1
|
Jiang Y, Tong W, Li Y, Ma Q, Chen Y. Melatonin inhibits the formation of intraplaque neovessels in ApoE-/- mice via PPARγ- RhoA-ROCK pathway. Biochem Biophys Res Commun 2024; 696:149391. [PMID: 38184922 DOI: 10.1016/j.bbrc.2023.149391] [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: 09/28/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND According to former research, the atherosclerotic plaque is thought to be aggravated by intraplaque neovessels (IPN) and intraplaque hemorrhage (IPH). Intriguingly, a lower incidence of IPH was found in plaque treated with melatonin. In this study, we attempted to investigate the impact and underlying mechanism regarding the influences of melatonin upon IPN. METHODS A mouse model was established by subjecting the high fat diet (HFD)-fed ApoE-/- mice to tandem stenosis (TS) surgery with melatonin and GW9662, a PPARγ antagonist, being given by gavage. In vitro experiment was conducted with HUVECs exposing to according treatments of VEGF, melatonin, GW9662, or Y27632. RESULTS Plaque and IPN were attenuated by treatment with melatonin, which was then reversed by blocking PPARγ. Western blotting results showed that melatonin increased PPARγ and decreased RhoA/ROCK signaling in carotid artery. Elevated RhoA/ROCK signaling was observed in melatonin-treated mice when PPARγ was blocked. In accordance with it, experiments using protein and mRNA from HUVECs revealed that melatonin inhibited the RhoA/ROCK signaling by enhancing PPARγ. According to in vitro study, melatonin was able to inhibit cell migration and angiogenesis, which was aborted by GW9662. Blockage of ROCK using Y27632 was able to cease the effect of GW9662 and restored the suppression on cell migration and angiogenesis by melatonin. CONCLUSIONS Our study demonstrates that melatonin is able to curb development of plaque and IPN formation by inhibiting the migration of endothelial cells via PPARγ- RhoA-ROCK pathway. That provides a therapeutic potential for both melatonin and PPARγ agonist targeting IPN, IPH, and atherosclerotic plaque.
Collapse
Affiliation(s)
- YuFan Jiang
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Tong
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yueyang Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiang Ma
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - YunDai Chen
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
2
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [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] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
3
|
Procopio A, Cesarelli G, Donisi L, Merola A, Amato F, Cosentino C. Combined mechanistic modeling and machine-learning approaches in systems biology - A systematic literature review. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 240:107681. [PMID: 37385142 DOI: 10.1016/j.cmpb.2023.107681] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND AND OBJECTIVE Mechanistic-based Model simulations (MM) are an effective approach commonly employed, for research and learning purposes, to better investigate and understand the inherent behavior of biological systems. Recent advancements in modern technologies and the large availability of omics data allowed the application of Machine Learning (ML) techniques to different research fields, including systems biology. However, the availability of information regarding the analyzed biological context, sufficient experimental data, as well as the degree of computational complexity, represent some of the issues that both MMs and ML techniques could present individually. For this reason, recently, several studies suggest overcoming or significantly reducing these drawbacks by combining the above-mentioned two methods. In the wake of the growing interest in this hybrid analysis approach, with the present review, we want to systematically investigate the studies available in the scientific literature in which both MMs and ML have been combined to explain biological processes at genomics, proteomics, and metabolomics levels, or the behavior of entire cellular populations. METHODS Elsevier Scopus®, Clarivate Web of Science™ and National Library of Medicine PubMed® databases were enquired using the queries reported in Table 1, resulting in 350 scientific articles. RESULTS Only 14 of the 350 documents returned by the comprehensive search conducted on the three major online databases met our search criteria, i.e. present a hybrid approach consisting of the synergistic combination of MMs and ML to treat a particular aspect of systems biology. CONCLUSIONS Despite the recent interest in this methodology, from a careful analysis of the selected papers, it emerged how examples of integration between MMs and ML are already present in systems biology, highlighting the great potential of this hybrid approach to both at micro and macro biological scales.
Collapse
Affiliation(s)
- Anna Procopio
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italia
| | - Giuseppe Cesarelli
- Department of Electrical Engineering and Information Technology, Università degli Studi di Napoli Federico II, Napoli, 80125, Italy
| | - Leandro Donisi
- Department of Advanced Medical and Surgical Sciences, Università della Campania Luigi Vanvitelli, Napoli, 80138, Italy
| | - Alessio Merola
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italia
| | - Francesco Amato
- Department of Electrical Engineering and Information Technology, Università degli Studi di Napoli Federico II, Napoli, 80125, Italy.
| | - Carlo Cosentino
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italia.
| |
Collapse
|
4
|
Ponsiglione AM, Montefusco F, Donisi L, Tedesco A, Cosentino C, Merola A, Romano M, Amato F. A General Approach for the Modelling of Negative Feedback Physiological Control Systems. Bioengineering (Basel) 2023; 10:835. [PMID: 37508862 PMCID: PMC10376068 DOI: 10.3390/bioengineering10070835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Mathematical models can improve the understanding of physiological systems behaviour, which is a fundamental topic in the bioengineering field. Having a reliable model enables researchers to carry out in silico experiments, which require less time and resources compared to their in vivo and in vitro counterparts. This work's objective is to capture the characteristics that a nonlinear dynamical mathematical model should exhibit, in order to describe physiological control systems at different scales. The similarities among various negative feedback physiological systems have been investigated and a unique general framework to describe them has been proposed. Within such a framework, both the existence and stability of equilibrium points are investigated. The model here introduced is based on a closed-loop topology, on which the homeostatic process is based. Finally, to validate the model, three paradigmatic examples of physiological control systems are illustrated and discussed: the ultrasensitivity mechanism for achieving homeostasis in biomolecular circuits, the blood glucose regulation, and the neuromuscular reflex arc (also referred to as muscle stretch reflex). The results show that, by a suitable choice of the modelling functions, the dynamic evolution of the systems under study can be described through the proposed general nonlinear model. Furthermore, the analysis of the equilibrium points and dynamics of the above-mentioned systems are consistent with the literature.
Collapse
Affiliation(s)
- Alfonso Maria Ponsiglione
- Dipartimento di Ingegneria Elettrica e delle Tecnologie dell'Informazione, Università degli Studi di Napoli Federico II, Via Claudio 21, 80125 Napoli, Italy
| | - Francesco Montefusco
- Dipartimento di Scienze Economiche, Giuridiche, Informatiche e Motorie, Università degli Studi di Napoli Parthenope, 80035 Nola, Italy
| | - Leandro Donisi
- Dipartimento di Scienze Mediche e Chirurgiche Avanzate, Università degli studi della Campania "Luigi Vanvitelli", P.zza L. Miraglia 2, 80138 Napoli, Italy
| | - Annarita Tedesco
- Dipartimento di Ingegneria per l'Innovazione, Universitá del Salento, 73100 Lecce, Italy
| | - Carlo Cosentino
- School of Computer and Biomedical Engineering, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi Magna Græcia di Catanzaro, Campus di Germaneto "Salvatore Venuta", 88100 Catanzaro, Italy
| | - Alessio Merola
- School of Computer and Biomedical Engineering, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi Magna Græcia di Catanzaro, Campus di Germaneto "Salvatore Venuta", 88100 Catanzaro, Italy
| | - Maria Romano
- Dipartimento di Ingegneria Elettrica e delle Tecnologie dell'Informazione, Università degli Studi di Napoli Federico II, Via Claudio 21, 80125 Napoli, Italy
| | - Francesco Amato
- Dipartimento di Ingegneria Elettrica e delle Tecnologie dell'Informazione, Università degli Studi di Napoli Federico II, Via Claudio 21, 80125 Napoli, Italy
| |
Collapse
|
5
|
Reisqs JB, Moreau A, Sleiman Y, Boutjdir M, Richard S, Chevalier P. Arrhythmogenic cardiomyopathy as a myogenic disease: highlights from cardiomyocytes derived from human induced pluripotent stem cells. Front Physiol 2023; 14:1191965. [PMID: 37250123 PMCID: PMC10210147 DOI: 10.3389/fphys.2023.1191965] [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: 03/22/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized by the replacement of myocardium by fibro-fatty infiltration and cardiomyocyte loss. ACM predisposes to a high risk for ventricular arrhythmias. ACM has initially been defined as a desmosomal disease because most of the known variants causing the disease concern genes encoding desmosomal proteins. Studying this pathology is complex, in particular because human samples are rare and, when available, reflect the most advanced stages of the disease. Usual cellular and animal models cannot reproduce all the hallmarks of human pathology. In the last decade, human-induced pluripotent stem cells (hiPSC) have been proposed as an innovative human cellular model. The differentiation of hiPSCs into cardiomyocytes (hiPSC-CM) is now well-controlled and widely used in many laboratories. This hiPSC-CM model recapitulates critical features of the pathology and enables a cardiomyocyte-centered comprehensive approach to the disease and the screening of anti-arrhythmic drugs (AAD) prescribed sometimes empirically to the patient. In this regard, this model provides unique opportunities to explore and develop new therapeutic approaches. The use of hiPSC-CMs will undoubtedly help the development of precision medicine to better cure patients suffering from ACM. This review aims to summarize the recent advances allowing the use of hiPSCs in the ACM context.
Collapse
Affiliation(s)
- J. B. Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - A. Moreau
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - Y. Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - M. Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, NY, United States
- Department of Medicine, New York University School of Medicine, NY, United States
| | - S. Richard
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - P. Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
6
|
Shoykhet M, Dervishi O, Menauer P, Hiermaier M, Moztarzadeh S, Osterloh C, Ludwig RJ, Williams T, Gerull B, Kääb S, Clauss S, Schüttler D, Waschke J, Yeruva S. EGFR inhibition leads to enhanced desmosome assembly and cardiomyocyte cohesion via ROCK activation. JCI Insight 2023; 8:163763. [PMID: 36795511 PMCID: PMC10070108 DOI: 10.1172/jci.insight.163763] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/15/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart disease partly caused by impaired desmosome turnover. Thus, stabilization of desmosome integrity may provide new treatment options. Desmosomes, apart from cellular cohesion, provide the structural framework of a signaling hub. Here, we investigated the role of the epidermal growth factor receptor (EGFR) in cardiomyocyte cohesion. We inhibited EGFR under physiological and pathophysiological conditions using the murine plakoglobin-KO AC model, in which EGFR was upregulated. EGFR inhibition enhanced cardiomyocyte cohesion. Immunoprecipitation showed an interaction of EGFR and desmoglein 2 (DSG2). Immunostaining and atomic force microscopy (AFM) revealed enhanced DSG2 localization and binding at cell borders upon EGFR inhibition. Enhanced area composita length and desmosome assembly were observed upon EGFR inhibition, confirmed by enhanced DSG2 and desmoplakin (DP) recruitment to cell borders. PamGene Kinase assay performed in HL-1 cardiomyocytes treated with erlotinib, an EGFR inhibitor, revealed upregulation of Rho-associated protein kinase (ROCK). Erlotinib-mediated desmosome assembly and cardiomyocyte cohesion were abolished upon ROCK inhibition. Thus, inhibiting EGFR and, thereby, stabilizing desmosome integrity via ROCK might provide treatment options for AC.
Collapse
Affiliation(s)
- Maria Shoykhet
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Orsela Dervishi
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Philipp Menauer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
7
|
Akoumianakis I, Polkinghorne M, Antoniades C. Non-canonical WNT signalling in cardiovascular disease: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:783-797. [PMID: 35697779 PMCID: PMC9191761 DOI: 10.1038/s41569-022-00718-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
WNT signalling comprises a diverse spectrum of receptor-mediated pathways activated by a large family of WNT ligands and influencing fundamental biological processes. WNT signalling includes the β-catenin canonical pathway and the non-canonical pathways, namely the planar cell polarity and the calcium-dependent pathways. Advances over the past decade have linked non-canonical WNT signalling with key mechanisms of atherosclerosis, including oxidative stress, endothelial dysfunction, macrophage activation and vascular smooth muscle cell phenotype regulation. In addition, non-canonical WNT signalling is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and oxidative stress. Importantly, non-canonical WNT signalling activation has complex effects in adipose tissue in the context of obesity, thereby potentially linking metabolic and vascular diseases. Tissue-specific targeting of non-canonical WNT signalling might be associated with substantial risks of off-target tumorigenesis, challenging its therapeutic potential. However, novel technologies, such as monoclonal antibodies, recombinant decoy receptors, tissue-specific gene silencing with small interfering RNAs and gene editing with CRISPR-Cas9, might enable more efficient therapeutic targeting of WNT signalling in the cardiovascular system. In this Review, we summarize the components of non-canonical WNT signalling, their links with the main mechanisms of atherosclerosis, heart failure and arrhythmias, and the rationale for targeting individual components of non-canonical WNT signalling for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Murray Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Abstract
Purpose of Review The advent of induced pluripotent stem cells (iPSC) has paved the way for new in vitro models of human cardiomyopathy. Herein, we will review existing models of disease as well as strengths and limitations of the system. Recent Findings Preclinical studies have now demonstrated that iPSCs generated from patients with both acquired or heritable genetic diseases retain properties of the disease in vitro and can be used as a model to study novel therapeutics. iPSCs can be differentiated in vitro into the cardiomyocyte lineage into cells resembling adult ventricular myocytes that retain properties of cardiovascular disease from their respective donor. iPSC pluripotency allows for them to be frozen, stored, and continually used to generate iPSC-derived myocytes for future experiments without need for invasive procedures or repeat myocyte isolations to obtain animal or human cardiac tissues. Summary While not without their limitations, iPSC models offer new ways for studying patient-specific cardiomyopathies. iPSCs offer a high-throughput avenue for drug development, modeling of disease pathophysiology in vitro, and enabling experimental repair strategies without need for invasive procedures to obtain cardiac tissues.
Collapse
|
9
|
Procopio A, Lagreca E, Jamaledin R, La Manna S, Corrado B, Di Natale C, Onesto V. Recent Fabrication Methods to Produce Polymer-Based Drug Delivery Matrices (Experimental and In Silico Approaches). Pharmaceutics 2022; 14:872. [PMID: 35456704 PMCID: PMC9027538 DOI: 10.3390/pharmaceutics14040872] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023] Open
Abstract
The study of novel drug delivery systems represents one of the frontiers of the biomedical research area. Multi-disciplinary scientific approaches combining traditional or engineered technologies are used to provide major advances in improving drug bioavailability, rate of release, cell/tissue specificity and therapeutic index. Biodegradable and bio-absorbable polymers are usually the building blocks of these systems, and their copolymers are employed to create delivery components. For example, poly (lactic acid) or poly (glycolic acid) are often used as bricks for the production drug-based delivery systems as polymeric microparticles (MPs) or micron-scale needles. To avoid time-consuming empirical approaches for the optimization of these formulations, in silico-supported models have been developed. These methods can predict and tune the release of different drugs starting from designed combinations. Starting from these considerations, this review has the aim of investigating recent approaches to the production of polymeric carriers and the combination of in silico and experimental methods as promising platforms in the biomedical field.
Collapse
Affiliation(s)
- Anna Procopio
- Biomechatronics Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Elena Lagreca
- Department of Chemical, Materials & Industrial Production Engineering, University of Naples Federico II, 80131 Naples, Italy; (E.L.); (R.J.)
- Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Rezvan Jamaledin
- Department of Chemical, Materials & Industrial Production Engineering, University of Naples Federico II, 80131 Naples, Italy; (E.L.); (R.J.)
| | - Sara La Manna
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
| | - Brunella Corrado
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80131 Naples, Italy;
| | - Concetta Di Natale
- Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80131 Naples, Italy;
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-Nanotec), Campus Ecotekne, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
10
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
11
|
Scaramuzzino L, Lucchino V, Scalise S, Lo Conte M, Zannino C, Sacco A, Biamonte F, Parrotta EI, Costanzo FS, Cuda G. Uncovering the Metabolic and Stress Responses of Human Embryonic Stem Cells to FTH1 Gene Silencing. Cells 2021; 10:2431. [PMID: 34572080 PMCID: PMC8469604 DOI: 10.3390/cells10092431] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Embryonic stem cells (ESCs) are pluripotent cells with indefinite self-renewal ability and differentiation properties. To function properly and maintain genomic stability, ESCs need to be endowed with an efficient repair system as well as effective redox homeostasis. In this study, we investigated different aspects involved in ESCs' response to iron accumulation following stable knockdown of the ferritin heavy chain (FTH1) gene, which encodes for a major iron storage protein with ferroxidase activity. Experimental findings highlight unexpected and, to a certain extent, paradoxical results. If on one hand FTH1 silencing does not correlate with increased ROS production nor with changes in the redox status, strengthening the concept that hESCs are extremely resistant and, to a certain extent, even refractory to intracellular iron imbalance, on the other, the differentiation potential of hESCs seems to be affected and apoptosis is observed. Interestingly, we found that FTH1 silencing is accompanied by a significant activation of the nuclear factor (erythroid-derived-2)-like 2 (Nrf2) signaling pathway and pentose phosphate pathway (PPP), which crosstalk in driving hESCs antioxidant cascade events. These findings shed new light on how hESCs perform under oxidative stress, dissecting the molecular mechanisms through which Nrf2, in combination with PPP, counteracts oxidative injury triggered by FTH1 knockdown.
Collapse
Affiliation(s)
- Luana Scaramuzzino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Valeria Lucchino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Stefania Scalise
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Michela Lo Conte
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Clara Zannino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Alessandro Sacco
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Flavia Biamonte
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
- Center of Interdepartmental Services (CIS), “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Francesco Saverio Costanzo
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
- Center of Interdepartmental Services (CIS), “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| |
Collapse
|