1
|
Vázquez-Cuevas FG, Reyna-Jeldes M, Velázquez-Miranda E, Coddou C. Transactivation of receptor tyrosine kinases by purinergic P2Y and adenosine receptors. Purinergic Signal 2023; 19:613-621. [PMID: 36529846 PMCID: PMC10754767 DOI: 10.1007/s11302-022-09913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Transactivation of receptor tyrosine kinases (RTK) is a crosstalk mechanism exhibited by G-protein-coupled receptors (GPCR) to activate signaling pathways classically associated with growth factors. The discovery of RTK transactivation was a breakthrough in signal transduction that contributed to developing current concepts in intracellular signaling. RTK transactivation links GPCR signaling to important cellular processes, such as cell proliferation and differentiation, and explains the functional diversity of these receptors. Purinergic (P2Y and adenosine) receptors belong to class A of GPCR; in the present work, we systematically review the experimental evidence showing that purinergic receptors have the ability to transactivate RTK in multiple tissues and physiopathological conditions resulting in the modulation of cellular physiology. Of particular relevance, the crosstalk between purinergic receptors and epidermal growth factor receptor is a redundant pathway that participates in multiple pathophysiological processes. Specific and detailed knowledge of purinergic receptor-regulated pathways advances our understanding of the complexity of GPCR signal transduction and opens the way for pharmacologic intervention in the pathological context.
Collapse
Affiliation(s)
- F G Vázquez-Cuevas
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México.
| | - M Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile
| | - E Velázquez-Miranda
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México
| | - C Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile.
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile.
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile.
| |
Collapse
|
2
|
Pinheiro da Silva F, Gonçalves ANA, Duarte‐Neto AN, Dias TL, Barbeiro HV, Breda CNS, Breda LCD, Câmara NOS, Nakaya HI. Transcriptome analysis of six tissues obtained post-mortem from sepsis patients. J Cell Mol Med 2023; 27:3157-3167. [PMID: 37731199 PMCID: PMC10568675 DOI: 10.1111/jcmm.17938] [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: 03/08/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Septic shock is a life-threatening clinical condition characterized by a robust immune inflammatory response to disseminated infection. Little is known about its impact on the transcriptome of distinct human tissues. To address this, we performed RNA sequencing of samples from the prefrontal cortex, hippocampus, heart, lung, kidney and colon of seven individuals who succumbed to sepsis and seven uninfected controls. We identified that the lungs and colon were the most affected organs. While gene activation dominated, strong inhibitory signals were also detected, particularly in the lungs. We found that septic shock is an extremely heterogeneous disease, not only when different individuals are investigated, but also when comparing different tissues of the same patient. However, several pathways, such as respiratory electron transport and other metabolic functions, revealed distinctive alterations, providing evidence that tissue specificity is a hallmark of sepsis. Strikingly, we found evident signals of accelerated ageing in our sepsis population.
Collapse
Affiliation(s)
| | | | | | | | - Hermes Vieira Barbeiro
- Laboratório de Emergências Clínicas, Faculdade de MedicinaUniversidade de São PauloSão PauloBrazil
| | | | | | | | - Helder I. Nakaya
- Faculdade de Ciências FarmacêuticasUniversidade de São PauloSão PauloBrazil
- Hospital Israelita Albert EinsteinSão PauloBrazil
| |
Collapse
|
3
|
Phillips J, Akemann C, Shields JN, Wu CC, Meyer DN, Baker BB, Pitts DK, Baker TR. Developmental phenotypic and transcriptomic effects of exposure to nanomolar levels of metformin in zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 87:103716. [PMID: 34311114 PMCID: PMC8446320 DOI: 10.1016/j.etap.2021.103716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 05/06/2023]
Abstract
Metformin is found in the majority of lakes and streams in the United States, leading to widespread environmental exposure. Results of the present study indicate that extended duration metformin exposure at critical developmental periods leads to decreased survival rates in zebrafish (danio rerio), an NIH approved human model. Significant abnormalities are seen with extended duration metformin exposure from 4 h post fertilization up to 5 days post fertilization, although short term metformin exposure for 24 h at 4-5 days post fertilization did not lead to any significant abnormalities. Both extended and short term duration did however have an impact on locomotor activity of zebrafish, and several genes involved in neurological and cardiovascular development were differentially expressed after exposure to metformin. The changes seen in behavior, gene expression and morphological abnormalities caused by metformin exposure should be examined further in future studies in order to assess their potential human health implications as metformin prescriptions continue to increase worldwide.
Collapse
Affiliation(s)
- Jessica Phillips
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Camille Akemann
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Jeremiah N Shields
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Chia-Chen Wu
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Danielle N Meyer
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Bridget B Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - David K Pitts
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Tracie R Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA.
| |
Collapse
|
4
|
Jin Y, Ai L, Chai X, Tang P, Zhang W, Yang L, Hu Y, Xu Y, Li S. Maternal Circulating Exosomal miRNAs as Non-invasive Biomarkers for the Prediction of Fetal Ventricular Septal Defect. Front Genet 2021; 12:717208. [PMID: 34567071 PMCID: PMC8458870 DOI: 10.3389/fgene.2021.717208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022] Open
Abstract
Objective: This study aimed to identify maternal circulating exosomal miRNAs as potential non-invasive biomarkers for the early detection of fetal ventricular septal defects (VSDs). Methods: In total, 182 pregnant women, comprising 91 VSD cases and 91 matched controls, were included in this study. Exosomes were isolated; dysregulated exosomal miRNAs were profiled using next-generation sequencing. Differential abundance of miRNAs was verified using quantitative real-time polymerase chain reaction (qRT-PCR). Diagnostic accuracy was evaluated by constructing receiver operating characteristic (ROC) curves. Results: In total, 77 serum exosomal miRNAs were found to be differentially expressed in the VSD group compared to their expression in the control group. Among these, five downregulated exosomal miRNAs were validated using qRT-PCR. hsa-miR-146a-5p was identified to be capable of distinguishing VSD cases from controls (area under the ROC curve [AUC]: 0.997; p < 1.00E-05). Conclusion: Circulating exosomal miRNAs, particularly hsa-miR-146a-5p, may be predictive biomarkers for the non-invasive prenatal diagnosis of fetal VSDs.
Collapse
Affiliation(s)
- Yuxia Jin
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Ling Ai
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Xiaojun Chai
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Ping Tang
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Weihua Zhang
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Li Yang
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Yue Hu
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| | - Ying Xu
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Suping Li
- Department of Prenatal Diagnostic, Jiaxing Maternity and Children Health Care Hospital, Jiaxing University, Jiaxing, China
| |
Collapse
|
5
|
Heliste J, Jokilammi A, Vaparanta K, Paatero I, Elenius K. Combined genetic and chemical screens indicate protective potential for EGFR inhibition to cardiomyocytes under hypoxia. Sci Rep 2021; 11:16661. [PMID: 34404849 PMCID: PMC8371130 DOI: 10.1038/s41598-021-96033-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
The return of blood flow to ischemic heart after myocardial infarction causes ischemia-reperfusion injury. There is a clinical need for novel therapeutic targets to treat myocardial ischemia-reperfusion injury. Here we screened for targets for the treatment of ischemia-reperfusion injury using a combination of shRNA and drug library analyses in HL-1 mouse cardiomyocytes subjected to hypoxia and reoxygenation. The shRNA library included lentiviral constructs targeting 4625 genes and the drug library 689 chemical compounds approved by the Food and Drug Administration (FDA). Data were analyzed using protein-protein interaction and pathway analyses. EGFR inhibition was identified as a cardioprotective mechanism in both approaches. Inhibition of EGFR kinase activity with gefitinib improved cardiomyocyte viability in vitro. In addition, gefitinib preserved cardiac contractility in zebrafish embryos exposed to hypoxia-reoxygenation in vivo. These findings indicate that the EGFR inhibitor gefitinib is a potential candidate for further studies of repurposing the drug for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Juho Heliste
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland.,Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Anne Jokilammi
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland
| | - Katri Vaparanta
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland.,Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland.,MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland.
| | - Klaus Elenius
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland. .,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland. .,MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland. .,Department of Oncology, Turku University Hospital, PO Box 52, 20521, Turku, Finland.
| |
Collapse
|
6
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Wang Z, Chan HW, Gambarotta G, Smith NJ, Purdue BW, Pennisi DJ, Porrello ER, O'Brien SL, Reichelt ME, Thomas WG, Paravicini TM. Stimulation of the four isoforms of receptor tyrosine kinase ErbB4, but not ErbB1, confers cardiomyocyte hypertrophy. J Cell Physiol 2021; 236:8160-8170. [PMID: 34170016 DOI: 10.1002/jcp.30487] [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: 04/01/2020] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 11/11/2022]
Abstract
Epidermal growth factor (EGF) receptors (ErbB1-ErbB4) promote cardiac development and growth, although the specific EGF ligands and receptor isoforms involved in growth/repair versus pathology remain undefined. We challenged ventricular cardiomyocytes with EGF-like ligands and observed that selective activation of ErbB4 (the receptor for neuregulin 1 [NRG1]), but not ErbB1 (the receptor for EGF, EGFR), stimulated hypertrophy. This lack of direct ErbB1-mediated hypertrophy occurred despite robust activation of extracellular-regulated kinase 1/2 (ERK) and protein kinase B. Hypertrophic responses to NRG1 were unaffected by the tyrosine kinase inhibitor (AG1478) at concentrations that are selective for ErbB1 over ErbB4. NRG1-induced cardiomyocyte enlargement was suppressed by small interfering RNA (siRNA) knockdown of ErbB4 and ErbB2, whereas ERK phosphorylation was only suppressed by ErbB4 siRNA. Four ErbB4 isoforms exist (JM-a/JM-b and CYT-1/CYT-2), generated by alternative splicing, and their expression declines postnatally and following cardiac hypertrophy. Silencing of all four isoforms in cardiomyocytes, using an ErbB4 siRNA, abrogated NRG1-induced hypertrophic promoter/reporter activity, which was rescued by coexpression of knockdown-resistant versions of the ErbB4 isoforms. Thus, ErbB4 confers cardiomyocyte hypertrophy to NRG1, and all four ErbB4 isoforms possess the capacity to mediate this effect.
Collapse
Affiliation(s)
- Zhen Wang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Hsiu-Wen Chan
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Nicola J Smith
- School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, New South Wales, Australia
| | - Brooke W Purdue
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - David J Pennisi
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shannon L O'Brien
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tamara M Paravicini
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Guan G, Huo D, Li Y, Zhao X, Li Y, Qin Z, Sun D, Yang G, Yang M, Tan J, Zeng W, Zhu C. Engineering hiPSC-CM and hiPSC-EC laden 3D nanofibrous splenic hydrogel for improving cardiac function through revascularization and remuscularization in infarcted heart. Bioact Mater 2021; 6:4415-4429. [PMID: 33997517 PMCID: PMC8113784 DOI: 10.1016/j.bioactmat.2021.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Cell therapy has been a promising strategy for cardiac repair after myocardial infarction (MI), but a poor ischemic environment and low cell delivery efficiency remain significant challenges. The spleen serves as a hematopoietic stem cell niche and secretes cardioprotective factors after MI, but it is unclear whether it could be used for human pluripotent stem cell (hiPSC) cultivation and provide a proper microenvironment for cell grafts against the ischemic environment. Herein, we developed a splenic extracellular matrix derived thermoresponsive hydrogel (SpGel). Proteomics analysis indicated that SpGel is enriched with proteins known to modulate the Wnt signaling pathway, cell-substrate adhesion, cardiac muscle contraction and oxidation-reduction processes. In vitro studies demonstrated that hiPSCs could be efficiently induced into endothelial cells (iECs) and cardiomyocytes (iCMs) with enhanced function on SpGel. The cytoprotective effect of SpGel on iECs/iCMs against oxidative stress damage was also proven. Furthermore, in vivo studies revealed that iEC/iCM-laden SpGel improved cardiac function and inhibited cardiac fibrosis of infarcted hearts by improving cell survival, revascularization and remuscularization. In conclusion, we successfully established a novel platform for the efficient generation and delivery of autologous cell grafts, which could be a promising clinical therapeutic strategy for cardiac repair and regeneration after MI.
Collapse
Affiliation(s)
- Ge Guan
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Da Huo
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yanzhao Li
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaolin Zhao
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yinghao Li
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zhongliang Qin
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Chongqing Institute of Zhong Zhi Yi Gu, Shapingba District, Chongqing, 400030, China
| | - Dayu Sun
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Guanyuan Yang
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Mingcan Yang
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ju Tan
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Army Medical University, Chongqing, 400038, China
| | - Chuhong Zhu
- Department of Anatomy, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| |
Collapse
|
9
|
Xu M, Yang Y, Deng QW, Shen JT, Liu WF, Yang WJ, Liu KX. Microarray Profiling and Functional Identification of LncRNA in Mice Intestinal Mucosa Following Intestinal Ischemia/Reperfusion. J Surg Res 2021; 258:389-404. [PMID: 33109405 DOI: 10.1016/j.jss.2020.08.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 08/06/2020] [Accepted: 08/25/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) injury is a common clinical event with high mortality, but its mechanism is elusive. Although long noncoding RNAs (lncRNAs) have recently emerged as critical molecules in I/R damage in other organs, the changes in their expression and potential roles in intestinal I/R remain unclear. METHODS The expression profiles of both lncRNAs and mRNAs in mouse intestinal mucosa after intestinal I/R were explored by a microarray approach, and their biological functions were elucidated by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Then, some lncRNAs were further verified by qRT-PCR. Based on the coding-noncoding gene coexpression (CNC) network analyses, the role of lncRNA AK089510 in intestinal I/R-induced intestinal mucosa apoptosis was investigated by knockdown assay in vitro. RESULTS A total of 3602 aberrantly expressed lncRNAs (1503 upregulated and 2099 downregulated) and 3158 mRNAs (1528 upregulated and 1630 downregulated) were identified. The dysregulated transcripts were enriched in the lipid metabolic process, apoptotic process, reactive oxygen species metabolic process, MAPK, TNF, ErbB, mTOR, and FoxO signaling pathways, and so on. The overexpression of lncRNA AK089510 was validated by qRT-PCR, and the CNC analysis revealed its target mRNAs. AK089510-siRNA reduced Casp6 and Casp7 expression and suppressed intestinal epithelial cell apoptosis after oxygen-glucose deprivation treatment. CONCLUSIONS Our study revealed the lncRNA and mRNA expression patterns in mouse intestinal mucosa after intestinal I/R and predicted their potential functions and pathways. We identified AK089510 as a novel lncRNA involved in the apoptosis of intestinal mucosa, advancing our understanding of the molecular mechanisms of intestinal I/R injury.
Collapse
Affiliation(s)
- Miao Xu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Yang
- Department of Neurology, Guangzhou First People'(')s Hospital, Guangzhou, China
| | - Qi-Wen Deng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jian-Tong Shen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei-Feng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Jing Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Wang L, Yuan W, Huang J. Identification of Myocardial Infarction-Associated Genes Using Integrative microRNA-Gene Expression Network Analysis. DNA Cell Biol 2020; 40:348-358. [PMID: 33395357 DOI: 10.1089/dna.2020.6222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It is crucial to identify potential molecular targets and their interaction involved in myocardial infarction (MI). In our study, we obtained microarray data of MI from GEO database and identify differentially expressed mRNAs and microRNAs (miRNAs). Compared with normal tissues, 686 mRNAs and 16 miRNAs were differentially expressed in MI. Subsequently, function enrichment analysis was performed to further investigate their biological functions. Also, gene set enrichment analysis indicated they were enriched into Pathway in cancer. Besides, protein-protein interaction analysis was performed to assess the interactions of the differentially expressed mRNAs. Finally, we constructed an mRNA-miRNA interaction network based on the overlapping genes between the differentially expressed mRNAs and predicted target genes of dysregulated miRNAs. The network demonstrated three MI-associated miRNAs, miR-498, miR-181a, and miR-612, and 45 novel target genes, as well as their interaction involved in MI. What is more, in vitro and in vivo quantitative real-time PCR confirmed our results were consistent. In conclusion, miR-498, miR-181a, and miR-612 may participate in the pathogenesis of MI and may serve as the potential therapeutic targets or biomarkers.
Collapse
Affiliation(s)
- Long Wang
- Department of Cardiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Yuan
- Department of Cardiology, Jiangsu Taizhou People's Hospital, Taizhou, China
| | - Jinyu Huang
- Department of Cardiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Wang H, Yuan R, Cao Q, Wang M, Ren D, Huang X, Wu M, Zhang L, Zhao X, Huo X, Pan Y, Liu Q. Astragaloside III activates TACE/ADAM17‐dependent anti‐inflammatory and growth factor signaling in endothelial cells in a p38‐dependent fashion. Phytother Res 2020; 34:1096-1107. [PMID: 32197276 DOI: 10.1002/ptr.6603] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/31/2019] [Accepted: 11/30/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Haifang Wang
- Shaanxi and Xianyang Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrated MedicineShaanxi University of Chinese Medicine Xianyang China
| | - Ruihua Yuan
- Shaanxi and Xianyang Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrated MedicineShaanxi University of Chinese Medicine Xianyang China
| | - Qingwen Cao
- Division of Medical ManagementShaanxi Provincial Hospital of Traditional Chinese Medicine Xi'an China
| | - Mian Wang
- Shaanxi and Xianyang Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrated MedicineShaanxi University of Chinese Medicine Xianyang China
| | - Dezhi Ren
- Department of CardiologyShaanxi Provincial Hospital of Traditional Chinese Medicine Xi'an China
| | - Xiaoyan Huang
- Laboratory CenterShaanxi Provincial People's Hospital Xi'an China
| | - Min Wu
- Laboratory CenterShaanxi Provincial People's Hospital Xi'an China
| | - Linping Zhang
- Laboratory CenterShaanxi Provincial People's Hospital Xi'an China
| | - Xiangrong Zhao
- Laboratory CenterShaanxi Provincial People's Hospital Xi'an China
| | - Xueping Huo
- Laboratory CenterShaanxi Provincial People's Hospital Xi'an China
| | - Yalei Pan
- Shaanxi Collaborative Innovation Center of Chinese Medicine Resources Industrialization, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research CenterShaanxi University of Chinese Medicine Xianyang China
| | - Qinshe Liu
- Shaanxi and Xianyang Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrated MedicineShaanxi University of Chinese Medicine Xianyang China
| |
Collapse
|
12
|
De Pascale MR, Della Mura N, Vacca M, Napoli C. Useful applications of growth factors for cardiovascular regenerative medicine. Growth Factors 2020; 38:35-63. [PMID: 33028111 DOI: 10.1080/08977194.2020.1825410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel advances for cardiovascular diseases (CVDs) include regenerative approaches for fibrosis, hypertrophy, and neoangiogenesis. Studies indicate that growth factor (GF) signaling could promote heart repair since most of the evidence is derived from preclinical models. Observational studies have evaluated GF serum/plasma levels as feasible biomarkers for risk stratification of CVDs. Noteworthy, two clinical interventional published studies showed that the administration of growth factors (GFs) induced beneficial effect on left ventricular ejection fraction (LVEF), myocardial perfusion, end-systolic volume index (ESVI). To date, large scale ongoing studies are in Phase I-II and mostly focussed on intramyocardial (IM), intracoronary (IC) or intravenous (IV) administration of vascular endothelial growth factor (VEGF) and fibroblast growth factor-23 (FGF-23) which result in the most investigated GFs in the last 10 years. Future data of ongoing randomized controlled studies will be crucial in understanding whether GF-based protocols could be in a concrete way effective in the clinical setting.
Collapse
Affiliation(s)
| | | | - Michele Vacca
- Division of Immunohematology and Transfusion Medicine, Cardarelli Hospital, Naples, Italy
| | - Claudio Napoli
- IRCCS Foundation SDN, Naples, Italy
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
13
|
Brea MS, Díaz RG, Escudero DS, Zavala MR, Portiansky EL, Villa-Abrille MC, Caldiz CI, Pérez NG, Morgan PE. Silencing of epidermal growth factor receptor reduces Na+/H+ exchanger 1 activity and hypertensive cardiac hypertrophy. Biochem Pharmacol 2019; 170:113667. [DOI: 10.1016/j.bcp.2019.113667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
|
14
|
Ogola B, Zhang Y, Iyer L, Thekkumkara T. 2-Methoxyestradiol causes matrix metalloproteinase 9-mediated transactivation of epidermal growth factor receptor and angiotensin type 1 receptor downregulation in rat aortic smooth muscle cells. Am J Physiol Cell Physiol 2018; 314:C554-C568. [DOI: 10.1152/ajpcell.00152.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Studies have demonstrated the therapeutic potential of estrogen metabolite 2-methoxyestradiol (2ME2) in several cardiovascular disorders, including hypertension. However, the exact mechanism(s) remains unknown. In this study, primary rat aortic smooth muscle cells (RASMCs) were exposed to 2ME2, and angiotensin type 1 receptor (AT1R) expression, function, and associated signaling pathways were evaluated. In RASMCs, 2ME2 downregulated AT1R expression in a concentration- and time-dependent manner, which was correlated with reduced mRNA expression. The 2ME2 effect was through G protein-coupled receptor 30 (GPR30) that inhibits second messenger cAMP. Moreover, 2ME2 exposure phosphorylated ERK1/2 that was sensitive to MEK inhibitor PD98059. Selective epidermal growth factor receptor (EGFR) inhibitor AG1478 blocked 2ME2-induced EGFR transactivation and attenuated subsequent phosphorylation of ERK1/2 preventing AT1R downregulation. The transactivation was dependent on 2ME2-induced release of matrix metalloproteinase 9 (MMP9) and epidermal growth factor demonstrated by ELISA. Furthermore, transfection with small interfering (si) RNA targeting MMP9 impeded ERK1/2 activation and AT1R downregulation in response to 2ME2 and G1 stimulation. Interestingly, under similar conditions, stimulation of GPR30 with the selective agonist G1 elicited similar signaling pathways and downregulated the AT1R expression that was reversed by GPR30 antagonist G15. Furthermore, 2ME2 and G1 inhibited angiotensin II (ANG II) induced Ca2+ release, a response consistent with AT1R downregulation. Collectively, our study demonstrates for the first time that 2ME2 binding to GPR30 induces MMP9 specific transactivation of EGFR that mediates ERK1/2-dependent downregulation of AT1R in RASMCs. The study provides critical insights into the newly discovered role and signaling pathways of 2ME2 in the regulation of AT1R in vascular cells and its potential to be developed as a therapeutic agent that ameliorates hypertension.
Collapse
Affiliation(s)
- Benard Ogola
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Yong Zhang
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Laxmi Iyer
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Thomas Thekkumkara
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| |
Collapse
|
15
|
Cappelli K, Capomaccio S, Viglino A, Silvestrelli M, Beccati F, Moscati L, Chiaradia E. Circulating miRNAs as Putative Biomarkers of Exercise Adaptation in Endurance Horses. Front Physiol 2018; 9:429. [PMID: 29740341 PMCID: PMC5928201 DOI: 10.3389/fphys.2018.00429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/06/2018] [Indexed: 12/13/2022] Open
Abstract
Endurance exercise induces metabolic adaptations and has recently been reported associated with the modulation of a particular class of small noncoding RNAs, microRNAs, that act as post-transcriptional regulators of gene expression. Released into body fluids, they termed circulating miRNAs, and they have been recognized as more effective and accurate biomarkers than classical serum markers. This study examined serum profile of miRNAs through massive parallel sequencing in response to prolonged endurance exercise in samples obtained from four competitive Arabian horses before and 2 h after the end of competition. MicroRNA identification, differential gene expression (DGE) analysis and a protein-protein interaction (PPI) network showing significantly enriched pathways of target gene clusters, were assessed and explored. Our results show modulation of more than 100 miRNAs probably arising from tissues involved in exercise responses and indicating the modulation of correlated processes as muscle remodeling, immune and inflammatory responses. Circulating miRNA high-throughput sequencing is a promising approach for sports medicine for the discovery of putative biomarkers for predicting risks related to prolonged activity and monitoring metabolic adaptations.
Collapse
Affiliation(s)
- Katia Cappelli
- Dipartimento di Medicina Veterinaria, Centro di Studio del Cavallo Sportivo, University of Perugia, Perugia, Italy
| | - Stefano Capomaccio
- Dipartimento di Medicina Veterinaria, Centro di Studio del Cavallo Sportivo, University of Perugia, Perugia, Italy
| | - Andrea Viglino
- Facoltà di Scienze Agrarie, Alimentari e Ambientali, Istituto di Zootecnica, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Maurizio Silvestrelli
- Dipartimento di Medicina Veterinaria, Centro di Studio del Cavallo Sportivo, University of Perugia, Perugia, Italy
| | - Francesca Beccati
- Dipartimento di Medicina Veterinaria, Centro di Studio del Cavallo Sportivo, University of Perugia, Perugia, Italy
| | - Livia Moscati
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche, Perugia, Italy
| | - Elisabetta Chiaradia
- Dipartimento di Medicina Veterinaria, Centro di Studio del Cavallo Sportivo, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Altara R, Zouein FA, Brandão RD, Bajestani SN, Cataliotti A, Booz GW. In Silico Analysis of Differential Gene Expression in Three Common Rat Models of Diastolic Dysfunction. Front Cardiovasc Med 2018; 5:11. [PMID: 29556499 PMCID: PMC5850854 DOI: 10.3389/fcvm.2018.00011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Standard therapies for heart failure with preserved ejection fraction (HFpEF) have been unsuccessful, demonstrating that the contribution of the underlying diastolic dysfunction pathophysiology differs from that of systolic dysfunction in heart failure and currently is far from being understood. Complicating the investigation of HFpEF is the contribution of several comorbidities. Here, we selected three established rat models of diastolic dysfunction defined by three major risk factors associated with HFpEF and researched their commonalities and differences. The top differentially expressed genes in the left ventricle of Dahl salt sensitive (Dahl/SS), spontaneous hypertensive heart failure (SHHF), and diabetes 1 induced HFpEF models were derived from published data in Gene Expression Omnibus and used for a comprehensive interpretation of the underlying pathophysiological context of each model. The diversity of the underlying transcriptomic of the heart of each model is clearly observed by the different panel of top regulated genes: the diabetic model has 20 genes in common with the Dahl/SS and 15 with the SHHF models. Advanced analytics performed in Ingenuity Pathway Analysis (IPA®) revealed that Dahl/SS heart tissue transcripts triggered by upstream regulators lead to dilated cardiomyopathy, hypertrophy of heart, arrhythmia, and failure of heart. In the heart of SHHF, a total of 26 genes were closely linked to cardiovascular disease including cardiotoxicity, pericarditis, ST-elevated myocardial infarction, and dilated cardiomyopathy. IPA Upstream Regulator analyses revealed that protection of cardiomyocytes is hampered by inhibition of the ERBB2 plasma membrane-bound receptor tyrosine kinases. Cardioprotective markers such as natriuretic peptide A (NPPA), heat shock 27 kDa protein 1 (HSPB1), and angiogenin (ANG) were upregulated in the diabetes 1 induced model; however, the model showed a different underlying mechanism with a majority of the regulated genes involved in metabolic disorders. In conclusion, our findings suggest that multiple mechanisms may contribute to diastolic dysfunction and HFpEF, and thus drug therapies may need to be guided more by phenotypic characteristics of the cardiac remodeling events than by the underlying molecular processes.
Collapse
Affiliation(s)
- Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway.,Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fouad A Zouein
- Faculty of Medicine, Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Rita Dias Brandão
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Saeed N Bajestani
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Ophthalmology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
17
|
Shim JV, Chun B, van Hasselt JGC, Birtwistle MR, Saucerman JJ, Sobie EA. Mechanistic Systems Modeling to Improve Understanding and Prediction of Cardiotoxicity Caused by Targeted Cancer Therapeutics. Front Physiol 2017; 8:651. [PMID: 28951721 PMCID: PMC5599787 DOI: 10.3389/fphys.2017.00651] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are highly potent cancer therapeutics that have been linked with serious cardiotoxicity, including left ventricular dysfunction, heart failure, and QT prolongation. TKI-induced cardiotoxicity is thought to result from interference with tyrosine kinase activity in cardiomyocytes, where these signaling pathways help to control critical processes such as survival signaling, energy homeostasis, and excitation–contraction coupling. However, mechanistic understanding is limited at present due to the complexities of tyrosine kinase signaling, and the wide range of targets inhibited by TKIs. Here, we review the use of TKIs in cancer and the cardiotoxicities that have been reported, discuss potential mechanisms underlying cardiotoxicity, and describe recent progress in achieving a more systematic understanding of cardiotoxicity via the use of mechanistic models. In particular, we argue that future advances are likely to be enabled by studies that combine large-scale experimental measurements with Quantitative Systems Pharmacology (QSP) models describing biological mechanisms and dynamics. As such approaches have proven extremely valuable for understanding and predicting other drug toxicities, it is likely that QSP modeling can be successfully applied to cardiotoxicity induced by TKIs. We conclude by discussing a potential strategy for integrating genome-wide expression measurements with models, illustrate initial advances in applying this approach to cardiotoxicity, and describe challenges that must be overcome to truly develop a mechanistic and systematic understanding of cardiotoxicity caused by TKIs.
Collapse
Affiliation(s)
- Jaehee V Shim
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Bryan Chun
- Department of Biomedical Engineering, University of VirginiaCharlottesville, VA, United States
| | - Johan G C van Hasselt
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of VirginiaCharlottesville, VA, United States
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| |
Collapse
|