and protein-protein interaction network studies. Specially, an optimized in silico quantitative pathway analysis that maximally extracted gene expression information was developed to reveal differentially expressed pathways (DEPs) among various groups, and is publicly available as R package QPA on GitHub (https://github.com/github-gs/QPA). Finally, the HF-related genes predicted using DEP approach were validated by quantitative real-time polymerase chain reaction and western blot. RESULTS Multiple key genes and the associated signaling pathways were shown to be highly relevant for the therapeutic effect of QSG. Decreased expression of Spp1 gene required for inflammatory signaling and profibrotic signaling were observed in failing hearts treated with QJ, WYYQ and HX. Decreased expression of Cx3cr1 gene required for inflammatory signaling was observed in failing hearts treated with WYYQ and HX. Decreased expression of Myc gene required for oxidative stress and Fgfr2 gene required for profibrotic signaling were observed in failing hearts treated with HX and WYYQ, respectively. Increased expression of Adcy1 gene required for cAMP-PKA signaling cascade was observed in failing hearts treated with WYYQ and HX. CONCLUSIONS Our study suggests that QJ, WYYQ and HX recipes in QSG achieve synergistic and complementary therapeutic effects through alleviating inflammatory responses, attenuating ventricular remodeling and enhancing myocardial energy supply.
Collapse
Affiliation(s)
- Sheng Gao
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Qian Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Chuan Tian
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794, United States.
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Yunzheng Lin
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Wenxing Gao
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Dingfeng Wu
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Na Jiao
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China.
| | - Lixin Zhu
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China; Department of Biochemistry, Genome, Environment and Microbiome Community of Excellence, The State University of New York at Buffalo, New York, 14214, United States.
| | - Wenzhe Li
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Ruixin Zhu
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
41
|
Elucidating cyclic AMP signaling in subcellular domains with optogenetic tools and fluorescent biosensors. Biochem Soc Trans 2020; 47:1733-1747. [PMID: 31724693 DOI: 10.1042/bst20190246] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022]
Abstract
The second messenger 3',5'-cyclic nucleoside adenosine monophosphate (cAMP) plays a key role in signal transduction across prokaryotes and eukaryotes. Cyclic AMP signaling is compartmentalized into microdomains to fulfil specific functions. To define the function of cAMP within these microdomains, signaling needs to be analyzed with spatio-temporal precision. To this end, optogenetic approaches and genetically encoded fluorescent biosensors are particularly well suited. Synthesis and hydrolysis of cAMP can be directly manipulated by photoactivated adenylyl cyclases (PACs) and light-regulated phosphodiesterases (PDEs), respectively. In addition, many biosensors have been designed to spatially and temporarily resolve cAMP dynamics in the cell. This review provides an overview about optogenetic tools and biosensors to shed light on the subcellular organization of cAMP signaling.
Collapse
|
42
|
Stone G, Choi A, Meritxell O, Gorham J, Heydarpour M, Seidman CE, Seidman JG, Aranki SF, Body SC, Carey VJ, Raby BA, Stranger BE, Muehlschlegel JD. Sex differences in gene expression in response to ischemia in the human left ventricular myocardium. Hum Mol Genet 2020; 28:1682-1693. [PMID: 30649309 DOI: 10.1093/hmg/ddz014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 01/28/2023] Open
Abstract
Sex differences exist in the prevalence, presentation and outcomes of ischemic heart disease (IHD). Females have higher risk of heart failure post-myocardial infarction relative to males and are two to three times more likely to die after coronary artery bypass grafting surgery. We examined sex differences in human myocardial gene expression in response to ischemia. Left ventricular biopsies from 68 male/46 female patients undergoing aortic valve replacement surgery were obtained at baseline and after a median 74 min of cold cardioplegic arrest/ischemia. Transcriptomes were quantified by RNA-sequencing. Cell-type enrichment analysis was used to estimate the identity and relative proportions of different cell types in each sample. A sex-specific response to ischemia was observed for 271 genes. Notably, the expression FAM5C, PLA2G4E and CYP1A1 showed an increased expression in females compared to males due to ischemia and DIO3, MT1G and CMA1 showed a decreased expression in females compared to males due to ischemia. Functional annotation analysis revealed sex-specific modulation of the oxytocin signaling pathway and common pathway of fibrin clot formation. Expression quantitative trait locus (eQTL) analysis identified variant-by-sex interaction eQTLs, indicative of sex differences in the genotypic effects on gene expression. Cell-type enrichment analysis showed sex-bias in proportion of specific cell types. Common lymphoid progenitor cells and M2 macrophages were found to increase in female samples from pre- to post-ischemia, but no change was observed in male samples. These differences in response to myocardial ischemia provide insight into the sexual dimorphism of IHD and may aid in the development of sex-specific therapies that reduce myocardial injury.
Collapse
Affiliation(s)
- Gregory Stone
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashley Choi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliva Meritxell
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jon G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sary F Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vincent J Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara E Stranger
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
White SM, Avantaggiati ML, Nemazanyy I, Di Poto C, Yang Y, Pende M, Gibney GT, Ressom HW, Field J, Atkins MB, Yi C. YAP/TAZ Inhibition Induces Metabolic and Signaling Rewiring Resulting in Targetable Vulnerabilities in NF2-Deficient Tumor Cells. Dev Cell 2020; 49:425-443.e9. [PMID: 31063758 DOI: 10.1016/j.devcel.2019.04.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 02/04/2019] [Accepted: 04/08/2019] [Indexed: 02/09/2023]
Abstract
Merlin/NF2 is a bona fide tumor suppressor whose mutations underlie inherited tumor syndrome neurofibromatosis type 2 (NF2), as well as various sporadic cancers including kidney cancer. Multiple Merlin/NF2 effector pathways including the Hippo-YAP/TAZ pathway have been identified. However, the molecular mechanisms underpinning the growth and survival of NF2-mutant tumors remain poorly understood. Using an inducible orthotopic kidney tumor model, we demonstrate that YAP/TAZ silencing is sufficient to induce regression of pre-established NF2-deficient tumors. Mechanistically, YAP/TAZ depletion diminishes glycolysis-dependent growth and increases mitochondrial respiration and reactive oxygen species (ROS) buildup, resulting in oxidative-stress-induced cell death when challenged by nutrient stress. Furthermore, we identify lysosome-mediated cAMP-PKA/EPAC-dependent activation of RAF-MEK-ERK signaling as a resistance mechanism to YAP/TAZ inhibition. Finally, unbiased analysis of TCGA primary kidney tumor transcriptomes confirms a positive correlation of a YAP/TAZ signature with glycolysis and inverse correlations with oxidative phosphorylation and lysosomal gene expression, supporting the clinical relevance of our findings.
Collapse
Affiliation(s)
- Shannon M White
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | | | - Ivan Nemazanyy
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cristina Di Poto
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Yang Yang
- Department of Systems Pharmacology and Translational Therapeutics, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mario Pende
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Jeffery Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael B Atkins
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Chunling Yi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
44
|
Jayarajan V, Appukuttan A, Aslam M, Reusch P, Regitz-Zagrosek V, Ladilov Y. Regulation of AMPK activity by type 10 adenylyl cyclase: contribution to the mitochondrial biology, cellular redox and energy homeostasis. Cell Mol Life Sci 2019; 76:4945-4959. [PMID: 31172217 PMCID: PMC11105217 DOI: 10.1007/s00018-019-03152-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 01/28/2023]
Abstract
The downregulation of AMP-activated protein kinase (AMPK) activity contributes to numerous pathologies. Recent reports suggest that the elevation of cellular cAMP promotes AMPK activity. However, the source of the cAMP pool that controls AMPK activity remains unknown. Mammalian cells possess two cAMP sources: membrane-bound adenylyl cyclase (tmAC) and intracellularly localized, type 10 soluble adenylyl cyclase (sAC). Due to the localization of sAC and AMPK in similar intracellular compartments, we hypothesized that sAC may control AMPK activity. In this study, sAC expression and activity were manipulated in H9C2 cells, adult rat cardiomyocytes or endothelial cells. sAC knockdown depleted the cellular cAMP content and decreased AMPK activity in an EPAC-dependent manner. Functionally, sAC knockdown reduced cellular ATP content, increased mitochondrial ROS formation and led to mitochondrial depolarization. Furthermore, sAC downregulation led to EPAC-dependent mitophagy disturbance, indicated by an increased mitochondrial mass and unaffected mitochondrial biogenesis. Consistently, sAC overexpression or stimulation with bicarbonate significantly increased AMPK activity and cellular ATP content. In contrast, tmAC inhibition or stimulation produced no effect on AMPK activity. Therefore, the sAC-EPAC axis may regulate basal and induced AMPK activity and support mitophagy, cellular energy and redox homeostasis. The study argues for sAC as a potential target in treating pathologies associated with AMPK downregulation.
Collapse
Affiliation(s)
- Vignesh Jayarajan
- Charité, Universitätsmedizin Berlin, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin Partner Site, Berlin, Germany
- Department of Clinical Pharmacology, Ruhr-University Bochum, Bochum, Germany
| | - Avinash Appukuttan
- Department of Clinical Pharmacology, Ruhr-University Bochum, Bochum, Germany
| | - Muhammad Aslam
- Internal Medicine I/Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany
- Experimental Cardiology, Justus Liebig University Giessen, Giessen, Germany
| | - Peter Reusch
- Department of Clinical Pharmacology, Ruhr-University Bochum, Bochum, Germany
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin Partner Site, Berlin, Germany
| | - Yury Ladilov
- Charité, Universitätsmedizin Berlin, Institute of Gender in Medicine, Center for Cardiovascular Research, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Berlin Partner Site, Berlin, Germany.
| |
Collapse
|
45
|
The Epac1 Protein: Pharmacological Modulators, Cardiac Signalosome and Pathophysiology. Cells 2019; 8:cells8121543. [PMID: 31795450 PMCID: PMC6953115 DOI: 10.3390/cells8121543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022] Open
Abstract
The second messenger 3′,5′-cyclic adenosine monophosphate (cAMP) is one of the most important signalling molecules in the heart as it regulates many physiological and pathophysiological processes. In addition to the classical protein kinase A (PKA) signalling route, the exchange proteins directly activated by cAMP (Epac) mediate the intracellular functions of cAMP and are now emerging as a new key cAMP effector in cardiac pathophysiology. In this review, we provide a perspective on recent advances in the discovery of new chemical entities targeting the Epac1 isoform and illustrate their use to study the Epac1 signalosome and functional characterisation in cardiac cells. We summarize the role of Epac1 in different subcompartments of the cardiomyocyte and discuss how cAMP–Epac1 specific signalling networks may contribute to the development of cardiac diseases. We also highlight ongoing work on the therapeutic potential of Epac1-selective small molecules for the treatment of cardiac disorders.
Collapse
|
46
|
Espejo MS, Orlowski A, Ibañez AM, Di Mattía RA, Velásquez FC, Rossetti NS, Ciancio MC, De Giusti VC, Aiello EA. The functional association between the sodium/bicarbonate cotransporter (NBC) and the soluble adenylyl cyclase (sAC) modulates cardiac contractility. Pflugers Arch 2019; 472:103-115. [PMID: 31754830 DOI: 10.1007/s00424-019-02331-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/15/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022]
Abstract
The soluble adenylyl cyclase (sAC) was identified in the heart as another source of cyclic AMP (cAMP). However, its cardiac physiological function is unknown. On the other hand, the cardiac Na+/HCO3- cotransporter (NBC) promotes the cellular co-influx of HCO3- and Na+. Since sAC activity is regulated by HCO3-, our purpose was to investigate the potential functional relationship between NBC and sAC in the cardiomyocyte. Rat ventricular myocytes were loaded with Fura-2, Fluo-3, or BCECF to measure Ca2+ transient (Ca2+i) by epifluorescence, Ca2+ sparks frequency (CaSF) by confocal microscopy, or intracellular pH (pHi) by epifluorescence, respectively. Sarcomere or cell shortening was measured with a video camera as an index of contractility. The NBC blocker S0859 (10 μM), the selective inhibitor of sAC KH7 (1 μM), and the PKA inhibitor H89 (0.1 μM) induced a negative inotropic effect which was associated with a decrease in Ca2+i. Since PKA increases Ca2+ release through sarcoplasmic reticulum RyR channels, CaSF was measured as an index of RyR open probability. The generation of CaSF was prevented by KH7. Finally, we investigated the potential role of sAC activation on NBC activity. NBC-mediated recovery from acidosis was faster in the presence of KH7 or H89, suggesting that the pathway sAC-PKA is negatively regulating NBC function, consistent with a negative feedback modulation of the HCO3- influx that activates sAC. In summary, the results demonstrated that the complex NBC-sAC-PKA plays a relevant role in Ca2+ handling and basal cardiac contractility.
Collapse
Affiliation(s)
- María S Espejo
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Alejandro M Ibañez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Romina A Di Mattía
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Fernanda Carrizo Velásquez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Noelia S Rossetti
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - María C Ciancio
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Verónica C De Giusti
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina.
| | - Ernesto A Aiello
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina.
| |
Collapse
|
47
|
Particulate Matter 2.5 Mediates Cutaneous Cellular Injury by Inducing Mitochondria-Associated Endoplasmic Reticulum Stress: Protective Effects of Ginsenoside Rb1. Antioxidants (Basel) 2019; 8:antiox8090383. [PMID: 31505827 PMCID: PMC6769862 DOI: 10.3390/antiox8090383] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/25/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022] Open
Abstract
The prevalence of fine particulate matter-induced harm to the human body is increasing daily. The aim of this study was to elucidate the mechanism by which particulate matter 2.5 (PM2.5) induces damage in human HaCaT keratinocytes and normal human dermal fibroblasts, and to evaluate the preventive capacity of the ginsenoside Rb1. PM2.5 induced oxidative stress by increasing the production of reactive oxygen species, leading to DNA damage, lipid peroxidation, and protein carbonylation; this effect was inhibited by ginsenoside Rb1. Through gene silencing of endoplasmic reticulum (ER) stress-related genes such as PERK, IRE1, ATF, and CHOP, and through the use of the ER stress inhibitor tauroursodeoxycholic acid (TUDCA), it was demonstrated that PM2.5-induced ER stress also causes apoptosis and ultimately leads to cell death; however, this phenomenon was reversed by ginsenoside Rb1. We also found that TUDCA partially restored the production of ATP that was inhibited by PM2.5, and its recovery ability was significantly higher than that of ginsenoside Rb1, indicating that the process of ER stress leading to cell damage may also occur via the mitochondrial pathway. We concluded that ER stress acts alone or via the mitochondrial pathway in the induction of cell damage by PM2.5, and that ginsenoside Rb1 blocks this process. Ginsenoside Rb1 shows potential for use in skin care products to protect the skin against damage by fine particles.
Collapse
|
48
|
Palmeira CM, Teodoro JS, Amorim JA, Steegborn C, Sinclair DA, Rolo AP. Mitohormesis and metabolic health: The interplay between ROS, cAMP and sirtuins. Free Radic Biol Med 2019; 141:483-491. [PMID: 31349039 PMCID: PMC6718302 DOI: 10.1016/j.freeradbiomed.2019.07.017] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
The key role of mitochondria in oxidative metabolism and redox homeostasis explains the link between mitochondrial dysfunction and the development of metabolic disorders. Mitochondria's highly dynamic nature, based on alterations in biogenesis, mitophagy, fusion and fission, allows adjusting sequential redox reactions of the electron transport chain (ETC) and dissipation of the membrane potential by ATP synthase, to different environmental cues. With reactive oxygen species being an inevitable by-product of oxidative phosphorylation (OXPHOS), alterations on mitochondrial oxidative rate with a consequent excessive load of reactive oxygen species have been traditionally associated with pathological conditions. However, reactive oxygen species have also been suggested as promoters of mitohormesis, a process in which low, non-cytotoxic concentrations of reactive oxygen species promote mitochondrial homeostasis. Therefore, signaling systems involved in the regulation of mitochondrial homeostasis are attractive candidates for drug development for metabolic diseases triggered by mitochondrial dysfunction. Reversible phosphorylation downstream the cyclic AMP (cAMP) signaling cascade and deacetylation mediated by sirtuins are recognized as major mitochondrial regulators.
Collapse
Affiliation(s)
- Carlos Marques Palmeira
- Department of Life Sciences, University of Coimbra, Portugal; Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - João Soeiro Teodoro
- Department of Life Sciences, University of Coimbra, Portugal; Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - João Alves Amorim
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; IIIUC - Institute of Interdisciplinary Research, University of Coimbra, Portugal; Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, 95440, Bayreuth, Germany
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Anabela Pinto Rolo
- Department of Life Sciences, University of Coimbra, Portugal; Center for Neurosciences and Cell Biology, University of Coimbra, Portugal.
| |
Collapse
|
49
|
Talmon M, Rossi S, Lim D, Pollastro F, Palattella G, Ruffinatti FA, Marotta P, Boldorini R, Genazzani AA, Fresu LG. Absinthin, an agonist of the bitter taste receptor hTAS2R46, uncovers an ER-to-mitochondria Ca 2+-shuttling event. J Biol Chem 2019; 294:12472-12482. [PMID: 31248983 DOI: 10.1074/jbc.ra119.007763] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/17/2019] [Indexed: 01/04/2023] Open
Abstract
Type 2 taste receptors (TAS2R) are G protein-coupled receptors first described in the gustatory system, but have also been shown to have extraoral localizations, including airway smooth muscle (ASM) cells, in which TAS2R have been reported to induce relaxation. TAS2R46 is an unexplored subtype that responds to its highly specific agonist absinthin. Here, we first demonstrate that, unlike other bitter-taste receptor agonists, absinthin alone (1 μm) in ASM cells does not induce Ca2+ signals but reduces histamine-induced cytosolic Ca2+ increases. To investigate this mechanism, we introduced into ASM cells aequorin-based Ca2+ probes targeted to the cytosol, subplasma membrane domain, or the mitochondrial matrix. We show that absinthin reduces cytosolic histamine-induced Ca2+ rises and simultaneously increases Ca2+ influx into mitochondria. We found that this effect is inhibited by the potent human TAS2R46 (hTAS2R46) antagonist 3β-hydroxydihydrocostunolide and is no longer evident in hTAS2R46-silenced ASM cells, indicating that it is hTAS2R46-dependent. Furthermore, these changes were sensitive to the mitochondrial uncoupler carbonyl cyanide p-(trifluoromethoxy)phenyl-hydrazone (FCCP); the mitochondrial calcium uniporter inhibitor KB-R7943 (carbamimidothioic acid); the cytoskeletal disrupter latrunculin; and an inhibitor of the exchange protein directly activated by cAMP (EPAC), ESI-09. Similarly, the β2 agonist salbutamol also could induce Ca2+ shuttling from cytoplasm to mitochondria, suggesting that this new mechanism might be generalizable. Moreover, forskolin and an EPAC activator mimicked this effect in HeLa cells. Our findings support the hypothesis that plasma membrane receptors can positively regulate mitochondrial Ca2+ uptake, adding a further facet to the ability of cells to encode complex Ca2+ signals.
Collapse
Affiliation(s)
- Maria Talmon
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17-28100 Novara, Italy
| | - Silvia Rossi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17-28100 Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Via Bovio, 6-28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Via Bovio, 6-28100 Novara, Italy
| | - Gioele Palattella
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17-28100 Novara, Italy
| | - Federico A Ruffinatti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Via Bovio, 6-28100 Novara, Italy
| | - Patrizia Marotta
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Via Bovio, 6-28100 Novara, Italy
| | - Renzo Boldorini
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17-28100 Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Via Bovio, 6-28100 Novara, Italy.
| | - Luigia G Fresu
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Via Solaroli, 17-28100 Novara, Italy.
| |
Collapse
|
50
|
STIM2 knockdown protects against ischemia/reperfusion injury through reducing mitochondrial calcium overload and preserving mitochondrial function. Life Sci 2019; 247:116560. [PMID: 31200000 DOI: 10.1016/j.lfs.2019.116560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 01/03/2023]
Abstract
Mitochondrial dysfunction caused by calcium overload is a vital factor for mediating cardiomyocyte death following ischemia/reperfusion (I/R) injury. The stromal interactive molecule 2 (STIM2) is a calcium sensor protein that regulates the store-operated calcium entry (SOCE). Whereas, whether STIM2 is associated with I/R injury remains largely unclear. We report here that STIM2, but not its homologue STIM1, is upregulated in cultured H9c2 cells, a cell model for cardiomyocytes, following I/R injury. In addition, the knockdown of STIM2, but not STIM1, reduces H9c2 cell apoptosis following I/R injury, and similar results were obtained in primary neonatal cardiomyocytes. This anti-apoptotic effect could be attributed to the inhibited activation of mitochondrial apoptosis pathway. Moreover, STIM2 knockdown reduces ER calcium release and simultaneously alleviates mitochondrial calcium overload in H9c2 cells following I/R injury. Furthermore, STIM2 knockdown decreases mitochondrial injury and preserves mitochondrial function following I/R injury. Collectively, these results suggest that the protective role of STIM2 knockdown against I/R injury in cardiomyocytes is associated with the reduced mitochondrial calcium overload and preserved mitochondrial function. Hence, our study may provide a novel insight into the regulation of mitochondrial-mediated cardiomyocyte apoptosis following I/R injury.
Collapse
|