1
|
Fayyaz AU, Eltony M, Prokop LJ, Koepp KE, Borlaug BA, Dasari S, Bois MC, Margulies KB, Maleszewski JJ, Wang Y, Redfield MM. Pathophysiological insights into HFpEF from studies of human cardiac tissue. Nat Rev Cardiol 2024:10.1038/s41569-024-01067-1. [PMID: 39198624 DOI: 10.1038/s41569-024-01067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 09/01/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major, worldwide health-care problem. Few therapies for HFpEF exist because the pathophysiology of this condition is poorly defined and, increasingly, postulated to be diverse. Although perturbations in other organs contribute to the clinical profile in HFpEF, altered cardiac structure, function or both are the primary causes of this heart failure syndrome. Therefore, studying myocardial tissue is fundamental to improve pathophysiological insights and therapeutic discovery in HFpEF. Most studies of myocardial changes in HFpEF have relied on cardiac tissue from animal models without (or with limited) confirmatory studies in human cardiac tissue. Animal models of HFpEF have evolved based on theoretical HFpEF aetiologies, but these models might not reflect the complex pathophysiology of human HFpEF. The focus of this Review is the pathophysiological insights gained from studies of human HFpEF myocardium. We outline the rationale for these studies, the challenges and opportunities in obtaining myocardial tissue from patients with HFpEF and relevant comparator groups, the analytical approaches, the pathophysiological insights gained to date and the remaining knowledge gaps. Our objective is to provide a roadmap for future studies of cardiac tissue from diverse cohorts of patients with HFpEF, coupling discovery biology with measures to account for pathophysiological diversity.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Muhammad Eltony
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Larry J Prokop
- Mayo Clinic College of Medicine and Science, Library Reference Service, Rochester, MN, USA
| | - Katlyn E Koepp
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Barry A Borlaug
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Mayo Clinic College of Medicine and Science, Computational Biology, Rochester, MN, USA
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joesph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ying Wang
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Margaret M Redfield
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Chaubal R, Gardi N, Joshi S, Pantvaidya G, Kadam R, Vanmali V, Hawaldar R, Talker E, Chitra J, Gera P, Bhatia D, Kalkar P, Gurav M, Shetty O, Desai S, Krishnan NM, Nair N, Parmar V, Dutt A, Panda B, Gupta S, Badwe R. Surgical Tumor Resection Deregulates Hallmarks of Cancer in Resected Tissue and the Surrounding Microenvironment. Mol Cancer Res 2024; 22:572-584. [PMID: 38394149 PMCID: PMC11148542 DOI: 10.1158/1541-7786.mcr-23-0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/24/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Surgery exposes tumor tissue to severe hypoxia and mechanical stress leading to rapid gene expression changes in the tumor and its microenvironment, which remain poorly characterized. We biopsied tumor and adjacent normal tissues from patients with breast (n = 81) and head/neck squamous cancers (HNSC; n = 10) at the beginning (A), during (B), and end of surgery (C). Tumor/normal RNA from 46/81 patients with breast cancer was subjected to mRNA-Seq using Illumina short-read technology, and from nine patients with HNSC to whole-transcriptome microarray with Illumina BeadArray. Pathways and genes involved in 7 of 10 known cancer hallmarks, namely, tumor-promoting inflammation (TNF-A, NFK-B, IL18 pathways), activation of invasion and migration (various extracellular matrix-related pathways, cell migration), sustained proliferative signaling (K-Ras Signaling), evasion of growth suppressors (P53 signaling, regulation of cell death), deregulating cellular energetics (response to lipid, secreted factors, and adipogenesis), inducing angiogenesis (hypoxia signaling, myogenesis), and avoiding immune destruction (CTLA4 and PDL1) were significantly deregulated during surgical resection (time points A vs. B vs. C). These findings were validated using NanoString assays in independent pre/intra/post-operative breast cancer samples from 48 patients. In a comparison of gene expression data from biopsy (analogous to time point A) with surgical resection samples (analogous to time point C) from The Cancer Genome Atlas study, the top deregulated genes were the same as identified in our analysis, in five of the seven studied cancer types. This study suggests that surgical extirpation deregulates the hallmarks of cancer in primary tumors and adjacent normal tissue across different cancers. IMPLICATIONS Surgery deregulates hallmarks of cancer in human tissue.
Collapse
Affiliation(s)
- Rohan Chaubal
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Nilesh Gardi
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Shalaka Joshi
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Gouri Pantvaidya
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Rasika Kadam
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Vaibhav Vanmali
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Clinical Research Secretariat, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Rohini Hawaldar
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Clinical Research Secretariat, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Elizabeth Talker
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Jaya Chitra
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Poonam Gera
- Biorepository, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Dimple Bhatia
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Prajakta Kalkar
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Mamta Gurav
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Omshree Shetty
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Sangeeta Desai
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | | | - Nita Nair
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Vani Parmar
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- 3D Printing Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Amit Dutt
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Binay Panda
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sudeep Gupta
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Rajendra Badwe
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Hypoxia and Clinical Genomics Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Yazdani A, Tiwari S, Heydarpour M. WITHDRAWN: The effect of ischemia on expression quantitative trait loci (eQTL) in human myocardium and insights into myocardial injury etiology. RESEARCH SQUARE 2024:rs.3.rs-3967889. [PMID: 38464039 PMCID: PMC10925459 DOI: 10.21203/rs.3.rs-3967889/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
26 February, 2024. Research Square has withdrawn this preprint as it was submitted and made public without the full consent of all the authors and without the full consent of the principle investigator of the registered clinical trial. Therefore, this work should not be cited as a reference.
Collapse
|
4
|
Scarpa JR, Elemento O. Multi-omic molecular profiling and network biology for precision anaesthesiology: a narrative review. Br J Anaesth 2023:S0007-0912(23)00125-3. [PMID: 37055274 DOI: 10.1016/j.bja.2023.03.006] [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: 11/11/2022] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 04/15/2023] Open
Abstract
Technological advancement, data democratisation, and decreasing costs have led to a revolution in molecular biology in which the entire set of DNA, RNA, proteins, and various other molecules - the 'multi-omic' profile - can be measured in humans. Sequencing 1 million bases of human DNA now costs US$0.01, and emerging technologies soon promise to reduce the cost of sequencing the whole genome to US$100. These trends have made it feasible to sample the multi-omic profile of millions of people, much of which is publicly available for medical research. Can anaesthesiologists use these data to improve patient care? This narrative review brings together a rapidly growing literature in multi-omic profiling across numerous fields that points to the future of precision anaesthesiology. Here, we discuss how DNA, RNA, proteins, and other molecules interact in molecular networks that can be used for preoperative risk stratification, intraoperative optimisation, and postoperative monitoring. This literature provides evidence for four fundamental insights: (1) Clinically similar patients have different molecular profiles and, as a consequence, different outcomes. (2) Vast, publicly available, and rapidly growing molecular datasets have been generated in chronic disease patients and can be repurposed to estimate perioperative risk. (3) Multi-omic networks are altered in the perioperative period and influence postoperative outcomes. (4) Multi-omic networks can serve as empirical, molecular measurements of a successful postoperative course. With this burgeoning universe of molecular data, the anaesthesiologist-of-the-future will tailor their clinical management to an individual's multi-omic profile to optimise postoperative outcomes and long-term health.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
5
|
Zhang F, Duan B, Zhou Z, Han L, Huang P, Ye Y, Wang Q, Huang F, Li J. Integration of metabolomics and transcriptomics to reveal anti-chronic myocardial ischemia mechanism of Gualou Xiebai decoction. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115530. [PMID: 35830899 DOI: 10.1016/j.jep.2022.115530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gualou Xiebai decoction (GLXB), a well-known classic traditional Chinese medicine formula, is a recorded and proven therapy for the management of cardiac diseases. However, its pharmacological characteristics and mechanism of action are unclear. MATERIALS AND METHODS The effects of GLXB and its mechanism of action in an isoprenaline-induced rat model of chronic myocardial ischemia (CMI) were investigated by incorporating metabonomics and transcriptomics. Meanwhile, the echocardiographic evaluation, histopathological analysis, serum biochemistry assay, TUNEL assay and western blot analysis were detected to revealed the protective effects of GLXB on CMI. RESULTS The results of echocardiographic evaluation, histopathological analysis and serum biochemistry assay revealed that GLXB had a significantly cardioprotective performance by reversing echocardiographic abnormalities, restoring pathological disorders and converting the serum biochemistry perturbations. Further, the omics analysis indicated that many genes and metabolites were regulated after modeling and GLXB administration, and maintained the marked "high-low" or "low-high" trends. Meanwhile, the results from integrated bioinformatics analysis suggested that the interaction network mainly consisted of amino acid and organic acid metabolism. The results of TUNEL assay and western blot analysis complemented the findings of integrated analysis of metabolomics and transcriptomics. CONCLUSION These findings suggested that GLXB has a curative effect in isoproterenol-induced CMI in rats. Integrated analysis based on transcriptomics and metabolomics studies revealed that the mechanism of GLXB in alleviating CMI was principally by the regulation of energy homeostasis and apoptosis, which was through a multi-component and multi-target treatment modality.
Collapse
Affiliation(s)
- Fengyun Zhang
- Pharmacy School, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430061, China
| | - Bailu Duan
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Zhenxiang Zhou
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Lintao Han
- Pharmacy School, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430061, China
| | - Ping Huang
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430061, China; College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yan Ye
- Pharmacy School, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430061, China
| | - Qiong Wang
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei, 430061, China; College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Fang Huang
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Jingjing Li
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
6
|
Gaballah M, Penttinen K, Kreutzer J, Mäki AJ, Kallio P, Aalto-Setälä K. Cardiac Ischemia On-a-Chip: Antiarrhythmic Effect of Levosimendan on Ischemic Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2022; 11:cells11061045. [PMID: 35326497 PMCID: PMC8947267 DOI: 10.3390/cells11061045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
Ischemic heart disease (IHD) is one of the leading causes of mortality worldwide. Preserving functionality and preventing arrhythmias of the heart are key principles in the management of patients with IHD. Levosimendan, a unique calcium (Ca2+) enhancer with inotropic activity, has been introduced into clinical usage for heart failure treatment. Human-induced pluripotent cell-derived cardiomyocytes (hiPSC-CMs) offer an opportunity to better understand the pathophysiological mechanisms of the disease as well as to serve as a platform for drug screening. Here, we developed an in vitro IHD model using hiPSC-CMs in hypoxic conditions and defined the effects of the subsequent hypoxic stress on CMs functionality. Furthermore, the effect of levosimendan on hiPSC-CMs functionality was evaluated during and after hypoxic stress. The morphology, contractile, Ca2+-handling, and gene expression properties of hiPSC-CMs were investigated in response to hypoxia. Hypoxia resulted in significant cardiac arrhythmia and decreased Ca2+ transient amplitude. In addition, disorganization of sarcomere structure was observed after hypoxia induction. Interestingly, levosimendan presented significant antiarrhythmic properties, as the arrhythmia was abolished or markedly reduced with levosimendan treatment either during or after the hypoxic stress. Moreover, levosimendan presented significant protection from the sarcomere alterations induced by hypoxia. In conclusion, this chip model appears to be a suitable preclinical representation of IHD. With this hypoxia platform, detailed knowledge of the disease pathophysiology can be obtained. The antiarrhythmic effect of levosimendan was clearly observed, suggesting a possible new clinical use for the drug.
Collapse
Affiliation(s)
- Mahmoud Gaballah
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (K.P.); (K.A.-S.)
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Menoufia 32897, Egypt
- Correspondence: ; Tel.: +358-402574148
| | - Kirsi Penttinen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (K.P.); (K.A.-S.)
| | - Joose Kreutzer
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (J.K.); (A.-J.M.); (P.K.)
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (J.K.); (A.-J.M.); (P.K.)
| | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (J.K.); (A.-J.M.); (P.K.)
| | - Katriina Aalto-Setälä
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (K.P.); (K.A.-S.)
- Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland
| |
Collapse
|
7
|
Scott SR, March KL, Wang IW, Singh K, Liu J, Turrentine M, Sen CK, Wang M. Bone marrow- or adipose-mesenchymal stromal cell secretome preserves myocardial transcriptome profile and ameliorates cardiac damage following ex vivo cold storage. J Mol Cell Cardiol 2022; 164:1-12. [PMID: 34774548 PMCID: PMC8860861 DOI: 10.1016/j.yjmcc.2021.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 10/24/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart transplantation, a life-saving approach for patients with end-stage heart disease, is limited by shortage of donor organs. While prolonged storage provides more organs, it increases the extent of ischemia. Therefore, we seek to understand molecular mechanisms underlying pathophysiological changes of donor hearts during prolonged storage. Additionally, considering mesenchymal stromal cell (MSC)-derived paracrine protection, we aim to test if MSC secretome preserves myocardial transcriptome profile and whether MSC secretome from a certain source provides the optimal protection in donor hearts during cold storage. METHODS AND RESULTS Isolated mouse hearts were divided into: no cold storage (control), 6 h cold storage (6 h-I), 6 h-I + conditioned media from bone marrow MSCs (BM-MSC CM), and 6 h-I + adipose-MSC CM (Ad-MSC CM). Deep RNA sequencing analysis revealed that compared to control, 6 h-I led to 266 differentially expressed genes, many of which were implicated in modulating mitochondrial performance, oxidative stress response, myocardial function, and apoptosis. BM-MSC CM and Ad-MSC CM restored these gene expression towards control. They also improved 6 h-I-induced myocardial functional depression, reduced inflammatory cytokine production, decreased apoptosis, and reduced myocardial H2O2. However, neither MSC-exosomes nor exosome-depleted CM recapitulated MSC CM-ameliorated apoptosis and CM-improved mitochondrial preservation during cold ischemia. Knockdown of Per2 by specific siRNA abolished MSC CM-mediated these protective effects in cardiomyocytes following 6 h cold storage. CONCLUSIONS Our results demonstrated that using MSC secretome (BM-MSCs and Ad-MSCs) during prolonged cold storage confers preservation of the normal transcriptional "fingerprint", and reduces donor heart damage. MSC-released soluble factors and exosomes may synergistically act for donor heart protection.
Collapse
Affiliation(s)
- Susan R Scott
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A
| | - Keith L March
- Division of Cardiovascular Medicine, Department of Medicine, IU School of Medicine, Indianapolis, IN, U.S.A,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, U.S.A
| | - I-wen Wang
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A,Methodist Hospital, IU Health, IU School of Medicine, Indianapolis, IN, U.S.A
| | - Kanhaiya Singh
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A,Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, U.S.A
| | - Jianyun Liu
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A
| | - Mark Turrentine
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A
| | - Chandan K Sen
- Department of Surgery, IU School of Medicine, Indianapolis, IN, U.S.A,Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, U.S.A
| | - Meijing Wang
- Department of Surgery, IU School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Wang S, Wang E, Chen Q, Yang Y, Xu L, Zhang X, Wu R, Hu X, Wu Z. Uncovering Potential lncRNAs and mRNAs in the Progression From Acute Myocardial Infarction to Myocardial Fibrosis to Heart Failure. Front Cardiovasc Med 2021; 8:664044. [PMID: 34336943 PMCID: PMC8322527 DOI: 10.3389/fcvm.2021.664044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Morbidity and mortality of heart failure (HF) post-myocardial infarction (MI) remain elevated. The aim of this study was to find potential long non-coding RNAs (lncRNAs) and mRNAs in the progression from acute myocardial infarction (AMI) to myocardial fibrosis (MF) to HF. Methods: Firstly, blood samples from AMI, MF, and HF patients were used for RNA sequencing. Secondly, differentially expressed lncRNAs and mRNAs were obtained in MF vs. AMI and HF vs. MF, followed by functional analysis of shared differentially expressed mRNAs between two groups. Thirdly, interaction networks of lncRNA-nearby targeted mRNA and lncRNA-co-expressed mRNA were constructed in MF vs. AMI and HF vs. MF. Finally, expression validation and diagnostic capability analysis of selected lncRNAs and mRNAs were performed. Results: Several lncRNA-co-expressed/nearby targeted mRNA pairs including AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 were identified. Several signaling pathways including TNF and cytokine–cytokine receptor interaction, fructose and mannose metabolism and HIF-1, hematopoietic cell lineage and fluid shear stress, and atherosclerosis and estrogen were selected. IL1R2, IRAK3, LRG1, and PLAC4 had a potential diagnostic value for both AMI and HF. Conclusion: Identified AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 lncRNA-co-expressed/nearby targeted mRNA pairs may play crucial roles in the development of AMI, MF, and HF.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Enmao Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Qincong Chen
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yan Yang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Lei Xu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaolei Zhang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Rubing Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xitian Hu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhihong Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
9
|
Yamasaki Y, Matsuura K, Sasaki D, Shimizu T. Assessment of human bioengineered cardiac tissue function in hypoxic and re-oxygenized environments to understand functional recovery in heart failure. Regen Ther 2021; 18:66-75. [PMID: 33869689 PMCID: PMC8044384 DOI: 10.1016/j.reth.2021.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/09/2021] [Accepted: 03/21/2021] [Indexed: 01/30/2023] Open
Abstract
Introduction Myocardial recovery is one of the targets for heart failure treatment. A non-negligible number of heart failure with reduced ejection fraction (EF) patients experience myocardial recovery through treatment. Although myocardial hypoxia has been reported to contribute to the progression of heart failure even in non-ischemic cardiomyopathy, the relationship between contractile recovery and re-oxygenation and its underlying mechanisms remain unclear. The present study investigated the effects of hypoxia/re-oxygenation on bioengineered cardiac cell sheets-tissue function and the underlying mechanisms. Methods Bioengineered cardiac cell sheets-tissue was fabricated with human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) using temperature-responsive culture dishes. Cardiac tissue functions in the following conditions were evaluated with a contractile force measurement system: continuous normoxia (20% O2) for 12 days; hypoxia (1% O2) for 4 days followed by normoxia (20% O2) for 8 days; or continuous hypoxia (1% O2) for 8 days. Cell number, sarcomere structure, ATP levels, mRNA expressions and Ca2+ transients of hiPSC-CM in those conditions were also assessed. Results Hypoxia (4 days) elicited progressive decreases in contractile force, maximum contraction velocity, maximum relaxation velocity, Ca2+ transient amplitude and ATP level, but sarcomere structure and cell number were not affected. Re-oxygenation (8 days) after hypoxia (4 days) was associated with progressive increases in contractile force, maximum contraction velocity and relaxation time to the similar extent levels of continuous normoxia group, while maximum relaxation velocity was still significantly low even after re-oxygenation. Ca2+ transient magnitude, cell number, sarcomere structure and ATP level after re-oxygenation were similar to those in the continuous normoxia group. Hypoxia/re-oxygenation up-regulated mRNA expression of PLN. Conclusions Hypoxia and re-oxygenation condition directly affected human bioengineered cardiac tissue function. Further understanding the molecular mechanisms of functional recovery of cardiac tissue after re-oxygenation might provide us the new insight on heart failure with recovered ejection fraction and preserved ejection fraction.
Collapse
Key Words
- ATP, adenosine triphosphate
- Cardiac cell sheet
- Contractile force
- DMEM, Dulbecco's Modified Eagle Medium
- EF, ejection fraction
- FBS, fetal bovine serum
- HFmrEF, heart failure with midrange EF
- HFpEF, heart failure with preserved EF
- HFrEF, heart failure with reduced EF
- Heart failure
- Human induced pluripotent stem cells
- Hypoxia
- NPPA, natriuretic peptide precursor A
- PLN, phospholamban
- Re-oxygenation
- SERCA, sarco/endoplasmic reticulum Ca2+ ATPase
- cTnT, cardiac troponin T
- hiPSC-CMs, human induced pluripotent stem cell-derived cardiomyocytes
Collapse
Affiliation(s)
- Yu Yamasaki
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
- Corresponding author. Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| | - Daisuke Sasaki
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
10
|
Obal D, Wu S, McKinstry-Wu A, Tawfik VL. A Guide to Understanding "State-of-the-Art" Basic Research Techniques in Anesthesiology. Anesth Analg 2020; 131:450-463. [PMID: 32371742 DOI: 10.1213/ane.0000000000004801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Perioperative medicine is changing from a "protocol-based" approach to a progressively personalized care model. New molecular techniques and comprehensive perioperative medical records allow for detection of patient-specific phenotypes that may better explain, or even predict, a patient's response to perioperative stress and anesthetic care. Basic science technology has significantly evolved in recent years with the advent of powerful approaches that have translational relevance. It is incumbent on us as a primarily clinical specialty to have an in-depth understanding of rapidly evolving underlying basic science techniques to incorporate such approaches into our own research, critically interpret the literature, and improve future anesthesia patient care. This review focuses on 3 important and most likely practice-changing basic science techniques: next-generation sequencing (NGS), clustered regularly interspaced short palindromic repeat (CRISPR) modulations, and inducible pluripotent stem cells (iPSCs). Each technique will be described, potential advantages and limitations discussed, open questions and challenges addressed, and future developments outlined. We hope to provide insight for practicing physicians when confronted with basic science articles and encourage investigators to apply "state-of-the-art" technology to their future experiments.
Collapse
Affiliation(s)
- Detlef Obal
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California.,Department of Anesthesiology, Perioperative, and Pain Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
| | - Shaogen Wu
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Andrew McKinstry-Wu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vivianne L Tawfik
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
11
|
Stone G, Choi A, Meritxell O, Gorham J, Heydarpour M, Seidman CE, Seidman JG, Aranki SF, Body SC, Carey VJ, Raby BA, Stranger BE, Muehlschlegel JD. Sex differences in gene expression in response to ischemia in the human left ventricular myocardium. Hum Mol Genet 2020; 28:1682-1693. [PMID: 30649309 DOI: 10.1093/hmg/ddz014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 01/28/2023] Open
Abstract
Sex differences exist in the prevalence, presentation and outcomes of ischemic heart disease (IHD). Females have higher risk of heart failure post-myocardial infarction relative to males and are two to three times more likely to die after coronary artery bypass grafting surgery. We examined sex differences in human myocardial gene expression in response to ischemia. Left ventricular biopsies from 68 male/46 female patients undergoing aortic valve replacement surgery were obtained at baseline and after a median 74 min of cold cardioplegic arrest/ischemia. Transcriptomes were quantified by RNA-sequencing. Cell-type enrichment analysis was used to estimate the identity and relative proportions of different cell types in each sample. A sex-specific response to ischemia was observed for 271 genes. Notably, the expression FAM5C, PLA2G4E and CYP1A1 showed an increased expression in females compared to males due to ischemia and DIO3, MT1G and CMA1 showed a decreased expression in females compared to males due to ischemia. Functional annotation analysis revealed sex-specific modulation of the oxytocin signaling pathway and common pathway of fibrin clot formation. Expression quantitative trait locus (eQTL) analysis identified variant-by-sex interaction eQTLs, indicative of sex differences in the genotypic effects on gene expression. Cell-type enrichment analysis showed sex-bias in proportion of specific cell types. Common lymphoid progenitor cells and M2 macrophages were found to increase in female samples from pre- to post-ischemia, but no change was observed in male samples. These differences in response to myocardial ischemia provide insight into the sexual dimorphism of IHD and may aid in the development of sex-specific therapies that reduce myocardial injury.
Collapse
Affiliation(s)
- Gregory Stone
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashley Choi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliva Meritxell
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jon G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sary F Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vincent J Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara E Stranger
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Raggi F, Cangelosi D, Becherini P, Blengio F, Morini M, Acquaviva M, Belli ML, Panizzon G, Cervo G, Varesio L, Eva A, Bosco MC. Transcriptome analysis defines myocardium gene signatures in children with ToF and ASD and reveals disease-specific molecular reprogramming in response to surgery with cardiopulmonary bypass. J Transl Med 2020; 18:21. [PMID: 31924244 PMCID: PMC6954611 DOI: 10.1186/s12967-020-02210-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 01/03/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tetralogy of Fallot (ToF) and Atrial Septal Defects (ASD) are the most common types of congenital heart diseases and a major cause of childhood morbidity and mortality. Cardiopulmonary bypass (CPB) is used during corrective cardiac surgery to support circulation and heart stabilization. However, this procedure triggers systemic inflammatory and stress response and consequent increased risk of postoperative complications. The aim of this study was to define the molecular bases of ToF and ASD pathogenesis and response to CPB and identify new potential biomarkers. METHODS Comparative transcriptome analysis of right atrium specimens collected from 10 ToF and 10 ASD patients was conducted before (Pre-CPB) and after (Post-CPB) corrective surgery. Total RNA isolated from each sample was individually hybridized on Affymetrix HG-U133 Plus Array Strips containing 38,500 unique human genes. Differences in the gene expression profiles and functional enrichment/network analyses were assessed using bioinformatic tools. qRT-PCR analysis was used to validate gene modulation. RESULTS Pre-CPB samples showed significant differential expression of a total of 72 genes, 28 of which were overexpressed in ToF and 44 in ASD. According to Gene Ontology annotation, the mostly enriched biological processes were represented by matrix organization and cell adhesion in ToF and by muscle development and contractility in ASD specimens. GSEA highlighted the specific enrichment of hypoxia gene sets in ToF samples, pointing to a role for hypoxia in disease pathogenesis. The post-CPB myocardium exhibited significant alterations in the expression profile of genes related to transcription regulation, growth/apoptosis, inflammation, adhesion/matrix organization, and oxidative stress. Among them, only 70 were common to the two disease groups, whereas 110 and 24 were unique in ToF and ASD, respectively. Multiple functional interactions among differentially expressed gene products were predicted by network analysis. Interestingly, gene expression changes in ASD samples followed a consensus hypoxia profile. CONCLUSION Our results provide a comprehensive view of gene reprogramming in right atrium tissues of ToF and ASD patients before and after CPB, defining specific molecular pathways underlying disease pathophysiology and myocardium response to CPB. These findings have potential translational value because they identify new candidate prognostic markers and targets for tailored cardioprotective post-surgical therapies.
Collapse
Affiliation(s)
- Federica Raggi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Davide Cangelosi
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Pamela Becherini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy
| | - Fabiola Blengio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,INSERM U955 Equipe 16, Creteil, France
| | - Martina Morini
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Massimo Acquaviva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology INSPE, Ospedale San Raffaele, Milano, Italy
| | - Maria Luisa Belli
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.,Cytomorphology Laboratory, Heamo-Onco-TMO Department, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Panizzon
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Cervo
- Department of Cardiology, IRCSS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, IRCSS Istituto Giannina Gaslini, Padiglione 2, L.go G.Gaslini 5, 16147, Genova, Italy.
| |
Collapse
|
13
|
Heydarpour M, Ejiofor J, Gilfeather M, Stone G, Gorham J, Seidman CE, Seidman JG, Yammine M, Body SC, Aranki SF, Muehlschlegel JD. Molecular Genetics of Lidocaine-Containing Cardioplegia in the Human Heart During Cardiac Surgery. Ann Thorac Surg 2018; 106:1379-1387. [PMID: 30028983 DOI: 10.1016/j.athoracsur.2018.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/14/2018] [Accepted: 06/04/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND During cardiac surgery with cardiopulmonary bypass, delivery of cardioplegia solution to achieve electromechanical cardiac quiescence is obligatory. The addition of lidocaine to cardioplegia has advantages, although its consequences at a molecular level remain unclear. We performed whole-genome RNA sequencing of the human left ventricular (LV) myocardium to elucidate the differences between whole-blood (WB) cardioplegia with and without addition of lidocaine (LC) on gene expression. METHODS We prospectively enrolled 130 patients undergoing aortic valve replacement surgery. Patients received high-potassium blood cardioplegia either with (n = 37) or without (n = 93) lidocaine. The LV apex was biopsied at baseline, and after an average of 74 minutes of cold cardioplegic arrest. We performed differential gene expression analysis for 18,258 genes between these 2 groups. Clinical and demographic variables were adjusted in the model. Gene ontology (GO) and network enrichment analysis of the retained genes were performed using g:Profiler and Cytoscape. RESULTS A total of 1,298 genes were differentially expressed between cardioplegic treatments. Compared with the WB group, genes upregulated in the LC group were identified by network enrichment to play a protective role in ischemic injury by inhibiting apoptosis, increasing transferrin endocytosis, and increasing cell viability. Downregulated genes in the LC group were identified to play a role in inflammatory diseases, oxygen transport, and neutrophil aggregation. CONCLUSIONS The addition of lidocaine to cardioplegia had pronounced effects on a molecular level with genes responsible for decreased inflammation, reduced intracellular calcium binding, enhanced antiapoptotic protection, augmented oxygen accessibility through transferrins, and increased cell viability showing measurable differences.
Collapse
Affiliation(s)
- Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julius Ejiofor
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael Gilfeather
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gregory Stone
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Josh Gorham
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Christine E Seidman
- Howard Hughes Medical Institute, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jon G Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Maroun Yammine
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sary F Aranki
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
14
|
Du Z, Shu Z, Lei W, Li C, Zeng K, Guo X, Zhao M, Tu P, Jiang Y. Integration of Metabonomics and Transcriptomics Reveals the Therapeutic Effects and Mechanisms of Baoyuan Decoction for Myocardial Ischemia. Front Pharmacol 2018; 9:514. [PMID: 29875658 PMCID: PMC5974172 DOI: 10.3389/fphar.2018.00514] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022] Open
Abstract
Myocardial ischemia (MI) is an escalating public health care burden worldwide. Baoyuan decoction (BYD) is a traditional Chinese medicine formula with cardioprotective activity; however, its pharmacological characteristics and mechanisms are obscured. Herein, a multi-omics strategy via incorporating the metabonomics, transcriptomics, and pharmacodynamics was adopted to investigate the effects and molecular mechanisms of BYD for treating MI in a rat model of left anterior descending coronary artery (LADCA) ligation. The results indicated that BYD has a significantly cardioprotective role against MI by decreasing the infarct size, converting the echocardiographic abnormalities and myocardial enzyme markers, and reversing the serum metabolic disorders and myocardial transcriptional perturbations resulting from MI. Integrated bioinformatics analysis and literature reports constructed the interaction network based on the changes of the key MI targeted-metabolites and transcripts after BYD treatment and disclosed that the cardioprotection of BYD is mainly involved in the regulation of energy homeostasis, oxidative stress, apoptosis, inflammation, cardiac contractile dysfunction, and extracellular matrix remodeling. The results of histopathological examination, quantitative RT-PCR assay, cardiac energy synthesis, and serum antioxidant assessment complemented the multi-omics findings, and indicated the multi-pathway modulation mechanisms of BYD. Our investigation demonstrated that the multi-omics approach could achieve a complementary and verified view for the comprehensive evaluation of therapeutic effects and complex mechanisms of TCMF like BYD.
Collapse
Affiliation(s)
- Zhiyong Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zeliu Shu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wei Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyu Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Mingbo Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
15
|
Martin B, Gabris-Weber BA, Reddy R, Romero G, Chattopadhyay A, Salama G. Relaxin reverses inflammatory and immune signals in aged hearts. PLoS One 2018; 13:e0190935. [PMID: 29346407 PMCID: PMC5773192 DOI: 10.1371/journal.pone.0190935] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023] Open
Abstract
Background ‘Healthy’ aging drives structural and functional changes in the heart including maladaptive electrical remodeling, fibrosis and inflammation, which lower the threshold for cardiovascular diseases such as heart failure (HF) and atrial fibrillation (AF). Despite mixed results in recent clinical trials, Relaxin-therapy for 2-days could reduce mortality by 37% at 180-days post-treatment, in patients with acute decompensated HF. Relaxin’s short life-span (hours) but long-lasting protective actions led us to test the hypothesis that relaxin acts at a genomic level to reverse maladaptive remodeling in aging and HF. Methods and results Young (9-month) and aged (24-month), male and female F-344/Brown Norway rats were treated with relaxin (0.4 mg/kg/day) for 2-weeks delivered by subcutaneous osmotic mini-pumps or with sodium acetate (controls). The genomic effects of aging and relaxin were evaluated by extracting RNA from the left ventricles and analyzing genomic changes by RNA-sequencing, Ingenuity Pathway Analysis, MetaCore and tissue immunohistochemistry. We found that aging promotes a native inflammatory response with distinct sex-differences and relaxin suppresses transcription of multiple genes and signaling pathways associated with inflammation and HF in both genders. In addition, aging significantly increased: macrophage infiltration and atrial natriuretic peptide levels in female ventricles, and activation of the complement cascade, whereas relaxin reversed these age-related effects. Conclusion These data support the hypothesis that relaxin alters gene transcription and suppresses inflammatory pathways and genes associated with HF and aging. Relaxin’s suppression of inflammation and fibrosis supports its potential as a therapy for cardiovascular and inflammation-related diseases, such as HF, AF and diabetes.
Collapse
Affiliation(s)
- Brian Martin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- School of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Beth Ann Gabris-Weber
- School of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Rajiv Reddy
- School of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Guillermo Romero
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ansuman Chattopadhyay
- Molecular Biology Information Service, Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Guy Salama
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- School of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
16
|
Abstract
Cold cardioplegia is used to induce heart arrest during cardiac surgery. However, endothelial function may be compromised after this procedure. Accordingly, interventions such as adenosine, that mimic the effects of preconditioning, may minimize endothelial injury. Herein, we investigated whether adenosine prevents cold-induced injury to the endothelium. Cultured human cardiac microvascular endothelial cells were treated with adenosine for different durations. Phosphorylation and expression of endothelial nitric oxide synthase (eNOS), p38MAPK, ERK1/2, and p70S6K6 were measured along with nitric oxide (NO) production using diaminofluorescein-2 diacetate (DAF-2DA) probe. Cold-induced injury by hypothermia to 4°C for 45 minutes to mimic conditions of cold cardioplegia during open heart surgery was induced in human cardiac microvascular endothelial cells. Under basal conditions, adenosine stimulated NO production, eNOS phosphorylation at serine 1177 from 5 minutes to 4 hours and inhibited eNOS phosphorylation at threonine 495 from 5 minutes to 6 hours, but increased phosphorylation of ERK1/2, p38MAPK, and p70S6K only after exposure for 5 minutes. Cold-induced injury inhibited NO production and the phosphorylation of the different enzymes. Importantly, adenosine prevented these effects of hypothermic injury. Our data demonstrated that adenosine prevents hypothermic injury to the endothelium by activating ERK1/2, eNOS, p70S6K, and p38MAPK signaling pathways at early time points. These findings also indicated that 5 minutes after administration of adenosine or release of adenosine is an important time window for cardioprotection during cardiac surgery.
Collapse
|
17
|
Saddic LA, Sigurdsson MI, Chang TW, Mazaika E, Heydarpour M, Shernan SK, Seidman CE, Seidman JG, Aranki SF, Body SC, Muehlschlegel JD. The Long Noncoding RNA Landscape of the Ischemic Human Left Ventricle. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.116.001534. [PMID: 28115490 DOI: 10.1161/circgenetics.116.001534] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The discovery of functional classes of long noncoding RNAs (lncRNAs) has expanded our understanding of the variety of RNA species that exist in cells. In the heart, lncRNAs have been implicated in the regulation of development, ischemic and dilated cardiomyopathy, and myocardial infarction. Nevertheless, there is a limited description of expression profiles for these transcripts in human subjects. METHODS AND RESULTS We obtained left ventricular tissue from human patients undergoing cardiac surgery and used RNA sequencing to describe an lncRNA profile. We then identified a list of lncRNAs that were differentially expressed between pairs of samples before and after the ischemic insult of cardiopulmonary bypass. The expression of some of these lncRNAs correlates with ischemic time. Coding genes in close proximity to differentially expressed lncRNAs and coding genes that have coordinated expression with these lncRNAs are enriched in functional categories related to myocardial infarction, including heart function, metabolism, the stress response, and the immune system. CONCLUSIONS We describe a list of lncRNAs that are differentially expressed after ischemia in the human heart. These genes are predicted to function in pathways consistent with myocardial injury. As a result, lncRNAs may serve as novel diagnostic and therapeutic targets for ischemic heart disease. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00985049.
Collapse
Affiliation(s)
- Louis A Saddic
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Martin I Sigurdsson
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Tzuu-Wang Chang
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Erica Mazaika
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Mahyar Heydarpour
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Stanton K Shernan
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Christine E Seidman
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Jon G Seidman
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Sary F Aranki
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Simon C Body
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA
| | - Jochen D Muehlschlegel
- From the Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital (L.A.S., M.I.S., T.-W.C., M.H., S.K.S., S.C.B., J.D.M.), Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital (S.F.A.), and Department of Genetics (E.M., C.E.S., J.G.S.), Harvard Medical School, Boston, MA.
| |
Collapse
|
18
|
Saddic LA, Nicoloro SM, Gupta OT, Czech MP, Gorham J, Shernan SK, Seidman CE, Seidman JG, Aranki SF, Body SC, Fitzgibbons TP, Muehlschlegel JD. Joint analysis of left ventricular expression and circulating plasma levels of Omentin after myocardial ischemia. Cardiovasc Diabetol 2017; 16:87. [PMID: 28687077 PMCID: PMC5501278 DOI: 10.1186/s12933-017-0567-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Omentin-1, also known as Intelectin-1 (ITLN1), is an adipokine with plasma levels associated with diabetes, obesity, and coronary artery disease. Recent studies suggest that ITLN1 can mitigate myocardial ischemic injury but the expression of ITLN1 in the heart itself has not been well characterized. The purpose of this study is to discern the relationship between the expression pattern of ITLN1 RNA in the human heart and the level of circulating ITLN1 protein in plasma from the same patients following myocardial ischemia. METHODS A large cohort of patients (n = 140) undergoing elective cardiac surgery for aortic valve replacement were enrolled in this study. Plasma and left ventricular biopsy samples were taken at the beginning of cardiopulmonary bypass and after an average of 82 min of ischemic cross clamp time. The localization of ITLN1 in epicardial adipose tissue (EAT) was also further characterized with immunoassays and cell fate transition studies. RESULTS mRNA expression of ITLN1 decreases in left ventricular tissue after acute ischemia in human patients (mean difference 280.48, p = 0.001) whereas plasma protein levels of ITLN1 increase (mean difference 5.24, p < 0.001). Immunohistochemistry localized ITLN1 to the mesothelium or visceral pericardium of EAT. Epithelial to mesenchymal transition in mesothelial cells leads to a downregulation of ITLN1 expression. CONCLUSIONS Myocardial injury leads to a decrease in ITLN1 expression in the heart and a corresponding increase in plasma levels. These changes may in part be due to an epithelial to mesenchymal transition of the cells that express ITLN1 following ischemia. Trial Registration Clinicaltrials.gov ID: NCT00985049.
Collapse
Affiliation(s)
- Louis A. Saddic
- Department of Anesthesiology and Perioperative Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Sarah M. Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical Center, Worcester, MA USA
| | | | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Stanton K. Shernan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, CWN L1, 75 Francis Street, Boston, MA 02115 USA
| | - Christine E. Seidman
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Howard Hughes Medical Institute, Boston, MA 02115 USA
| | - Jon G. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Sary F. Aranki
- Division of Cardiac Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Simon C. Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, CWN L1, 75 Francis Street, Boston, MA 02115 USA
| | - Timothy P. Fitzgibbons
- Cardiovascular Division, Department of Medicine, University of Massachusetts Medical School, Worcester, MA USA
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, CWN L1, 75 Francis Street, Boston, MA 02115 USA
| |
Collapse
|
19
|
Increased Atrial β-Adrenergic Receptors and GRK-2 Gene Expression Can Play a Fundamental Role in Heart Failure After Repair of Congenital Heart Disease with Cardiopulmonary Bypass. Pediatr Cardiol 2017; 38:734-745. [PMID: 28214967 DOI: 10.1007/s00246-017-1573-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/19/2017] [Indexed: 01/15/2023]
Abstract
Surgeries to correct congenital heart diseases are increasing in Brazil and worldwide. However, even with the advances in surgical techniques and perfusion, some cases, especially the more complex ones, can develop heart failure and death. A retrospective study of patients who underwent surgery for correction of congenital heart diseases with cardiopulmonary bypass (CPB) in a university tertiary-care hospital that died, showed infarction in different stages of evolution and scattered microcalcifications in the myocardium, even without coronary obstruction. CPB is a process routinely used during cardiac surgery for congenital heart disease. However, CPB has been related to increased endogenous catecholamines that can lead to major injuries in cardiomyocytes. The mechanisms involved are not completely understood. The aim of this study was to evaluate the alterations induced in the β-adrenergic receptors and GRK-2 present in atrial cardiomyocytes of infants with congenital heart disease undergoing surgical repair with CPB and correlate the alterations with functional and biochemical markers of ischemia/myocardial injury. The study consisted of right atrial biopsies of infants undergoing surgical correction in HC-FMRPUSP. Thirty-three cases were selected. Atrial biopsies were obtained at the beginning of CPB (group G1) and at the end of CPB (group G2). Real-time PCR, Western blotting, and immunofluorescence analysis were conducted to evaluate the expression of β1, β2-adrenergic receptors, and GRK-2 in atrial myocardium. Cardiac function was evaluated by echocardiography and biochemical analysis (N-terminal pro-brain natriuretic peptide (NT-ProBNP), lactate, and cardiac troponin I). We observed an increase in serum lactate, NT-proBNP, and troponin I at the end of CPB indicating tissue hypoxia/ischemia. Even without major clinical consequences in cardiac function, these alterations were followed by a significant increase in gene expression of β1 and β2 receptors and GRK-2, suggesting that this is one of the mechanisms responsible for the exacerbated response of cardiomyocytes to circulating catecholamines. These alterations could explain the irreversible myocardial damage and lipid peroxidation of membranes classically attributed to catecholamine excess, observed in some infants who develop heart failure and postoperative death. Although other factors may be involved, this study confirms that CPB acts as a potent inducer of increased gene expression of β- adrenergic receptors and GRK-2, making the myocardium of these infants more susceptible to the effects of circulating endogenous catecholamines, which may contribute to the development of irreversible myocardial damage and death.
Collapse
|
20
|
Sigurdsson MI, Saddic L, Heydarpour M, Chang TW, Shekar P, Aranki S, Couper GS, Shernan SK, Seidman JG, Body SC, Muehlschlegel JD. Allele-specific expression in the human heart and its application to postoperative atrial fibrillation and myocardial ischemia. Genome Med 2016; 8:127. [PMID: 27923400 PMCID: PMC5139013 DOI: 10.1186/s13073-016-0381-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/18/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Allele-specific expression (ASE) is differential expression of each of the two chromosomal alleles of an autosomal gene. We assessed ASE patterns in the human left atrium (LA, n = 62) and paired samples from the left ventricle (LV, n = 76) before and after ischemia, and tested the utility of differential ASE to identify genes associated with postoperative atrial fibrillation (poAF) and myocardial ischemia. METHODS Following genotyping from whole blood and whole-genome sequencing of LA and LV samples, we called ASE using sequences overlapping heterozygous SNPs using rigorous quality control to minimize false ASE calling. ASE patterns were compared between cardiac chambers and with a validation cohort from cadaveric tissue. ASE patterns in the LA were compared between patients who had poAF and those who did not. Changes in ASE in the LV were compared between paired baseline and post-ischemia samples. RESULTS ASE was found for 3404 (5.1%) and 8642 (4.0%) of SNPs analyzed in the LA and LV, respectively. Out of 6157 SNPs with ASE analyzed in both chambers, 2078 had evidence of ASE in both LA and LV (p < 0.0001). The SNP with the greatest ASE difference in the LA of patients with and without postoperative atrial fibrillation was within the gelsolin (GSN) gene, previously associated with atrial fibrillation in mice. The genes with differential ASE in poAF were enriched for myocardial structure genes, indicating the importance of atrial remodeling in the pathophysiology of AF. The greatest change in ASE between paired post-ischemic and baseline samples of the LV was in the zinc finger and homeodomain protein 2 (ZHX2) gene, a modulator of plasma lipids. Genes with differential ASE in ischemia were enriched in the ubiquitin ligase complex pathway associated with the ischemia-reperfusion response. CONCLUSIONS Our results establish a pattern of ASE in the human heart, with a high degree of shared ASE between cardiac chambers as well as chamber-specific ASE. Furthermore, ASE analysis can be used to identify novel genes associated with (poAF) and myocardial ischemia.
Collapse
Affiliation(s)
- Martin I Sigurdsson
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Louis Saddic
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Tzuu-Wang Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Prem Shekar
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital / Harvard Medical School, Boston, MA, USA
| | - Sary Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital / Harvard Medical School, Boston, MA, USA
| | - Gregory S Couper
- Division of Cardiac Surgery, Department of Surgery, Tufts Medical Center, Boston, MA, USA
| | - Stanton K Shernan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jon G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital / Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| |
Collapse
|
21
|
McKean DM, Homsy J, Wakimoto H, Patel N, Gorham J, DePalma SR, Ware JS, Zaidi S, Ma W, Patel N, Lifton RP, Chung WK, Kim R, Shen Y, Brueckner M, Goldmuntz E, Sharp AJ, Seidman CE, Gelb BD, Seidman JG. Loss of RNA expression and allele-specific expression associated with congenital heart disease. Nat Commun 2016; 7:12824. [PMID: 27670201 PMCID: PMC5052634 DOI: 10.1038/ncomms12824] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 08/04/2016] [Indexed: 12/22/2022] Open
Abstract
Congenital heart disease (CHD), a prevalent birth defect occurring in 1% of newborns, likely results from aberrant expression of cardiac developmental genes. Mutations in a variety of cardiac transcription factors, developmental signalling molecules and molecules that modify chromatin cause at least 20% of disease, but most CHD remains unexplained. We employ RNAseq analyses to assess allele-specific expression (ASE) and biallelic loss-of-expression (LOE) in 172 tissue samples from 144 surgically repaired CHD subjects. Here we show that only 5% of known imprinted genes with paternal allele silencing are monoallelic versus 56% with paternal allele expression-this cardiac-specific phenomenon seems unrelated to CHD. Further, compared with control subjects, CHD subjects have a significant burden of both LOE genes and ASE events associated with altered gene expression. These studies identify FGFBP2, LBH, RBFOX2, SGSM1 and ZBTB16 as candidate CHD genes because of significantly altered transcriptional expression.
Collapse
Affiliation(s)
- David M McKean
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts 02115, USA
| | - Jason Homsy
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts 02115, USA.,Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Neil Patel
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Steven R DePalma
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Howard Hughes Medical Institute, Harvard University, Boston, Massachusetts 02115, USA
| | - James S Ware
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,National Institute for Health Research Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield National Health Service Foundation Trust and Imperial College London, London SW3 6NP, UK.,National Heart and Lung Institute, Imperial College London, London SW3 6NP, UK
| | - Samir Zaidi
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Wenji Ma
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032, USA
| | - Nihir Patel
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Howard Hughes Medical Institute, Yale University, Connecticut 06510, USA
| | - Wendy K Chung
- Department of Pediatrics and Medicine, Columbia University Medical Center, New York, New York 10032, USA
| | - Richard Kim
- Section of Cardiothoracic Surgery, University of Southern California Keck School of Medicine, Los Angeles, California 90089, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Medical Center, New York, New York 10032, USA.,Department of Biomedical Informatics, Columbia University Medical Center, New York, New York 10032, USA
| | - Martina Brueckner
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Elizabeth Goldmuntz
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Andrew J Sharp
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Cardiovascular Division, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts 02115, USA.,Howard Hughes Medical Institute, Harvard University, Boston, Massachusetts 02115, USA
| | - Bruce D Gelb
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - J G Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
22
|
|
23
|
Varelmann DJ, Muehlschlegel JD. Noteworthy Articles in 2015 for the Cardiothoracic Anesthesiologist. Semin Cardiothorac Vasc Anesth 2016; 20:7-13. [PMID: 26783263 DOI: 10.1177/1089253215626729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Large multicenter, randomized controlled trials published in reputable journals had a large impact on the world of cardiothoracic anesthesia in 2015. We as cardiac anesthesiologists pride ourselves as being experts in applied physiology, physics, ultrasonography, and pharmacology/pharmacotherapy. The selected studies added to our knowledge in the fields of echocardiography, pharmacology, molecular biology, and genetics. Outcome studies shine a light on important topics that are relevant to all cardiac anesthesiologists: does surgical atrial fibrillation ablation during mitral valve surgery reduce the recurrence of atrial fibrillation at 1 year after surgery? Does remote ischemic preconditioning live up to its promise to reduce postoperative major cardiac and cerebral events? Although we still do not have the answer to all the questions, the year 2015 has been a great step toward the goal of understanding molecular mechanisms of ischemic myocardial injury and toward providing evidence-based medicine for improving patient outcome.
Collapse
Affiliation(s)
- Dirk J Varelmann
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
24
|
Study of the activated macrophage transcriptome. Exp Mol Pathol 2015; 99:575-80. [PMID: 26439118 DOI: 10.1016/j.yexmp.2015.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 11/22/2022]
Abstract
Transcriptome analysis is a powerful modern tool to study possible alterations of gene expression associated with human diseases. It turns out to be especially promising for evaluation of gene expression changes in immunopathology, as immune cells have flexible gene expression patterns that can be switched in response to infection, inflammatory stimuli and exposure to various cytokines. In particular, macrophage polarization towards pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes can be successfully studied using the modern transcriptome analysis approaches. The two mostly used techniques for transcriptome analysis are microarray and next generation sequencing. In this review we will provide an overview of known gene expression changes associated with immunopathology and discuss the advantage and limitations of different methods of transcriptome analysis.
Collapse
|
25
|
Next generation of cardiovascular studies: transcriptional responses of the human myocardium during cardiac surgery. Anesthesiology 2015; 122:486-8. [PMID: 25569809 DOI: 10.1097/aln.0000000000000581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|