1
|
Yang X, Liu Y, Cao J, Wu C, Tang L, Bian W, Chen Y, Yu L, Wu Y, Li S, Shen Y, Xia J, Du J. Targeting epigenetic and post-translational modifications of NRF2: key regulatory factors in disease treatment. Cell Death Discov 2025; 11:189. [PMID: 40258841 PMCID: PMC12012105 DOI: 10.1038/s41420-025-02491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor involved in regulating cellular antioxidant defense and detoxification mechanisms. It mitigates oxidative stress and xenobiotic-induced damage by inducing the expression of cytoprotective enzymes, including HO-1 and NQO1. NRF2 also modulates inflammatory responses by inhibiting pro-inflammatory genes and mediates cell death pathways, including apoptosis and ferroptosis. Targeting NRF2 offers potential therapeutic avenues for treating various diseases. NRF2 is regulated through two principal mechanisms: post-translational modifications (PTMs) and epigenetic alterations. PTMs, including phosphorylation, ubiquitination, and acetylation, play a pivotal role in modulating NRF2's stability, activity, and subcellular localization, thereby precisely controlling its function in the antioxidant response. For instance, ubiquitination can lead to NRF2 degradation and reduced antioxidant activity, while deubiquitination enhances its stability and function. Epigenetic modifications, such as DNA methylation, histone modifications, and interactions with non-coding RNAs (e.g., MALAT1, PVT1, MIR4435-2HG, and TUG1), are essential for regulating NRF2 expression by modulating chromatin architecture and gene accessibility. This paper systematically summarizes the molecular mechanisms by which PTMs and epigenetic alterations regulate NRF2, and elucidates its critical role in cellular defense and disease. By analyzing the impact of PTMs, such as phosphorylation, ubiquitination, and acetylation, as well as DNA methylation, histone modifications, and non-coding RNA interactions on NRF2 stability, activity, and expression, the study reveals the complex cellular protection network mediated by NRF2. Furthermore, the paper explores how these regulatory mechanisms affect NRF2's roles in oxidative stress, inflammation, and cell death, identifying novel therapeutic targets and strategies. This provides new insights into the treatment of NRF2-related diseases, such as cancer, neurodegenerative disorders, and metabolic syndrome. This research deepens our understanding of NRF2's role in cellular homeostasis and lays the foundation for the development of NRF2-targeted therapies.
Collapse
Affiliation(s)
- Xinyi Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lusheng Tang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenxia Bian
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Sainan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
2
|
Fu X, Zhang G, Hou Z, Fu T, Cui G. PKN2 enhances the immunosuppressive activity of polymorphonuclear myeloid-derived suppressor cells in esophageal carcinoma by mediating fatty acid oxidation. Mol Med 2025; 31:92. [PMID: 40069590 PMCID: PMC11900251 DOI: 10.1186/s10020-025-01132-6] [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: 09/18/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) in tumor microenvironment reduce the efficacy of immunotherapy. PKN2 plays a role in colon cancer, but its function in esophageal cancer (EC) remains unclear. This study investigated PKN2 expression in MDSCs derived from EC tissues and determined whether PKN2 regulates immunosuppressive activity of MDSCs by mediating fatty acid oxidation (FAO). MATERIALS AND METHODS PKN2 expression was determined in GEO database, EC patients, and 4-NQO-induced EC mice, as well as in different types of immune cells. The effect of PKN2 on the function of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) was investigated by co-culture of PMN-MDSCs and CD4+/CD8+ T cells. The co-culture of patient-derived organoids and autologous immune cells was performed to observe the effect of PKN2 on the immunosuppressive function of PMN-MDSCs. RESULTS PKN2 is highly expressed in EC tumor tissues compared to normal tissues, especially in tumor-infiltrated PMN-MDSCs. Overexpressing PKN2 in PMN-MDSCs contributes to the immunosuppressive activity of PMN-MDSCs in vitro. PKN2-overexpressing PMN-MDSCs inhibited the killing ability of cytotoxic T lymphocytes and promoted EC organoid growth. PKN2 promotes FAO in PMN-MDSCs via CPT1B (a key enzyme of FAO). Mechanistically, PKN2 promotes CPT1B transcription by upregulating STAT3 phosphorylation. CONCLUSIONS PKN2 expression was increased in PMN-MDSCs derived from human and mouse EC tissues. PKN2 plays a role in enhancing the immunosuppressive activity of PMN-MDSCs by facilitating STAT3 phosphorylation and CPT1B transcription, which in turn leads to increased CPT1B-mediated FAO in PMN-MDSCs. Targeted inhibition of PKN2 is expected to improve immunotherapeutic efficacy in EC patients.
Collapse
Affiliation(s)
- Xiao Fu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 East Jianshe Road, Zhengzhou, 450052, China
| | - GuoQing Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 East Jianshe Road, Zhengzhou, 450052, China
| | - ZhiChao Hou
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 East Jianshe Road, Zhengzhou, 450052, China
| | - TingTing Fu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - GuangHui Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 East Jianshe Road, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Wei S, Shen R, Lu X, Li X, He L, Zhang Y, Yang J, Shu Z, Huang X. Mendelian randomization provides a multi-omics perspective on the regulation of genes involved in ribosome biogenesis in relation to cardiac structure and function. Clin Epigenetics 2025; 17:42. [PMID: 40045424 PMCID: PMC11884004 DOI: 10.1186/s13148-025-01850-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Ribosome biogenesis (RiboSis) is a complex process for generating ribosomes, the cellular machinery responsible for protein synthesis. Dysfunctional RiboSis can disrupt cardiac structure and function, contributing to cardiovascular diseases. This study employed a Mendelian randomization (MR) approach, integrating multi-omics data, to investigate the relationship between RiboSis-related genes and standard cardiac structure and function. METHODS We utilized summary stats for methylation, RNA splicing, and gene expression, and UK Biobank cardiopulm MRI genetic associations (N = 41,135). MR evaluated RiboSis gene features against traits, complemented by hypothesis prioritization for multi-trait colocalization (HyPrColoc) and colocalization. Composite scores ranked RiboSis genes, and phenome-wide association study (PheWAS) with scQTLbase instrumental variables (IVs) confirmed results. RESULTS We identified 15 RiboSis-related genes: HEATR1, SENP3, ERI1, ERCC2, TSR1, UTP11, DDX17, SMARCB1, NIP7, ERAL1, NOP56, RPL10A, EIF6, EXOSC9, and NOP58. Notably, HEATR1 and SENP3 were ranked in the top quartile (Q1), scoring 25. In validation cohort, 12 genes associated with cardiac structures, functions, diseases. Only ERAL1, TSR1, and NIP7 lacked significant associations with cardiac traits. CONCLUSION Our multi-omics MR analysis identified 15 RiboSis-related genes associated with cardiac risk, with 12 further validated through gene set enrichment analysis. These findings suggest a link between RiboSis and cardiac health, enhancing understanding of cardiac disease mechanisms.
Collapse
Affiliation(s)
- Shuxu Wei
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
| | - Ronghuai Shen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
| | - Xiaojia Lu
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
| | - Xinyi Li
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
| | - Lingbin He
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China
| | - Youti Zhang
- Department of Cardiology, Jie xi's People Hospital, Jieyang, Guangdong, China
| | - Jiahang Yang
- Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Zhouwu Shu
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China.
| | - Xianxi Huang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China.
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, No.57, Changping Road, Shantou, 515041, China.
- Department of Cardiology, Jie xi's People Hospital, Jieyang, Guangdong, China.
| |
Collapse
|
4
|
Schopohl B, Kohlhaas M, Nickel AG, Schiuma AF, Maas SL, van der Vorst EPC, Shia YX, Maack C, Steffens S, Puhl SL. Gpr55 deficiency crucially alters cardiomyocyte homeostasis and counteracts angiotensin II induced maladaption in female mice. Br J Pharmacol 2025; 182:670-691. [PMID: 39428581 DOI: 10.1111/bph.17350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/24/2024] [Accepted: 07/20/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Cannabis stimulates several G-protein-coupled-receptors and causes bradycardia and hypotension upon sustained consumption. Moreover, in vitro studies suggest an interference of cannabinoid-signalling with cardiomyocyte contractility and hypertrophy. We aimed at revealing a functional contribution of the cannabinoid-sensitive receptor GPR55 to cardiomyocyte homeostasis and neurohumorally induced hypertrophy in vivo. EXPERIMENTAL APPROACH Gpr55-/- and wild-type (WT) mice were characterized after 28-day angiotensin II (AngII; 1·μg·kg-1 min-1) or vehicle infusion. In isolated adult Gpr55-/- and WT cardiomyocytes, mitochondrial function was assessed under naïve conditions, while cytosolic Ca2+ handling was additionally determined following application of the selective GPR55 antagonist CID16020046. KEY RESULTS Gpr55 deficiency did not affect angiotensin II (AngII) mediated hypertrophic growth, yet, especially in females, it alleviated maladaptive pro-hypertrophic and -inflammatory gene expression and improved inotropy and adrenergic responsiveness compared to WT. In-depth analyses implied increased cytosolic Ca2+ concentrations and transient amplitudes, and accelerated sarcomere contraction kinetics in Gpr55-/- myocytes, which could be mimicked by GPR55 blockade with CID16020046 in female WT cells. Moreover, Gpr55 deficiency up-regulated factors involved in glucose and fatty acid transport independent of the AngII challenge, accelerated basal mitochondrial respiration and reduced basal protein kinase (PK) A, G and C activity and phospholemman (PLM) phosphorylation. CONCLUSIONS AND IMPLICATIONS Our study suggests GPR55 as crucial regulator of cardiomyocyte hypertrophy and homeostasis presumably by regulating PKC/PKA-PLM and PKG signalling, and identifies the receptor as potential target to counteract maladaptation, adrenergic desensitization and metabolic shifts as unfavourable features of the hypertrophied heart in females.
Collapse
Affiliation(s)
- Brigitte Schopohl
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Alexander G Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | | | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany
| | - Yi Xuan Shia
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
- Medical Clinic I, University Clinic Würzburg, Würzburg, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sarah-Lena Puhl
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Bogomolovas J, Chen J. Illuminating understudied kinases: a generalizable biosensor development method applied to protein kinase N. Commun Biol 2025; 8:109. [PMID: 39843538 PMCID: PMC11754634 DOI: 10.1038/s42003-025-07510-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
Protein kinases play crucial roles in regulating cellular processes, making real-time visualization of their activity essential for understanding signaling dynamics. While genetically encoded fluorescent biosensors have emerged as powerful tools for studying kinase activity, their development for many kinases remains challenging due to the lack of suitable substrate peptides. Here, we present a novel approach for identifying peptide substrates and demonstrate its effectiveness by developing a biosensor for Protein Kinase N (PKN) activity. Our method identified a new PKN substrate peptide that we optimized for use in a fluorescent biosensor design. The resulting biosensor shows specificity for PKN family kinases and can detect both overexpressed and endogenous PKN activity in live cells. Importantly, our biosensor revealed sustained basal PKN2 activity at the plasma membrane, identifying it as a PKN2 activity hotspot. This work not only provides a valuable tool for studying PKN signaling but also demonstrates a promising strategy for developing biosensors for other understudied kinases, potentially expanding our ability to monitor kinase activity across the human kinome.
Collapse
Affiliation(s)
| | - Ju Chen
- Department of Medicine, UCSD, La Jolla, CA, USA.
| |
Collapse
|
6
|
Song Y, Li F, Ali M, Li X, Zhang X, Ahmed ZFR. Advances in Protein Kinase Regulation of Stress Responses in Fruits and Vegetables. Int J Mol Sci 2025; 26:768. [PMID: 39859482 PMCID: PMC11765796 DOI: 10.3390/ijms26020768] [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: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Fruits and vegetables (F&Vs) are essential in daily life and industrial production. These perishable produces are vulnerable to various biotic and abiotic stresses during their growth, postharvest storage, and handling. As the fruit detaches from the plant, these stresses become more intense. This unique biological process involves substantial changes in a variety of cellular metabolisms. To counter these stresses, plants have evolved complex physiological defense mechanisms, including regulating cellular activities through reversible phosphorylation of proteins. Protein kinases, key components of reversible protein phosphorylation, facilitate the transfer of the γ-phosphate group from adenosine triphosphate (ATP) to specific amino acid residues on substrates. This phosphorylation alters proteins' structure, function, and interactions, thereby playing a crucial role in regulating cellular activity. Recent studies have identified various protein kinases in F&Vs, underscoring their significant roles in plant growth, development, and stress responses. This article reviews the various types of protein kinases found in F&Vs, emphasizing their roles and regulatory mechanisms in managing stress responses. This research sheds light on the involvement of protein kinases in metabolic regulation, offering key insights to advance the quality characteristics of F&Vs.
Collapse
Affiliation(s)
- Yanan Song
- College of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; (Y.S.); (F.L.); (M.A.); (X.L.)
| | - Fujun Li
- College of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; (Y.S.); (F.L.); (M.A.); (X.L.)
| | - Maratab Ali
- College of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; (Y.S.); (F.L.); (M.A.); (X.L.)
- School of Food and Agricultural Sciences, University of Management and Technology, Lahore 54000, Pakistan
| | - Xiaoan Li
- College of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; (Y.S.); (F.L.); (M.A.); (X.L.)
| | - Xinhua Zhang
- College of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; (Y.S.); (F.L.); (M.A.); (X.L.)
| | - Zienab F. R. Ahmed
- Integrative Agriculture Department, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| |
Collapse
|
7
|
Kakkar C, Sharma V, Mannan A, Gupta G, Singh S, Kumar P, Dua K, Kaur A, Singh S, Dhiman S, Singh TG. Diabetic Cardiomyopathy: An Update on Emerging Pathological Mechanisms. Curr Cardiol Rev 2025; 21:88-107. [PMID: 39501954 DOI: 10.2174/011573403x331870241025094307] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 04/25/2025] Open
Abstract
Diabetic Cardiomyopathy (DCM) is a notable consequence of diabetes mellitus, distinguished by cardiac dysfunction that occurs separately from coronary artery disease or hypertension. A recent study has revealed an intricate interaction of pathogenic processes that contribute to DCM. Important aspects involve the dysregulation of glucose metabolism, resulting in heightened oxidative stress and impaired mitochondrial function. In addition, persistent high blood sugar levels stimulate inflammatory pathways, which contribute to the development of heart fibrosis and remodelling. Additionally, changes in the way calcium is managed and the presence of insulin resistance are crucial factors in the formation and advancement of DCM. This may be due to the involvement of many molecular mechanistic pathways such as NLRP3, NF-κB, PKC, and MAPK with their downstream associated signaling pathways. Gaining a comprehensive understanding of these newly identified pathogenic pathways is crucial in order to design precise therapy approaches that can enhance the results for individuals suffering from diabetes. In addition, this review offers an in-depth review of not just pathogenic pathways and molecular mechanistic pathways but also diagnostic methods, treatment options, and clinical trials.
Collapse
Affiliation(s)
- Chirag Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Gaurav Gupta
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, 346, United Arab Emirates
| | - Sachin Singh
- Lovely Institute of Technology (Pharmacy), Lovely Professional University, Phagwara, Punjab, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Broadway, P.O. Box 123, Ultimo, NSW, 2007, Australia
| | - Puneet Kumar
- Department of Pharmacology, School of Pharmaceutical Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Broadway, P.O. Box 123, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
8
|
Liu DZ, Luo XZ, Lu CH, Feng YY, Chen DX, Zeng ZY, Huang F. Y4 RNA fragments from cardiosphere-derived cells ameliorate diabetic myocardial ischemia‒reperfusion injury by inhibiting protein kinase C β-mediated macrophage polarization. Cardiovasc Diabetol 2024; 23:202. [PMID: 38867293 PMCID: PMC11170846 DOI: 10.1186/s12933-024-02247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024] Open
Abstract
The specific pathophysiological pathways through which diabetes exacerbates myocardial ischemia/reperfusion (I/R) injury remain unclear; however, dysregulation of immune and inflammatory cells, potentially driven by abnormalities in their number and function due to diabetes, may play a significant role. In the present investigation, we simulated myocardial I/R injury by inducing ischemia through ligation of the left anterior descending coronary artery in mice for 40 min, followed by reperfusion for 24 h. Previous studies have indicated that protein kinase Cβ (PKCβ) is upregulated under hyperglycemic conditions and is implicated in the development of various diabetic complications. The Y4 RNA fragment is identified as the predominant small RNA component present in the extracellular vesicles of cardio sphere-derived cells (CDCs), exhibiting notable anti-inflammatory properties in the contexts of myocardial infarction and cardiac hypertrophy. Our investigation revealed that the administration of Y4 RNA into the ventricular cavity of db/db mice following myocardial I/R injury markedly enhanced cardiac function. Furthermore, Y4 RNA was observed to facilitate M2 macrophage polarization and interleukin-10 secretion through the suppression of PKCβ activation. The mechanism by which Y4 RNA affects PKCβ by regulating macrophage activation within the inflammatory environment involves the inhibition of ERK1/2 phosphorylation In our study, the role of PKCβ in regulating macrophage polarization during myocardial I/R injury was investigated through the use of PKCβ knockout mice. Our findings indicate that PKCβ plays a crucial role in modulating the inflammatory response associated with macrophage activation in db/db mice experiencing myocardial I/R, with a notable exacerbation of this response observed upon significant upregulation of PKCβ expression. In vitro studies further elucidated the protective mechanism by which Y4 RNA modulates the PKCβ/ERK1/2 signaling pathway to induce M2 macrophage activation. Overall, our findings suggest that Y4 RNA plays an anti-inflammatory role in diabetic I/R injury, suggesting a novel therapeutic approach for managing myocardial I/R injury in diabetic individuals.
Collapse
Affiliation(s)
- De-Zhao Liu
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xiao-Zhi Luo
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Chuang-Hong Lu
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yang-Yi Feng
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - De-Xin Chen
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhi-Yu Zeng
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Feng Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
9
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
10
|
Tang X, Shen Y, Lu Y, He W, Nie Y, Fang X, Cai J, Si X, Zhu Y. Identification and validation of pyroptosis-related genes as potential biomarkers for hypertrophic cardiomyopathy: A comprehensive bioinformatics analysis. Medicine (Baltimore) 2024; 103:e36799. [PMID: 38277535 PMCID: PMC10817039 DOI: 10.1097/md.0000000000036799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/06/2023] [Indexed: 01/28/2024] Open
Abstract
Pyroptosis plays a key role in the death of cells including cardiomyocytes, and it is associated with a variety of cardiovascular diseases. However, the role of pyroptosis-related genes (PRGs) in hypertrophic cardiomyopathy (HCM) is not well characterized. This study aimed to identify key biomarkers and explore the molecular mechanisms underlying the functions of the PRGs in HCM. The differentially expressed genes were identified by GEO2R, and the differentially expressed pyroptosis-related genes (DEPRGs) of HCM were identified by combining with PRGs. Enrichment analysis was performed using the "clusterProfiler" package of the R software. Protein-protein interactions (PPI) network analysis was performed using the STRING database, and hub genes were screened using cytoHubba. TF-miRNA coregulatory networks and protein-chemical interactions were analyzed using NetworkAnalyst. RT-PCR/WB was used for expression validation of HCM diagnostic markers. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western Blot (WB) were used to measure and compare the expression of the identified genes in the cardiac hypertrophy model and the control group. A total of 20 DEPRGs were identified, which primarily showed enrichment for the positive regulation of cytokine production, regulation of response to biotic stimulus, tumor necrosis factor production, and other biological processes. These processes primarily involved pathways related to Renin-angiotensin system, Adipocytokine signaling pathway and NF-kappa B signaling pathway. Then, a PPI network was constructed, and 8 hub genes were identified. After verification analysis, the finally identified HCM-related diagnostic markers were upregulated gene protein tyrosine phosphatase non-receptor type 11 (PTPN11), downregulated genes interleukin-1 receptor-associated kinase 3 (IRAK3), and annexin A2 (ANXA2). Further GSEA analysis revealed these 3 biomarkers primarily related to cardiac muscle contraction, hypertrophic cardiomyopathy, fatty acid degradation and ECM - receptor interaction. Moreover, we also elucidated the interaction network of these biomarkers with the miRNA network and known compounds, respectively. RT-PCR/WB results indicated that PTPN11 expression was significantly increased, and IRAK3 and ANXA2 expressions were significantly decreased in HCM. This study identified PTPN11, IRAK3, and ANXA2 as pyroptosis-associated biomarkers of HCM, with the potential to reveal the development and pathogenesis of HCM and could be potential therapeutic targets.
Collapse
Affiliation(s)
- Xin Tang
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yi Shen
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Lu
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Wanya He
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Ying Nie
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Xue Fang
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jinghui Cai
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Xiaoyun Si
- Department of Cardiovascular Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Zhu
- School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Li W, Shen MY, Liu RB, Zhang JY, Li RY, Wang GG. Deletion of protein kinase C θ attenuates hepatic ischemia/reperfusion injury and further elucidates its mechanism in pathophysiology. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1323-1330. [PMID: 39229579 PMCID: PMC11366945 DOI: 10.22038/ijbms.2024.77365.16730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/06/2024] [Indexed: 09/05/2024]
Abstract
Objectives Hepatic ischemia-reperfusion (HIR) is a severe process in pathophysiology that occurs clinically in hepatectomy, and hepatic transplantations. The present study aimed to investigate the effect of PKC θ deletion against HIR injury and elucidate its mechanism in pathophysiology. Materials and Methods HIR injury was induced in wild-type and PKC θ deletion mice treated with or without heme. The ALT and AST levels were determined to evaluate liver function. HIR injury was observed via histological examination. Oxidative stress and inflammatory response markers, and their signaling pathways were detected. Results The study found that PKC θ knockout decreased serum AST and ALT levels when compared to the WT mice. Furthermore, heme treatment significantly reduced the ALT and AST levels of the PKC θ deletion mice compared with the untreated PKC θ deletion mice. PKC θ deletion markedly elevated superoxide dismutase activity in the liver tissue, reduced malondialdehyde content in the tissue, and the serum TNF-α and IL-6 levels compared with the WT mice. Heme treatment was observed to elevate the activity of SOD and reduced MDA content and serum of TNF-α and IL 6 in the PKC θ deletion animals. Meanwhile, heme treatment increased HO-1 and Nrf 2 protein expression, and reduced the levels of TLR4, phosphorylated NF-κB, and IKB-α. Conclusion These findings suggested that PKC θ deletion ameliorates HIR, and heme treatment further improves HIR, which is related to regulation of PKC θ deletion on Nrf 2/HO-1 and TLR4/NF-κB/IKB α pathway.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathophysiology, Wannan Medical College, Wuhu, China
| | - Meng-Yuan Shen
- School of Medical Imaging, Wannan Medical College, Wuhu, China
| | - Ruo-Bing Liu
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jun-Yang Zhang
- School of Medical Imaging, Wannan Medical College, Wuhu, China
| | - Rong-Yu Li
- Department of Immunology, Wannan Medical College, Wuhu, China
| | - Guo-Guang Wang
- Department of Pathophysiology, Wannan Medical College, Wuhu, China
| |
Collapse
|
12
|
Silnitsky S, Rubin SJS, Zerihun M, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases. Int J Mol Sci 2023; 24:17600. [PMID: 38139428 PMCID: PMC10743896 DOI: 10.3390/ijms242417600] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor-kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II).
Collapse
Affiliation(s)
- Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| |
Collapse
|
13
|
Zheng N, Wei J, Wu D, Xu Y, Guo J. Master kinase PDK1 in tumorigenesis. Biochim Biophys Acta Rev Cancer 2023; 1878:188971. [PMID: 37640147 DOI: 10.1016/j.bbcan.2023.188971] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/13/2023] [Accepted: 08/05/2023] [Indexed: 08/31/2023]
Abstract
3-phosphoinositide-dependent protein kinase 1 (PDK1) is considered as master kinase regulating AGC kinase family members such as AKT, SGK, PLK, S6K and RSK. Although autophosphorylation regulates PDK1 activity, accumulating evidence suggests that PDK1 is manipulated by many other mechanisms, including S6K-mediated phosphorylation, and the E3 ligase SPOP-mediated ubiquitination and degradation. Dysregulation of these upstream regulators or downstream signals involves in cancer development, as PDK1 regulating cell growth, metastasis, invasion, apoptosis and survival time. Meanwhile, overexpression of PDK1 is also exposed in a plethora of cancers, whereas inhibition of PDK1 reduces cell size and inhibits tumor growth and progression. More importantly, PDK1 also modulates the tumor microenvironments and markedly influences tumor immunotherapies. In summary, we comprehensively summarize the downstream signals, upstream regulators, mouse models, inhibitors, tumor microenvironment and clinical treatments for PDK1, and highlight PDK1 as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Nana Zheng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Jiaqi Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
14
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
15
|
Enzyme Inhibitors from Gorgonians and Soft Corals. Mar Drugs 2023; 21:md21020104. [PMID: 36827145 PMCID: PMC9963996 DOI: 10.3390/md21020104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
For decades, gorgonians and soft corals have been considered promising sources of bioactive compounds, attracting the interest of scientists from different fields. As the most abundant bioactive compounds within these organisms, terpenoids, steroids, and alkaloids have received the highest coverage in the scientific literature. However, enzyme inhibitors, a functional class of bioactive compounds with high potential for industry and biomedicine, have received much less notoriety. Thus, we revised scientific literature (1974-2022) on the field of marine natural products searching for enzyme inhibitors isolated from these taxonomic groups. In this review, we present representative enzyme inhibitors from an enzymological perspective, highlighting, when available, data on specific targets, structures, potencies, mechanisms of inhibition, and physiological roles for these molecules. As most of the characterization studies for the new inhibitors remain incomplete, we also included a methodological section presenting a general strategy to face this goal by accomplishing STRENDA (Standards for Reporting Enzymology Data) project guidelines.
Collapse
|
16
|
Bengel P, Elkenani M, Beuthner BE, Pietzner M, Mohamed BA, Pollok-Kopp B, Krätzner R, Toischer K, Puls M, Fischer A, Binder L, Hasenfuß G, Schnelle M. Metabolomic Profiling in Patients with Different Hemodynamic Subtypes of Severe Aortic Valve Stenosis. Biomolecules 2023; 13:95. [PMID: 36671480 PMCID: PMC9855798 DOI: 10.3390/biom13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Abstract
Severe aortic stenosis (AS) is a common pathological condition in an ageing population imposing significant morbidity and mortality. Based on distinct hemodynamic features, i.e., ejection fraction (EF), transvalvular gradient and stroke volume, four different AS subtypes can be distinguished: (i) normal EF and high gradient, (ii) reduced EF and high gradient, (iii) reduced EF and low gradient, and (iv) normal EF and low gradient. These subtypes differ with respect to pathophysiological mechanisms, cardiac remodeling, and prognosis. However, little is known about metabolic changes in these different hemodynamic conditions of AS. Thus, we carried out metabolomic analyses in serum samples of 40 AS patients (n = 10 per subtype) and 10 healthy blood donors (controls) using ultrahigh-performance liquid chromatography-tandem mass spectroscopy. A total of 1293 biochemicals could be identified. Principal component analysis revealed different metabolic profiles in all of the subgroups of AS (All-AS) vs. controls. Out of the determined biochemicals, 48% (n = 620) were altered in All-AS vs. controls (p < 0.05). In this regard, levels of various acylcarnitines (e.g., myristoylcarnitine, fold-change 1.85, p < 0.05), ketone bodies (e.g., 3-hydroxybutyrate, fold-change 11.14, p < 0.05) as well as sugar metabolites (e.g., glucose, fold-change 1.22, p < 0.05) were predominantly increased, whereas amino acids (e.g., leucine, fold-change 0.8, p < 0.05) were mainly reduced in All-AS. Interestingly, these changes appeared to be consistent amongst all AS subtypes. Distinct differences between AS subtypes were found for metabolites belonging to hemoglobin metabolism, diacylglycerols, and dihydrosphingomyelins. These findings indicate that relevant changes in substrate utilization appear to be consistent for different hemodynamic subtypes of AS and may therefore reflect common mechanisms during AS-induced heart failure. Additionally, distinct metabolites could be identified to significantly differ between certain AS subtypes. Future studies need to define their pathophysiological implications.
Collapse
Affiliation(s)
- Philipp Bengel
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Manar Elkenani
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Bo E. Beuthner
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Computational Medicine, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Belal A. Mohamed
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Beatrix Pollok-Kopp
- Department of Transfusion Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ralph Krätzner
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Karl Toischer
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Miriam Puls
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Andreas Fischer
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
- Division Vascular Signaling and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Lutz Binder
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Gerd Hasenfuß
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
17
|
Woodland M, Al-Horani RA. New Era: Mavacamten for Obstructive Hypertrophic Cardiomyopathy. Cardiovasc Hematol Agents Med Chem 2023; 21:78-83. [PMID: 36278454 PMCID: PMC10249146 DOI: 10.2174/1871525721666221019095218] [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: 05/26/2022] [Revised: 08/23/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022]
Abstract
Obstructive hypertrophic cardiomyopathy results from asymmetric septal hypertrophy, which eventually obstructs the outflow of the left ventricle. Obstructive hypertrophic cardiomyopathy is linked to mutations in genes that encode for sarcomere proteins, including actin, β-myosin heavy chain, titin, and troponin. The mutations lead to structural abnormalities in myocytes and myofibrils, causing conduction irregularities and abnormal force generation. Obstructive hypertrophic cardiomyopathy is a chronic disease that worsens over time, and patients become at higher risk of developing atrial fibrillation, heart failure, and stroke. Up until recently, there were no disease- specific medications for obstructive hypertrophic cardiomyopathy. Nevertheless, the US Food and Drug Administration approved mavacamten on April 28, 2022, for the treatment of symptomatic obstructive hypertrophic cardiomyopathy (New York Heart Association class II to III) in adults to improve functional capacity and symptoms. Its approval was based on data from EXPLORER- HCM and EXPLORER-LTE (NCT03723655). Mavacamten is a novel, first-in-class, orally active, allosteric inhibitor of cardiac myosin ATPase, which decreases the formation of actin- myosin cross-bridges, and thus, it reduces myocardial contractility, and it improves myocardial energetics. It represents a paradigm-shifting pharmacological treatment of obstructive hypertrophic cardiomyopathy. In this review, we describe its chemical and mechanistic aspects as well as its pharmacokinetics, adverse effects and warnings, potential drug-drug interactions, and contraindications.
Collapse
Affiliation(s)
- Ma’Lik Woodland
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans LA 70125, USA
| | - Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans LA 70125, USA
| |
Collapse
|
18
|
A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology. Biochem Pharmacol 2022; 206:115321. [DOI: 10.1016/j.bcp.2022.115321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/24/2022]
|
19
|
Xu M, Bermea KC, Ayati M, Kim HB, Yang X, Medina A, Fu Z, Heravi A, Zhang X, Na CH, Everett AD, Gabrielson K, Foster DB, Paolocci N, Murphy AM, Ramirez-Correa GA. Alteration in tyrosine phosphorylation of cardiac proteome and EGFR pathway contribute to hypertrophic cardiomyopathy. Commun Biol 2022; 5:1251. [PMID: 36380187 PMCID: PMC9666710 DOI: 10.1038/s42003-022-04021-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Alterations of serine/threonine phosphorylation of the cardiac proteome are a hallmark of heart failure. However, the contribution of tyrosine phosphorylation (pTyr) to the pathogenesis of cardiac hypertrophy remains unclear. We use global mapping to discover and quantify site-specific pTyr in two cardiac hypertrophic mouse models, i.e., cardiac overexpression of ErbB2 (TgErbB2) and α myosin heavy chain R403Q (R403Q-αMyHC Tg), compared to control hearts. From this, there are significant phosphoproteomic alterations in TgErbB2 mice in right ventricular cardiomyopathy, hypertrophic cardiomyopathy (HCM), and dilated cardiomyopathy (DCM) pathways. On the other hand, R403Q-αMyHC Tg mice indicated that the EGFR1 pathway is central for cardiac hypertrophy, along with angiopoietin, ErbB, growth hormone, and chemokine signaling pathways activation. Surprisingly, most myofilament proteins have downregulation of pTyr rather than upregulation. Kinase-substrate enrichment analysis (KSEA) shows a marked downregulation of MAPK pathway activity downstream of k-Ras in TgErbB2 mice and activation of EGFR, focal adhesion, PDGFR, and actin cytoskeleton pathways. In vivo ErbB2 inhibition by AG-825 decreases cardiomyocyte disarray. Serine/threonine and tyrosine phosphoproteome confirm the above-described pathways and the effectiveness of AG-825 Treatment. Thus, altered pTyr may play a regulatory role in cardiac hypertrophic models.
Collapse
Affiliation(s)
- Mingguo Xu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Pediatrics, The Third People’s Hospital of Longgang District, Shenzhen, 518115 China
| | - Kevin C. Bermea
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Marzieh Ayati
- grid.449717.80000 0004 5374 269XDeparment of Computer Science/College of Engineering and Computer Science, University of Texas Rio Grande Valley School of Medicine, Edinburgh, Texas USA
| | - Han Byeol Kim
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xiaomei Yang
- grid.27255.370000 0004 1761 1174Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Andres Medina
- Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| | - Zongming Fu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Amir Heravi
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xinyu Zhang
- grid.27255.370000 0004 1761 1174Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Chan Hyun Na
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Biological Chemistry/McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Allen D. Everett
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Kathleen Gabrielson
- grid.21107.350000 0001 2171 9311Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - D. Brian Foster
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Nazareno Paolocci
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anne M. Murphy
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Genaro A. Ramirez-Correa
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| |
Collapse
|
20
|
Bednarski TK, Duda MK, Dobrzyn P. Alterations of Lipid Metabolism in the Heart in Spontaneously Hypertensive Rats Precedes Left Ventricular Hypertrophy and Cardiac Dysfunction. Cells 2022; 11:cells11193032. [PMID: 36230994 PMCID: PMC9563594 DOI: 10.3390/cells11193032] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
Disturbances in cardiac lipid metabolism are associated with the development of cardiac hypertrophy and heart failure. Spontaneously hypertensive rats (SHRs), a genetic model of primary hypertension and pathological left ventricular (LV) hypertrophy, have high levels of diacylglycerols in cardiomyocytes early in development. However, the exact effect of lipids and pathways that are involved in their metabolism on the development of cardiac dysfunction in SHRs is unknown. Therefore, we used SHRs and Wistar Kyoto (WKY) rats at 6 and 18 weeks of age to analyze the impact of perturbations of processes that are involved in lipid synthesis and degradation in the development of LV hypertrophy in SHRs with age. Triglyceride levels were higher, whereas free fatty acid (FA) content was lower in the LV in SHRs compared with WKY rats. The expression of de novo FA synthesis proteins was lower in cardiomyocytes in SHRs compared with corresponding WKY controls. The higher expression of genes that are involved in TG synthesis in 6-week-old SHRs may explain the higher TG content in these rats. Adenosine monophosphate-activated protein kinase phosphorylation and peroxisome proliferator-activated receptor α protein content were lower in cardiomyocytes in 18-week-old SHRs, suggesting a lower rate of β-oxidation. The decreased protein content of α/β-hydrolase domain-containing 5, adipose triglyceride lipase (ATGL) activator, and increased content of G0/G1 switch protein 2, ATGL inhibitor, indicating a lower rate of lipolysis in the heart in SHRs. In conclusion, the present study showed that the development of LV hypertrophy and myocardial dysfunction in SHRs is associated with triglyceride accumulation, attributable to a lower rate of lipolysis and β-oxidation in cardiomyocytes.
Collapse
Affiliation(s)
- Tomasz K. Bednarski
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Monika K. Duda
- Centre of Postgraduate Medical Education, Department of Clinical Physiology, 01-813 Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
- Correspondence:
| |
Collapse
|
21
|
Reversing Cardiac Hypertrophy at the Source Using a Cardiac Targeting Peptide Linked to miRNA106a: Targeting Genes That Cause Cardiac Hypertrophy. Pharmaceuticals (Basel) 2022; 15:ph15070871. [PMID: 35890169 PMCID: PMC9317130 DOI: 10.3390/ph15070871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/04/2023] Open
Abstract
Causes and treatments for heart failure (HF) have been investigated for over a century culminating in data that have led to numerous pharmacological and surgical therapies. Unfortunately, to date, even with the most current treatments, HF remains a progressive disease with no therapies targeting the cardiomyocytes directly. Technological advances within the past two to three years have brought about new paradigms for treating many diseases that previously had been extremely difficult to resolve. One of these new paradigms has been a shift from pharmacological agents to antisense technology (e.g., microRNAs) to target the molecular underpinnings of pathological processes leading to disease onset. Although this paradigm shift may have been postulated over a decade ago, only within the past few years has it become feasible. Here, we show that miRNA106a targets genes that, when misregulated, have been shown to cause hypertrophy and eventual HF. The addition of miRNA106a suppresses misexpressed HF genes and reverses hypertrophy. Most importantly, using a cardiac targeting peptide reversibly linked to miRNA106a, we show delivery is specific to cardiomyocytes.
Collapse
|
22
|
Wang Y, Morishima M, Ono K. Protein Kinase C Regulates Expression and Function of the Cav3.2 T-Type Ca2+ Channel during Maturation of Neonatal Rat Cardiomyocyte. MEMBRANES 2022; 12:membranes12070686. [PMID: 35877889 PMCID: PMC9321535 DOI: 10.3390/membranes12070686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022]
Abstract
Two distinct isoforms of the T-type Ca2+ channel, Cav3.1 and Cav3.2, play a pivotal role in the generation of pacemaker potentials in nodal cells in the heart, although the isoform switches from Cav3.2 to Cav3.1 during the early neonatal period with an unknown mechanism. The present study was designed to investigate the molecular system of the parts that are responsible for the changes of T-type Ca2+ channel isoforms in neonatal cardiomyocytes using the whole-cell patch-clamp technique and mRNA quantification. The present study demonstrates that PKC activation accelerates the Ni2+-sensitive beating rate and upregulates the Ni2+-sensitive T-type Ca2+ channel current in neonatal cardiomyocytes as a long-term effect, whereas PKC inhibition delays the Ni2+-sensitive beating rate and downregulates the Ni2+-sensitive T-type Ca2+ channel current. Because the Ni2+-sensitive T-type Ca2+ channel current is largely composed of the Cav3.2-T-type Ca2+ channel, it is accordingly assumed that PKC activity plays a crucial role in the maintenance of the Cav3.2 channel. The expression of Cav3.2 mRNA was highly positively correlated with PKC activity. The expression of a transcription factor Nkx2.5 mRNA, possibly corresponding to the Cav3.2 channel gene, was decreased by an inhibition of PKCβII. These results suggest that PKC activation, presumably by PKCβII, is responsible for the upregulation of CaV3.2 T-type Ca2+ channel expression that interacts with a cardiac-specific transcription factor, Nkx2.5, in neonatal cardiomyocytes.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathophysiology, Oita University School of Medicine, Oita 879-5593, Japan; (Y.W.); (M.M.)
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Oita 870-1192, Japan
| | - Masaki Morishima
- Department of Pathophysiology, Oita University School of Medicine, Oita 879-5593, Japan; (Y.W.); (M.M.)
- Department of Food Science and Nutrition, Kindai University Faculty of Agriculture, Nara 631-8505, Japan
| | - Katsushige Ono
- Department of Pathophysiology, Oita University School of Medicine, Oita 879-5593, Japan; (Y.W.); (M.M.)
- Correspondence: ; Tel.: +81-97-586-5650
| |
Collapse
|
23
|
Mohammed MR, El-Bahkery AM, Shedid SM. The Influence of Different γ-Irradiation Patterns on Factors that May Affect Cell Cycle Progression in Male Rats. Dose Response 2022; 20:15593258221117898. [PMID: 35982824 PMCID: PMC9379971 DOI: 10.1177/15593258221117898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most studies of the biological effects of ionizing radiation have been done on a
single acute dose, while clinically and environmentally exposures occur under
chronic/repetitive conditions. It is important to study effects of different
patterns of ionizing radiation. In this study, a rat model was used to compare
the effects of repetitive and acute exposure. Groups: (I) control, (II, III)
were exposed to fractionated doses (1.5 GyX4) and (2 GyX4), respectively/24h
interval, and (IV, V) were exposed to 6 Gy and 8 Gy of whole-body gamma
irradiation, respectively. The gene expression of MAPT and tau phosphorylation
increased in all irradiated groups but the gene expression of PKN not affected.
TGFβ% increased at dose of 2 GyX4 only. In addition, the cell cycle was arrested
in S phase. Micronucleus (MN) increased and cell proliferation decreased. In
conclusion, the dose and pattern of ionizing radiation do not affect the MAPT
and PKN gene expression, but TGF-β, p-tau, MN assay and cell proliferation are
significantly affected. The dose of 2 GyX4 showed distinctive effect. Repetitive
exposure may increase TGF-β%, which causes radio-resistance and, G2/M delay.
Thus, the cell cycle could be regulated in a different manner according to the
dose and pattern of irradiation.
Collapse
|
24
|
Prem PN, Sivakumar B, Boovarahan SR, Kurian GA. Recent advances in potential of Fisetin in the management of myocardial ischemia-reperfusion injury-A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154123. [PMID: 35533608 DOI: 10.1016/j.phymed.2022.154123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 03/26/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The primary therapeutic strategy in managing ischemic heart diseases is to restore the perfusion of the myocardial ischemic area by surgical methods that often result in an unavoidable injury called ischemia-reperfusion injury (IR). Fisetin is an effective flavonoid with antioxidant and anti-inflammatory properties, proven to be cardioprotective against IR injury in both in-vitro and invivo models, apart from its promising health benefits against cancer, diabetes, and neurodegenerative ailments. PURPOSE The potential of fisetin in attenuating myocardial IR is inconclusive as the effectiveness of fisetin needs more understanding in terms of its possible target sites and underlying different mechanisms. Considering the surge in recent scientific interests in fisetin as a pharmacological agent, this review not only updates the existing preclinical and clinical studies with fisetin and its underlying mechanisms but also summarizes its possible targets during IR protection. METHODS We performed a literature survey using search engines Pubmed, PMC, Science direct, Google, and research gate published across the years 2006-2021. The relevant studies were extracted from the databases with the combinations of the following keywords and summarized: myocardial ischemia-reperfusion injury, natural products, flavonoid, fisetin, PI3K, JAK-STAT, Nrf2, PKC, JNK, autophagy. RESULTS Fisetin is reported to be effective in attenuating IR injury by delaying the clotting time, preserving the mitochondrial function, reducing oxidative stress, and inhibiting GSK 3β. But it failed to protect diseased cardiomyocytes challenged to IR. As discussed in the current review, fisetin not only acts as a conventional antioxidant and anti-inflammatory agent to exert its biological effect but may also exert modulatory action on the cellular metabolism and adaptation via direct action on various signalling pathways that comprise PI3K, JAK-STAT, Nrf2, PKC, JNK, and autophagy. Moreover, the dosage of fisetin and co-morbidities like diabetes and obesity are found to be detrimental factors for cardioprotection. CONCLUSION For further evaluation and smooth clinical translation of the fisetin molecule in IR treatment, researchers should pay close attention to the potential of fisetin to possibly alter the key cardioprotective pathways and dosage, as the efficacy of fisetin is tissue and cell type-specific and varies with different doses.
Collapse
Affiliation(s)
- Priyanka N Prem
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Bhavana Sivakumar
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Sri Rahavi Boovarahan
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Gino A Kurian
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India; School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
25
|
PKN2 deficiency leads both to prenatal congenital cardiomyopathy and defective angiotensin II stress responses. Biochem J 2022; 479:1467-1486. [PMID: 35730579 PMCID: PMC9342899 DOI: 10.1042/bcj20220281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022]
Abstract
The protein kinase PKN2 is required for embryonic development and PKN2 knockout mice die as a result of failure in the expansion of mesoderm, cardiac development and neural tube closure. In the adult, cardiomyocyte PKN2 and PKN1 (in combination) are required for cardiac adaptation to pressure-overload. The specific role of PKN2 in contractile cardiomyocytes during development and its role in the adult heart remain to be fully established. We used mice with cardiomyocyte-directed knockout of PKN2 or global PKN2 haploinsufficiency to assess cardiac development and function using high resolution episcopic microscopy, MRI, micro-CT and echocardiography. Biochemical and histological changes were also assessed. Cardiomyocyte-directed PKN2 knockout embryos displayed striking abnormalities in the compact myocardium, with frequent myocardial clefts and diverticula, ventricular septal defects and abnormal heart shape. The sub-Mendelian homozygous knockout survivors developed cardiac failure. RNASeq data showed up-regulation of PKN2 in patients with dilated cardiomyopathy, suggesting an involvement in adult heart disease. Given the rarity of homozygous survivors with cardiomyocyte-specific deletion of PKN2, the requirement for PKN2 in adult mice was explored using the constitutive heterozygous PKN2 knockout. Cardiac hypertrophy resulting from hypertension induced by angiotensin II was reduced in these haploinsufficient PKN2 mice relative to wild-type littermates, with suppression of cardiomyocyte hypertrophy and cardiac fibrosis. It is concluded that cardiomyocyte PKN2 is essential for heart development and the formation of compact myocardium and is also required for cardiac hypertrophy in hypertension. Thus, PKN signalling may offer therapeutic options for managing congenital and adult heart diseases.
Collapse
|
26
|
Zhong C, Zhao H, Xie X, Qi Z, Li Y, Jia L, Zhang J, Lu Y. Protein Kinase C-Mediated Hyperphosphorylation and Lateralization of Connexin 43 Are Involved in Autoimmune Myocarditis-Induced Prolongation of QRS Complex. Front Physiol 2022; 13:815301. [PMID: 35418879 PMCID: PMC9000987 DOI: 10.3389/fphys.2022.815301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Myocarditis is a serious and potentially life-threatening disease, which leads to cardiac dysfunction and sudden cardiac death. An increasing number of evidence suggests that myocarditis is also a malignant complication of coronavirus pneumonia, associated with heart failure and sudden cardiac death. Prolonged QRS complexes that are related to malignant arrhythmias caused by myocarditis significantly increase the risk of sudden cardiac death in patients. However, the molecular mechanisms are not fully known at present. In this study, we identify protein kinase C (PKC) as a new regulator of the QRS complex. In isolated hearts of normal rats, the PKC agonist, phorbol-12-myristate-13-acetate (PMA), induced prolongation of the QRS complex. Mechanistically, hyperphosphorylation and lateralization of connexin 43 (Cx43) by PKC induced depolymerization and internalization of Cx43 gap junction channels and prolongation of the QRS duration. Conversely, administration of the PKC inhibitor, Ro-32-0432, in experimental autoimmune myocarditis (EAM) rats after the most severe inflammation period still significantly rescued the stability of the Cx43 gap junction and alleviated prolongation of the QRS complex. Ro-32-0432 reduced phosphorylation and blocked translocation of Cx43 in EAM rat heart but did not regulate the mRNA expression level of ventricular ion channels and the other regulatory proteins, which indicates that the inhibition of PKC might have no protective effect on ion channels that generate ventricular action potential in EAM rats. These results suggest that the pharmacological inhibition of PKC ameliorates the prolongation of the QRS complex via suppression of Cx43 hyperphosphorylation, lateralization, and depolymerization of Cx43 gap junction channels in EAM rats, which provides a potential therapeutic strategy for myocarditis-induced arrhythmias.
Collapse
Affiliation(s)
- Chunlian Zhong
- School of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Fuzhou, China
| | - Huan Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
| | - Xinwen Xie
- Liancheng County General Hospital, Longyan, China
| | - Zhi Qi
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiamen, China
| | - Yumei Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Lee Jia
- School of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Fuzhou, China
| | - Jinwei Zhang
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital Xiamen University, Xiamen, China
- Hatherly Laboratories, Medical School, College of Medicine and Health, Institute of Biomedical and Clinical Sciences, University of Exeter, Exeter, United Kingdom
| | - Yusheng Lu
- School of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Fuzhou, China
| |
Collapse
|
27
|
Miao LN, Pan D, Shi J, Du JP, Chen PF, Gao J, Yu Y, Shi DZ, Guo M. Role and Mechanism of PKC-δ for Cardiovascular Disease: Current Status and Perspective. Front Cardiovasc Med 2022; 9:816369. [PMID: 35242825 PMCID: PMC8885814 DOI: 10.3389/fcvm.2022.816369] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022] Open
Abstract
Protein kinase C (PKC) is a protein kinase with important cellular functions. PKC-δ, a member of the novel PKC subfamily, has been well-documented over the years. Activation of PKC-δ plays an important regulatory role in myocardial ischemia/reperfusion (IRI) injury and myocardial fibrosis, and its activity and expression levels can regulate pathological cardiovascular diseases such as atherosclerosis, hypertension, cardiac hypertrophy, and heart failure. This article aims to review the structure and function of PKC-δ, summarize the current research regarding its activation mechanism and its role in cardiovascular disease, and provide novel insight into further research on the role of PKC-δ in cardiovascular diseases.
Collapse
Affiliation(s)
- Li-na Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Deng Pan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Junhe Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-peng Du
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng-fei Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Da-Zhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Da-Zhuo Shi
| | - Ming Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Ming Guo
| |
Collapse
|
28
|
Pluteanu F, Boknik P, Heinick A, König C, Müller FU, Weidlich A, Kirchhefer U. Activation of PKC results in improved contractile effects and Ca cycling by inhibition of PP2A-B56α. Am J Physiol Heart Circ Physiol 2022; 322:H427-H441. [PMID: 35119335 DOI: 10.1152/ajpheart.00539.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein phosphatase 2A (PP2A) represents a heterotrimer that is responsible for the dephosphorylation of important regulatory myocardial proteins. The present study was aimed to test whether the phosphorylation of PP2A-B56α at Ser41 by PKC is involved in the regulation of myocyte Ca2+ cycling and contraction. For this purpose, heart preparations of wild-type (WT) and transgenic mice overexpressing the non-phosphorylatable S41A mutant form (TG) were stimulated by administration of the direct PKC activator phorbol 12-myristate 13-acetate (PMA), and functional effects were studied. PKC activation was accompanied by the inhibition of PP2A activity in WT cardiomyocytes, whereas this effect was absent in TG. Consistently, the increase in the sarcomere length shortening and the peak amplitude of Ca2+ transients after PMA administration in WT cardiomyocytes was attenuated in TG. However, the co-stimulation with 1 µM isoprenaline was able to offset these functional deficits. Moreover, TG hearts did not show an increase in the phosphorylation of the myosin-binding protein C after administration of PMA but was detected in corresponding WT. PMA modulated voltage-dependent activation of the L-type Ca2+ channel (LTCC) differently in the two genotypes, shifting V1/2a by +1.5 mV in TG and by 2.4 mV in WT. In the presence of PMA, ICaL inactivation remained unchanged in TG, whereas it was slower in corresponding WT. Our data suggest that PKC-activated enhancement of myocyte contraction and intracellular Ca2+ signaling is mediated by phosphorylation of B56α at Ser41, leading to a decrease in PP2A activity.
Collapse
Affiliation(s)
- Florentina Pluteanu
- Department of Anatomy, Animal Physiology and Biophysics, University of Bucharest, Bucharest, Romania
| | - Peter Boknik
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Alexander Heinick
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Christiane König
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Frank U Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Adam Weidlich
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| |
Collapse
|
29
|
Llorach-Pares L, Nonell-Canals A, Avila C, Sanchez-Martinez M. Computer-Aided Drug Design (CADD) to De-Orphanize Marine Molecules: Finding Potential Therapeutic Agents for Neurodegenerative and Cardiovascular Diseases. Mar Drugs 2022; 20:53. [PMID: 35049908 PMCID: PMC8781171 DOI: 10.3390/md20010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022] Open
Abstract
Computer-aided drug design (CADD) techniques allow the identification of compounds capable of modulating protein functions in pathogenesis-related pathways, which is a promising line on drug discovery. Marine natural products (MNPs) are considered a rich source of bioactive compounds, as the oceans are home to much of the planet's biodiversity. Biodiversity is directly related to chemodiversity, which can inspire new drug discoveries. Therefore, natural products (NPs) in general, and MNPs in particular, have been used for decades as a source of inspiration for the design of new drugs. However, NPs present both opportunities and challenges. These difficulties can be technical, such as the need to dive or trawl to collect the organisms possessing the compounds, or biological, due to their particular marine habitats and the fact that they can be uncultivable in the laboratory. For all these difficulties, the contributions of CADD can play a very relevant role in simplifying their study, since, for example, no biological sample is needed to carry out an in-silico analysis. Therefore, the amount of natural product that needs to be used in the entire preclinical and clinical study is significantly reduced. Here, we exemplify how this combination between CADD and MNPs can help unlock their therapeutic potential. In this study, using a set of marine invertebrate molecules, we elucidate their possible molecular targets and associated therapeutic potential, establishing a pipeline that can be replicated in future studies.
Collapse
Affiliation(s)
- Laura Llorach-Pares
- Mind the Byte S.L., 08028 Barcelona, Catalonia, Spain; (L.L.-P.); (A.N.-C.)
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology and Biodiversity Research Institute (IRBio), University of Barcelona, 08028 Barcelona, Catalonia, Spain;
| | | | - Conxita Avila
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology and Biodiversity Research Institute (IRBio), University of Barcelona, 08028 Barcelona, Catalonia, Spain;
| | | |
Collapse
|
30
|
Sergienko NM, Donner DG, Delbridge LMD, McMullen JR, Weeks KL. Protein phosphatase 2A in the healthy and failing heart: New insights and therapeutic opportunities. Cell Signal 2021; 91:110213. [PMID: 34902541 DOI: 10.1016/j.cellsig.2021.110213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
Protein phosphatases have emerged as critical regulators of phosphoprotein homeostasis in settings of health and disease. Protein phosphatase 2A (PP2A) encompasses a large subfamily of enzymes that remove phosphate groups from serine/threonine residues within phosphoproteins. The heterogeneity in PP2A structure, which arises from the grouping of different catalytic, scaffolding and regulatory subunit isoforms, creates distinct populations of catalytically active enzymes (i.e. holoenzymes) that localise to different parts of the cell. This structural complexity, combined with other regulatory mechanisms, such as interaction of PP2A heterotrimers with accessory proteins and post-translational modification of the catalytic and/or regulatory subunits, enables PP2A holoenzymes to target phosphoprotein substrates in a highly specific manner. In this review, we summarise the roles of PP2A in cardiac physiology and disease. PP2A modulates numerous processes that are vital for heart function including calcium handling, contractility, β-adrenergic signalling, metabolism and transcription. Dysregulation of PP2A has been observed in human cardiac disease settings, including heart failure and atrial fibrillation. Efforts are underway, particularly in the cancer field, to develop therapeutics targeting PP2A activity. The development of small molecule activators of PP2A (SMAPs) and other compounds that selectively target specific PP2A holoenzymes (e.g. PP2A/B56α and PP2A/B56ε) will improve understanding of the function of different PP2A species in the heart, and may lead to the development of therapeutics for normalising aberrant protein phosphorylation in settings of cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Nicola M Sergienko
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Central Clinical School, Monash University, Clayton VIC 3800, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton VIC 3800, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| |
Collapse
|
31
|
Fulton DJ, Stepp DW. Protein kinase N2 connects blood flow with NO production in a double AKT. J Clin Invest 2021; 131:e154256. [PMID: 34720094 PMCID: PMC8553563 DOI: 10.1172/jci154256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Shear stress is an important regulator of blood flow, and luminal endothelial cells (ECs) sense increases in frictional forces and respond with an appropriate release of vasoactive mediators. In this issue of the JCI, Jin et al. identified a mechanism by which ECs respond to shear stress with endothelial NOS (eNOS) activation and NO release. The authors showed that PKN2 was activated by fluid shear stress and contributed to eNOS activation via a double play - indirect phosphorylation at serine 1177 (S1177) via AKT and direct phosphorylation of the S1179 site. Phosphorylation of both sites individually increased eNOS activity, but together they had an additive effect. In sum, these findings reveal exciting details about how shear stress regulates eNOS and have important implications for blood flow and blood pressure.
Collapse
|
32
|
Jin YJ, Chennupati R, Li R, Liang G, Wang S, Iring A, Graumann J, Wettschureck N, Offermanns S. Protein kinase N2 mediates flow-induced endothelial NOS activation and vascular tone regulation. J Clin Invest 2021; 131:e145734. [PMID: 34499618 DOI: 10.1172/jci145734] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 09/01/2021] [Indexed: 01/31/2023] Open
Abstract
Formation of NO by endothelial NOS (eNOS) is a central process in the homeostatic regulation of vascular functions including blood pressure regulation, and fluid shear stress exerted by the flowing blood is a main stimulus of eNOS activity. Previous work has identified several mechanosensing and -transducing processes in endothelial cells, which mediate this process and induce the stimulation of eNOS activity through phosphorylation of the enzyme via various kinases including AKT. How the initial mechanosensing and signaling processes are linked to eNOS phosphorylation is unclear. In human endothelial cells, we demonstrated that protein kinase N2 (PKN2), which is activated by flow through the mechanosensitive cation channel Piezo1 and Gq/G11-mediated signaling, as well as by Ca2+ and phosphoinositide-dependent protein kinase 1 (PDK1), plays a pivotal role in this process. Active PKN2 promoted the phosphorylation of human eNOS at serine 1177 and at a newly identified site, serine 1179. These phosphorylation events additively led to increased eNOS activity. PKN2-mediated eNOS phosphorylation at serine 1177 involved the phosphorylation of AKT synergistically with mTORC2-mediated AKT phosphorylation, whereas active PKN2 directly phosphorylated human eNOS at serine 1179. Mice with induced endothelium-specific deficiency of PKN2 showed strongly reduced flow-induced vasodilation and developed arterial hypertension accompanied by reduced eNOS activation. These results uncover a central mechanism that couples upstream mechanosignaling processes in endothelial cells to the regulation of eNOS-mediated NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ramesh Chennupati
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - András Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Laboratory of Molecular Medicine, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| |
Collapse
|
33
|
Muzorewa TT, Buerk DG, Jaron D, Barbee KA. Coordinated regulation of endothelial calcium signaling and shear stress-induced nitric oxide production by PKCβ and PKCη. Cell Signal 2021; 87:110125. [PMID: 34474112 DOI: 10.1016/j.cellsig.2021.110125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Protein Kinase C (PKC) is a promiscuous serine/threonine kinase regulating vasodilatory responses in vascular endothelial cells. Calcium-dependent PKCbeta (PKCβ) and calcium-independent PKCeta (PKCη) have both been implicated in the regulation and dysfunction of endothelial responses to shear stress and agonists. OBJECTIVE We hypothesized that PKCβ and PKCη differentially modulate shear stress-induced nitric oxide (NO) production by regulating the transduced calcium signals and the resultant eNOS activation. As such, this study sought to characterize the contribution of PKCη and PKCβ in regulating calcium signaling and endothelial nitric oxide synthase (eNOS) activation after exposure of endothelial cells to ATP or shear stress. METHODS Bovine aortic endothelial cells were stimulated in vitro under pharmacological inhibition of PKCβ with LY333531 or PKCη targeting with a pseudosubstrate inhibitor. The participation of PKC isozymes in calcium flux, eNOS phosphorylation and NO production was assessed following stimulation with ATP or shear stress. RESULTS PKCη proved to be a robust regulator of agonist- and shear stress-induced eNOS activation, modulating calcium fluxes and tuning eNOS activity by multi-site phosphorylation. PKCβ showed modest influence in this pathway, promoting eNOS activation basally and in response to shear stress. Both PKC isozymes contributed to the constitutive and induced phosphorylation of eNOS. The observed PKC signaling architecture is intricate, recruiting Src to mediate a portion of PKCη's control on calcium entry and eNOS phosphorylation. Elucidation of the importance of PKCη in this pathway was tempered by evidence of a single stimulus producing concurrent phosphorylation at ser1179 and thr497 which are antagonistic to eNOS activity. CONCLUSIONS We have, for the first time, shown in a single species in vitro that shear stress- and ATP-stimulated NO production are differentially regulated by classical and novel PKCs. This study furthers our understanding of the PKC isozyme interplay that optimizes NO production. These considerations will inform the ongoing design of drugs for the treatment of PKC-sensitive cardiovascular pathologies.
Collapse
Affiliation(s)
- Tenderano T Muzorewa
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
Activators and Inhibitors of Protein Kinase C (PKC): Their Applications in Clinical Trials. Pharmaceutics 2021; 13:pharmaceutics13111748. [PMID: 34834162 PMCID: PMC8621927 DOI: 10.3390/pharmaceutics13111748] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Protein kinase C (PKC), a family of phospholipid-dependent serine/threonine kinase, is classed into three subfamilies based on their structural and activation characteristics: conventional or classic PKC isozymes (cPKCs; α, βI, βII, and γ), novel or non-classic PKC isozymes (nPKCs; δ, ε, η, and θ), and atypical PKC isozymes (aPKCs; ζ, ι, and λ). PKC inhibitors and activators are used to understand PKC-mediated intracellular signaling pathways and for the diagnosis and treatment of various PKC-associated diseases, such as cancers, neurological diseases, cardiovascular diseases, and infections. Many clinical trials of PKC inhibitors in cancers showed no significant clinical benefits, meaning that there is a limitation to design a cancer therapeutic strategy targeting PKC alone. This review will focus on the activators and inhibitors of PKC and their applications in clinical trials.
Collapse
|
35
|
Lemoine KA, Fassas JM, Ohannesian SH, Purcell NH. On the PHLPPside: Emerging roles of PHLPP phosphatases in the heart. Cell Signal 2021; 86:110097. [PMID: 34320369 PMCID: PMC8403656 DOI: 10.1016/j.cellsig.2021.110097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
PH domain leucine-rich repeat protein phosphatase (PHLPP) is a family of enzymes made up of two isoforms (PHLPP1 and PHLPP2), whose actions modulate intracellular activity via the dephosphorylation of specific serine/threonine (Ser/Thr) residues on proteins such as Akt. Recent data generated in our lab, supported by findings from others, implicates the divergent roles of PHLPP1 and PHLPP2 in maintaining cellular homeostasis since dysregulation of these enzymes has been linked to various pathological states including cardiovascular disease, diabetes, ischemia/reperfusion injury, musculoskeletal disease, and cancer. Therefore, development of therapies to modulate specific isoforms of PHLPP could prove to be therapeutically beneficial in several diseases especially those targeting the cardiovascular system. This review is intended to provide a comprehensive summary of current literature detailing the role of the PHLPP isoforms in the development and progression of heart disease.
Collapse
Affiliation(s)
- Kellie A Lemoine
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Julianna M Fassas
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Shirag H Ohannesian
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA; Cardiovascular Molecular Signaling, Huntington Medical Research Institutes, Pasadena, CA 91105, USA.
| |
Collapse
|
36
|
Vahabi N, McDonough CW, Desai AA, Cavallari LH, Duarte JD, Michailidis G. Cox-sMBPLS: An Algorithm for Disease Survival Prediction and Multi-Omics Module Discovery Incorporating Cis-Regulatory Quantitative Effects. Front Genet 2021; 12:701405. [PMID: 34408773 PMCID: PMC8366414 DOI: 10.3389/fgene.2021.701405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/07/2021] [Indexed: 12/03/2022] Open
Abstract
Background The development of high-throughput techniques has enabled profiling a large number of biomolecules across a number of molecular compartments. The challenge then becomes to integrate such multimodal Omics data to gain insights into biological processes and disease onset and progression mechanisms. Further, given the high dimensionality of such data, incorporating prior biological information on interactions between molecular compartments when developing statistical models for data integration is beneficial, especially in settings involving a small number of samples. Results We develop a supervised model for time to event data (e.g., death, biochemical recurrence) that simultaneously accounts for redundant information within Omics profiles and leverages prior biological associations between them through a multi-block PLS framework. The interactions between data from different molecular compartments (e.g., epigenome, transcriptome, methylome, etc.) were captured by using cis-regulatory quantitative effects in the proposed model. The model, coined Cox-sMBPLS, exhibits superior prediction performance and improved feature selection based on both simulation studies and analysis of data from heart failure patients. Conclusion The proposed supervised Cox-sMBPLS model can effectively incorporate prior biological information in the survival prediction system, leading to improved prediction performance and feature selection. It also enables the identification of multi-Omics modules of biomolecules that impact the patients’ survival probability and also provides insights into potential relevant risk factors that merit further investigation.
Collapse
Affiliation(s)
- Nasim Vahabi
- Informatics Institute, University of Florida, Gainesville, FL, United States
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Larisa H Cavallari
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Julio D Duarte
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - George Michailidis
- Informatics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
37
|
Singh RK, Kumar S, Tomar MS, Verma PK, Kumar A, Kumar S, Kumar N, Singh JP, Acharya A. Putative role of natural products as Protein Kinase C modulator in different disease conditions. ACTA ACUST UNITED AC 2021; 29:397-414. [PMID: 34216003 DOI: 10.1007/s40199-021-00401-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Protein kinase C (PKC) is a promising drug target for various therapeutic areas. Natural products derived from plants, animals, microorganisms, and marine organisms have been used by humans as medicine from prehistoric times. Recently, several compounds derived from plants have been found to modulate PKC activities through competitive binding with ATP binding site, and other allosteric regions of PKC. As a result fresh race has been started in academia and pharmaceutical companies to develop an effective naturally derived small-molecule inhibitor to target PKC activities. Herein, in this review, we have discussed several natural products and their derivatives, which are reported to have an impact on PKC signaling cascade. METHODS All information presented in this review article regarding the regulation of PKC by natural products has been acquired by a systematic search of various electronic databases, including ScienceDirect, Scopus, Google Scholar, Web of science, ResearchGate, and PubMed. The keywords PKC, natural products, curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, protocatechuic acid, tannic acid, PKC modulators from marine organism, bryostatin, staurosporine, midostaurin, sangivamycin, and other relevant key words were explored. RESULTS The natural products and their derivatives including curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, bryostatin, staurosporine, and midostaurin play a major role in the management of PKC activity during various disease progression. CONCLUSION Based on the comprehensive literature survey, it could be concluded that various natural products can regulate PKC activity during disease progression. However, extensive research is needed to circumvent the challenge of isoform specific regulation of PKC by natural products.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | | | - Munendra Singh Tomar
- Department of Pharmaceutical Science, School of Pharmacy, University of Colorado, Denver, USA
| | | | - Amit Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Sandeep Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Naveen Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Jai Prakash Singh
- Department of Panchkarma, Institute of Medical Science, BHU, Varanasi, India, 221005
| | - Arbind Acharya
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India.
| |
Collapse
|
38
|
Yang CM, Yang CC, Hsiao LD, Yu CY, Tseng HC, Hsu CK, Situmorang JH. Upregulation of COX-2 and PGE 2 Induced by TNF-α Mediated Through TNFR1/MitoROS/PKCα/P38 MAPK, JNK1/2/FoxO1 Cascade in Human Cardiac Fibroblasts. J Inflamm Res 2021; 14:2807-2824. [PMID: 34234507 PMCID: PMC8254141 DOI: 10.2147/jir.s313665] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Tumor necrosis factor-α (TNF-α) has been shown to exert as a pathogenic factor in cardiac fibrosis and heart failure which were associated with the up-regulation of cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) axis. However, whether TNF-α-induced COX-2/PGE2 upregulation mediated through ROS-dependent cascade remains elusive in human cardiac fibroblasts (HCFs). This study aims to address the underlying mechanisms of TNF-α-induced COX-2/PGE2 expression. METHODS Here, we used TNF receptor neutralizing antibody (TNFR nAb), pharmacologic inhibitors, and siRNAs to dissect the involvement of signaling components examined by Western blot and ELISA in TNF-α-mediated responses in HCFs. MitoSOX Red was used to measure mitoROS generation. Isolation of subcellular fractions was performed to determine membrane translocation of PKCα. Promoter luciferase assay and chromatin immunoprecipitation (ChIP) assay were used to determine the role of transcription factor. RESULTS We found that TNF-α time- and concentration-dependently upregulated COX-2 protein and mRNA expression as well as PGE2 synthesis which was attenuated by TNFR1 nAb, the inhibitor of mitochondrial ROS scavenger (MitoTEMPO), protein kinase C [(PKC)α, Gö6976], p38 MAPK [p38 inhibitor VIII, (p38i VIII)], JNK1/2 (SP600125), or forkhead box protein O1 [(FoxO1), AS1842856], and transfection with their respective siRNAs in HCFs. TNF-α-stimulated PKCα phosphorylation was inhibited by TNFR1 nAb, MitoTEMPO, or Gö6976. TNF-α stimulated phosphorylation of p38 MAPK and JNK1/2 was attenuated by TNFR1 nAb, MitoTEMPO, Gö6976, and their inhibitors p38i VIII and SP600125. Moreover, TNF-α-triggered FoxO1 phosphorylation was abolished by AS1842856, TNFR1 nAb, and its upstream inhibitors MitoTEMPO, Gö6976, p38i VIII, and SP600125. Phosphorylation of FoxO1 could enhance its interaction with the COX-2 promoter element revealed by ChIP assay, which was attenuated by AS1842856. CONCLUSION Our results suggested that TNF-α-induced COX-2/PGE2 upregulation is mediated through TNFR1-dependent MitoROS/PKCα/p38 MAPK and JNK1/2 cascade to activate FoxO1 binding with the COX-2 promoter in HCFs.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, 40402, Taiwan
- Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung, 41354, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan, 33302, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Ying Yu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Hui-Ching Tseng
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chih-Kai Hsu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Jiro Hasegawa Situmorang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| |
Collapse
|
39
|
Chen L, Shi D, Guo M. The roles of PKC-δ and PKC-ε in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105716. [PMID: 34102229 DOI: 10.1016/j.phrs.2021.105716] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/01/2021] [Accepted: 06/03/2021] [Indexed: 01/14/2023]
Abstract
Ischemia and reperfusion (I/R) cause a reduction in arterial blood supply to tissues, followed by the restoration of perfusion and consequent reoxygenation. The reestablishment of blood flow triggers further damage to ischemic tissue through reactive oxygen species (ROS) accumulation, interference with cellular ion homeostasis, opening of mitochondrial permeability transition pores (mPTPs) and promotion of cell death (apoptosis or necrosis). PKC-δ and PKC-ε, belonging to a family of serine/threonine kinases, have been demonstrated to play important roles during I/R injury in cardiovascular diseases. However, the cardioprotective mechanisms of PKC-δ and PKC-ε in I/R injury have not been elaborated until now. This article discusses the roles of PKC-δ and PKC-ε during myocardial I/R in redox regulation (redox signaling and oxidative stress), cell death (apoptosis and necrosis), Ca2+ overload, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Li Chen
- Peking University Traditional Chinese Medicine Clinical Medical School (Xi yuan), Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ming Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
40
|
Protein Kinase C as a Therapeutic Target in Non-Small Cell Lung Cancer. Int J Mol Sci 2021; 22:ijms22115527. [PMID: 34073823 PMCID: PMC8197251 DOI: 10.3390/ijms22115527] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
Driver-directed therapeutics have revolutionized cancer treatment, presenting similar or better efficacy compared to traditional chemotherapy and substantially improving quality of life. Despite significant advances, targeted therapy is greatly limited by resistance acquisition, which emerges in nearly all patients receiving treatment. As a result, identifying the molecular modulators of resistance is of great interest. Recent work has implicated protein kinase C (PKC) isozymes as mediators of drug resistance in non-small cell lung cancer (NSCLC). Importantly, previous findings on PKC have implicated this family of enzymes in both tumor-promotive and tumor-suppressive biology in various tissues. Here, we review the biological role of PKC isozymes in NSCLC through extensive analysis of cell-line-based studies to better understand the rationale for PKC inhibition. PKC isoforms α, ε, η, ι, ζ upregulation has been reported in lung cancer, and overexpression correlates with worse prognosis in NSCLC patients. Most importantly, PKC isozymes have been established as mediators of resistance to tyrosine kinase inhibitors in NSCLC. Unfortunately, however, PKC-directed therapeutics have yielded unsatisfactory results, likely due to a lack of specific evaluation for PKC. To achieve satisfactory results in clinical trials, predictive biomarkers of PKC activity must be established and screened for prior to patient enrollment. Furthermore, tandem inhibition of PKC and molecular drivers may be a potential therapeutic strategy to prevent the emergence of resistance in NSCLC.
Collapse
|
41
|
Protein kinase C-mediated calcium signaling as the basis for cardiomyocyte plasticity. Arch Biochem Biophys 2021; 701:108817. [PMID: 33626379 DOI: 10.1016/j.abb.2021.108817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 01/08/2023]
Abstract
Protein kinase C is the superfamily of intracellular effector molecules which control crucial cellular functions. Here, we for the first time did the percentage estimation of all known PKC and PKC-related isozymes at the individual cadiomyocyte level. Broad spectrum of PKC transcripts is expressed in the left ventricular myocytes. In addition to the well-known 'heart-specific' PKCα, cardiomyocytes have the high expression levels of PKCN1, PKCδ, PKCD2, PKCε. In general, we detected all PKC isoforms excluding PKCη. In cardiomyocytes PKC activity tonically regulates voltage-gated Ca2+-currents, intracellular Ca2+ level and nitric oxide (NO) production. Imidazoline receptor of the first type (I1R)-mediated induction of the PKC activity positively modulates Ca2+ release through ryanodine receptor (RyR), increasing the Ca2+ leakage in the cytosol. In cardiomyocytes with the Ca2+-overloaded regions of > 9-10 μm size, the local PKC-induced Ca2+ signaling is transformed to global accompanied by spontaneous Ca2+ waves propagation across the entire cell perimeter. Such switching of Ca2+ signaling in cardiac cells can be important for the development of several cardiovascular pathologies and/or myocardial plasticity at the cardiomyocyte level.
Collapse
|
42
|
Kolczynska K, Loza-Valdes A, Hawro I, Sumara G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: a review. Lipids Health Dis 2020; 19:113. [PMID: 32466765 PMCID: PMC7257441 DOI: 10.1186/s12944-020-01286-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Protein kinase C (PKC) and Protein kinase D (PKD) isoforms can sense diacylglycerol (DAG) generated in the different cellular compartments in various physiological processes. DAG accumulates in multiple organs of the obese subjects, which leads to the disruption of metabolic homeostasis and the development of diabetes as well as associated diseases. Multiple studies proved that aberrant activation of PKCs and PKDs contributes to the development of metabolic diseases. DAG-sensing PKC and PKD isoforms play a crucial role in the regulation of metabolic homeostasis and therefore might serve as targets for the treatment of metabolic disorders such as obesity and diabetes.
Collapse
Affiliation(s)
- Katarzyna Kolczynska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Angel Loza-Valdes
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Izabela Hawro
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland.
| |
Collapse
|
43
|
Ai L, Perez E, Asimes A, Kampaengsri T, Heroux M, Zlobin A, Hiske MA, Chung CS, Pak TR, Kirk JA. Binge Alcohol Exposure in Adolescence Impairs Normal Heart Growth. J Am Heart Assoc 2020; 9:e015611. [PMID: 32319345 PMCID: PMC7428579 DOI: 10.1161/jaha.119.015611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Approximately 1 in 6 adolescents report regular binge alcohol consumption, and we hypothesize it affects heart growth during this period. Methods and Results Adolescent, genetically diverse, male Wistar rats were gavaged with water or ethanol once per day for 6 days. In vivo structure and function were assessed before and after exposure. Binge alcohol exposure in adolescence significantly impaired normal cardiac growth but did not affect whole‐body growth during adolescence, therefore this pathology was specific to the heart. Binge rats also exhibited signs of accelerated pathological growth (concentric cellular hypertrophy and thickening of the myocardial wall), suggesting a global reorientation from physiologic to pathologic growth. Binge rats compensated for their smaller filling volumes by increasing systolic function and sympathetic stimulation. Consequently, binge alcohol exposure increased PKA (protein kinase A) phosphorylation of troponin I, inducing myofilament calcium desensitization. Binge alcohol also impaired in vivo relaxation and increased titin‐based cellular stiffness due to titin phosphorylation by PKCα (protein kinase C α). Mechanistically, alcohol inhibited extracellular signal‐related kinase activity, a nodal signaling kinase activating physiology hypertrophy. Thus, binge alcohol exposure depressed genes involved in growth. These cardiac structural alterations from binge alcohol exposure persisted through adolescence even after cessation of ethanol exposure. Conclusions Alcohol negatively impacts function in the adult heart, but the adolescent heart is substantially more sensitive to its effects. This difference is likely because adolescent binge alcohol impedes the normal rapid physiological growth and reorients it towards pathological hypertrophy. Many adolescents regularly binge alcohol, and here we report a novel pathological consequence as well as mechanisms involved.
Collapse
Affiliation(s)
- Lizhuo Ai
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Edith Perez
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - AnnaDorothea Asimes
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Theerachat Kampaengsri
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Maxime Heroux
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Andrei Zlobin
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Mark A Hiske
- Department of Physiology Wayne State University Detroit MI
| | | | - Toni R Pak
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology Loyola University Chicago Stritch School of Medicine Maywood IL
| |
Collapse
|
44
|
Zhang Y, Zhai H. Bilobalide assuages morphine-induced addiction in hippocampal neuron cells through upregulation of microRNA-101. J Biochem Mol Toxicol 2020; 34:e22493. [PMID: 32319158 DOI: 10.1002/jbt.22493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/24/2019] [Accepted: 03/03/2020] [Indexed: 11/09/2022]
Abstract
Bilobalide exhibits many biological activities, but its effects on morphine stimulation have not been elucidated. The research aims to explore the function and underlying mechanisms of bilobalide in morphine-led hippocampal neuron cells. Cells were treated with or without morphine or oxaliplatin (OXA), bilobalide, or SCH772984 dilutions. miR-101 inhibitor and negative control were transfected into cells. Western blot and quantitative reverse transcription-polymerase chain reaction were, respectively, conducted to measure the relative expression of proteins or RNAs. Morphine improved the expression levels of orexin1 receptor (OX1R) and c-FOS, the p/t-ERK/PKC as well. The c-FOS protein level and p/t-ERK/PKC were significantly elevated by morphine + OXA. Bilobalide had no effect on OX1R and p/t-PKC but evidently decreased the c-FOS and p/t-ERK. The p-ERK and the c-FOS accumulation levels were remarkably reduced by SCH772984. The production of miR-101 was promoted by bilobalide but inhibited by the miR-101 inhibitor. miR-101 inhibitor abolished bilobalide's inhibitory effects on p/t-ERK. Bilobalide exhibited morphine-induced effects on hippocampal neuron cells by upregulating miR-101.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Hongyin Zhai
- Department of Children Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
45
|
Advanced glycation end products facilitate the proliferation and reduce early apoptosis of cardiac microvascular endothelial cells via PKCβ signaling pathway: Insight from diabetic cardiomyopathy. Anatol J Cardiol 2020; 23:141-150. [PMID: 32120359 PMCID: PMC7222633 DOI: 10.14744/anatoljcardiol.2019.21504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective: To investigate the effects of advanced glycation end products (AGEs) on the proliferation and apoptosis of cardiac microvascular endothelial cells (CMECs) in rats and their underlying signaling pathway. Methods: CMECs were isolated from Sprague–Dawley rats. We first examined the effects of AGEs on the proliferation and apoptosis of CMECs and then tested whether protein kinase C (PKC) β blockers could counteract the effects of AGEs. The PKC agonists phorbol 12-myristate 13-acetate (PMA) and PKCβ blockers were also used to verify whether PKC could act independently on CMECs. The receptor for AGEs (RAGE)–small interfering RNA (siRNA) transfection was used to verify the effect of AGEs on PKC. Following the above steps, we explained whether AGEs regulated the CMEC proliferation and early apoptosis through the PKCβ signaling pathway. Proliferation of CMECs was detected using the Cell Counting Kit-8 (CCK-8) assay, and early apoptosis was determined using the Annexin V- Fluorescein Isothiocyanate (FITC)/propidium iodide (PI) double staining. Expression of proliferation and apoptosis-related proteins and PKC phosphorylation were determined by western blotting analysis. Cell cycle distributions were assayed using a BD FACSCalibur cell-sorting system. Results: AGEs facilitated the proliferation of CMECs, upregulated phosphorylated extracellular signal regulated kinase (p-ERK), and accelerated the entry of cells from G1 phase to the S+G2/M phase, which was consistent with the upregulated cyclin D1 by AGEs. AGEs inhibited early apoptosis of CMECs by increasing the expression of survivin and decreasing the expression of cleaved-caspase3. All these effects can be reversed by PKCβ1/2inhibitors. In addition, AGE upregulated the RAGE expression and phosphorylation of PKCβ1/2 in CMECs, while the inhibition of RAGE reversed the phosphorylation, as well as the effects of AGEs on proliferation and apoptosis in CMECs. Conclusion: The study indicated that AGEs facilitated the proliferation and reduced early apoptosis of CMECs via the PKCβ signaling pathway.
Collapse
|
46
|
Gao J, Shao K, Chen X, Li Z, Liu Z, Yu Z, Aung LHH, Wang Y, Li P. The involvement of post-translational modifications in cardiovascular pathologies: Focus on SUMOylation, neddylation, succinylation, and prenylation. J Mol Cell Cardiol 2020; 138:49-58. [DOI: 10.1016/j.yjmcc.2019.11.146] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
|
47
|
Sakaguchi T, Takefuji M, Wettschureck N, Hamaguchi T, Amano M, Kato K, Tsuda T, Eguchi S, Ishihama S, Mori Y, Yura Y, Yoshida T, Unno K, Okumura T, Ishii H, Shimizu Y, Bando YK, Ohashi K, Ouchi N, Enomoto A, Offermanns S, Kaibuchi K, Murohara T. Protein Kinase N Promotes Stress-Induced Cardiac Dysfunction Through Phosphorylation of Myocardin-Related Transcription Factor A and Disruption of Its Interaction With Actin. Circulation 2019; 140:1737-1752. [PMID: 31564129 DOI: 10.1161/circulationaha.119.041019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart failure is a complex syndrome that results from structural or functional impairment of ventricular filling or blood ejection. Protein phosphorylation is a major and essential intracellular mechanism that mediates various cellular processes in cardiomyocytes in response to extracellular and intracellular signals. The RHOA-associated protein kinase (ROCK/Rho-kinase), an effector regulated by the small GTPase RHOA, causes pathological phosphorylation of proteins, resulting in cardiovascular diseases. RHOA also activates protein kinase N (PKN); however, the role of PKN in cardiovascular diseases remains unclear. METHODS To explore the role of PKNs in heart failure, we generated tamoxifen-inducible, cardiomyocyte-specific PKN1- and PKN2-knockout mice by intercrossing the αMHC-CreERT2 line with Pkn1flox/flox and Pkn2flox/flox mice and applied a mouse model of transverse aortic constriction- and angiotensin II-induced heart failure. To identify a novel substrate of PKNs, we incubated GST-tagged myocardin-related transcription factor A (MRTFA) with recombinant GST-PKN-catalytic domain or GST-ROCK-catalytic domain in the presence of radiolabeled ATP and detected radioactive GST-MRTFA as phosphorylated MRTFA. RESULTS We demonstrated that RHOA activates 2 members of the PKN family of proteins, PKN1 and PKN2, in cardiomyocytes of mice with cardiac dysfunction. Cardiomyocyte-specific deletion of the genes encoding Pkn1 and Pkn2 (cmc-PKN1/2 DKO) did not affect basal heart function but protected mice from pressure overload- and angiotensin II-induced cardiac dysfunction. Furthermore, we identified MRTFA as a novel substrate of PKN1 and PKN2 and found that MRTFA phosphorylation by PKN was considerably more effective than that by ROCK in vitro. We confirmed that endogenous MRTFA phosphorylation in the heart was induced by pressure overload- and angiotensin II-induced cardiac dysfunction in wild-type mice, whereas cmc-PKN1/2 DKO mice suppressed transverse aortic constriction- and angiotensin II-induced phosphorylation of MRTFA. Although RHOA-mediated actin polymerization accelerated MRTFA-induced gene transcription, PKN1 and PKN2 inhibited the interaction of MRTFA with globular actin by phosphorylating MRTFA, causing increased serum response factor-mediated expression of cardiac hypertrophy- and fibrosis-associated genes. CONCLUSIONS Our results indicate that PKN1 and PKN2 activation causes cardiac dysfunction and is involved in the transition to heart failure, thus providing unique targets for therapeutic intervention for heart failure.
Collapse
Affiliation(s)
- Teruhiro Sakaguchi
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Mikito Takefuji
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (N.W., S.O.)
| | - Tomonari Hamaguchi
- Cell Pharmacology (T.H., M.A., Y.Y., K. Kaibuchi), Nagoya University School of Medicine, Japan
| | - Mutsuki Amano
- Cell Pharmacology (T.H., M.A., Y.Y., K. Kaibuchi), Nagoya University School of Medicine, Japan
| | - Katsuhiro Kato
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Takuma Tsuda
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Shunsuke Eguchi
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Sohta Ishihama
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Yu Mori
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Yoshimitsu Yura
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan.,Cell Pharmacology (T.H., M.A., Y.Y., K. Kaibuchi), Nagoya University School of Medicine, Japan
| | - Tatsuya Yoshida
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Kazumasa Unno
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Takahiro Okumura
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Hideki Ishii
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Yuuki Shimizu
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Yasuko K Bando
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| | - Koji Ohashi
- Molecular Medicine and Cardiology (K.O., N.O.), Nagoya University School of Medicine, Japan
| | - Noriyuki Ouchi
- Molecular Medicine and Cardiology (K.O., N.O.), Nagoya University School of Medicine, Japan
| | - Atsushi Enomoto
- Pathology (A.E.), Nagoya University School of Medicine, Japan
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (N.W., S.O.)
| | - Kozo Kaibuchi
- Cell Pharmacology (T.H., M.A., Y.Y., K. Kaibuchi), Nagoya University School of Medicine, Japan
| | - Toyoaki Murohara
- Departments of Cardiology (T.S., M.T., K. Kato, T.T., S.E., S.I., Y.M., Y.Y., T.Y., K.U., T.O, H.I., Y.S., Y.K.B., T.M.), Nagoya University School of Medicine, Japan
| |
Collapse
|
48
|
Current State of Hypertrophic Cardiomyopathy Clinical Trials. Glob Heart 2019; 14:317-325. [DOI: 10.1016/j.gheart.2019.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
|
49
|
Lin JLJ. Characterization of the novel cardiolipin binding regions identified on the protease and lipid activated PKC-related kinase 1. Protein Sci 2019; 28:1473-1486. [PMID: 31125460 DOI: 10.1002/pro.3663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/22/2019] [Indexed: 11/09/2022]
Abstract
Protein kinase C-related kinase 1 (PRK1) or PKN is a protease and lipid activated protein kinase that acted downstream of the RhoA or Rac1 pathway. PRK1 comprises a unique regulatory domain and a PKC homologous kinase domain. The regulatory domain of PRK1 consists of homologous region -1 (HR1) and -2 (HR2). PRK1-(HR1) features a pseudosubstrate motif that overlapped with the putative cardiolipin and known RhoA binding sites. In fact, cardiolipin is the most potent lipid activator for PRK1 in respect of its either auto- or substrate phosphorylation activity. This study was thus aimed to characterize the binding region(s) of cardiolipin that was previously suggested for the regulatory domain of PRK1. The principal findings of this work established (i) PRK1-(HR1) folded into an active conformation where high affinity binding sites (mainly located in HR1a subdomain) were accessible for cardiolipin binding to protect against limited Lys-C digestion, (ii) the binding nature between acidic phospholipids and PRK1 (HR1) involved both polar and nonpolar components consistent with the amphipathic nature of the known cardiolipin-binding motifs, (iii) identification of the molecule masses of the Lys-C fragments of PRK1-(HR1) complexed with cardiolipin molecule, and (iv) appreciable reductions in the secondary structural contents at 222 nm measured by circular dichroism analyses demonstrated the binding of cardiolipin elicited the disruptive effect that was most evident among all phospholipids tested, suggestive of a functional correlation between the extents of helical disruption and PRK1 activation.
Collapse
Affiliation(s)
- Jason L J Lin
- Department of Biochemistry and Molecular Biology, University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
50
|
Goswami S. G protein-coupled receptor signaling in cardiovascular system: Specificity versus diversity. JOURNAL OF THE PRACTICE OF CARDIOVASCULAR SCIENCES 2019. [DOI: 10.4103/jpcs.jpcs_37_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|