1
|
Datta A, George N, Koppolu T, Kumar S U, Bithia R, Zayed H, Doss C GP. Unraveling the intricate physiological processes dysregulated in CHD-affected and Dan-Lou tablet-treated individuals. Comput Biol Chem 2024; 112:108151. [PMID: 39079284 DOI: 10.1016/j.compbiolchem.2024.108151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 09/13/2024]
Abstract
Coronary heart disease (CHD), a multifactorial cardiovascular condition, arises from the accumulation of atherosclerotic plaque in the coronary arteries, resulting in compromised blood flow to the heart and complications such as angina, myocardial infarction, or heart failure. Addressing global prevalence, risk factors, and genetics is crucial for effective management. The current study aims to identify molecular biomarkers for CHD by scrutinizing the expression patterns of differentially expressed genes (DEGs), utilizing various bioinformatic tools. In this investigation, a total of 24 samples underwent examination using the GEO2R tool. These included eight samples from individuals before treatment (GSM5434123-30), eight samples from patients after Dan-Lou tablet treatment (GSM5434131-38), and eight samples from healthy control subjects (GSM5434139-46). A suite of bioinformatics tools was used to detect enriched genes within the network, namely, Cytoscape (v3.10.1) and Molecular Complex Detection (MCODE). Functional analysis of the DEGs was conducted via clusterProfiler, a R-based package, and ClueGO. 182 and 174 DEGs corresponding to untreated and treated patient sample groups were functionally annotated for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) terms. ARF6 gene dysregulation was implicated in the myeloid cell apoptotic process (GO:0033028), regulation of actin cytoskeleton (hsa:04810), and other vital cellular functions. The myeloid cell apoptotic process (GO:0033028) was also observed to be regulated by the differential expression of the STAT5B gene. Additionally, STAT5B was found to be associated with the regulation of erythrocyte differentiation (GO:0045646). Providing targeted therapy based on the patient's idiosyncratic gene expression profiles could lead to the curing of various disorders in the near future.
Collapse
Affiliation(s)
- Ankur Datta
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Neethu George
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Tejaswini Koppolu
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Udhaya Kumar S
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India; Department of Medicine, Division Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA.
| | - R Bithia
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health and Sciences, Qatar University, QU Health, Doha 2713, Qatar.
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
2
|
Delgado-Arija M, Genovés P, Pérez-Carrillo L, González-Torrent I, Giménez-Escamilla I, Martínez-Dolz L, Portolés M, Tarazón E, Roselló-Lletí E. Plasma fibroblast activation protein is decreased in acute heart failure despite cardiac tissue upregulation. J Transl Med 2024; 22:124. [PMID: 38297310 PMCID: PMC10832198 DOI: 10.1186/s12967-024-04900-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Cardiac fibroblast activation protein (FAP) has an emerging role in heart failure (HF). A paradoxical reduction in its levels in pathological conditions associated with acute processes has been observed. We aimed to identify FAP cardiac tissue expression and its relationship with the main cardiac fibrosis-related signaling pathways, and to compare plasma FAP levels in acute and chronic HF patients. METHODS Transcriptomic changes were assessed via mRNA/ncRNA-seq in left ventricle tissue from HF patients (n = 57) and controls (n = 10). Western blotting and immunohistochemistry were used to explore FAP protein levels and localization in cardiac tissue. ELISA was performed to examine plasma FAP levels in acute HF (n = 48), chronic HF (n = 15) and control samples (n = 7). RESULTS FAP overexpression in cardiac tissue is related to the expression of molecules directly involved in cardiac fibrosis, such as POSTN, THBS4, MFAP5, COL1A2 and COL3A1 (P < 0.001), and is directly and inversely related to pro- and antifibrotic microRNAs, respectively. The observed FAP overexpression is not reflected in plasma. Circulating FAP levels were lower in acute HF patients than in controls (P < 0.05), while chronic HF patients did not show significant changes. The clinical variables analyzed, such as functional class or etiology, do not affect plasma FAP concentrations. CONCLUSIONS We determined that in HF cardiac tissue, FAP is related to the main cardiac fibrosis signaling pathways as well as to pro- and antifibrotic microRNAs. Additionally, an acute phase of HF decreases plasma FAP levels despite the upregulation observed in cardiac tissue and regardless of other clinical conditions.
Collapse
Affiliation(s)
- Marta Delgado-Arija
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Patricia Genovés
- Department of Physiology, Faculty of Medicine, Universitat de València, Avd. de Blasco Ibañez, 15, 46010, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Lorena Pérez-Carrillo
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Irene González-Torrent
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Isaac Giménez-Escamilla
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Luis Martínez-Dolz
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Manuel Portolés
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Estefanía Tarazón
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain.
| | - Esther Roselló-Lletí
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain.
| |
Collapse
|
3
|
Zhang L, Qi D, Peng M, Meng B, Wang X, Zhang X, Zuo Z, Li L, Wang Z, Zou W, Hu Z, Qian Z. Decoding molecular signature on heart of septic mice with distinct left ventricular ejection fraction. iScience 2023; 26:107825. [PMID: 37736036 PMCID: PMC10509301 DOI: 10.1016/j.isci.2023.107825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
Dysregulated cardiac function after sepsis in intensive care unit is known to predict poor long-term outcome and increase mortality. Their pathological feature and molecular mechanism remain unclear. We observed that septic patients with depressed left ventricular ejection fraction (LVEF) have the highest in-hospital and 28 days mortality comparing to patients with hyperdynamic LVEF or with heart failure with preserved LVEF. Echocardiograms reveal that survivors post cecum ligation and puncture (CLP) on rodents have stable LVEF and non-survivors have fluctuated LVEF at CLP early phase. CLP-induced mice fall into three groups based on LVEF 24 h post-surgery: high-, low-, and normal-LVEF. Transcriptomic and proteomic analyses identify jointly and distinctively changed genes, proteins and biologically essential pathways in left ventricles from three CLP groups. Notably, transmission electron microscopy shows different mitochondrial and sarcomere defects associated with LVEF variances. Together, this study systematically characterizes the molecular, morphological, and functional alterations in CLP-induced cardiac injury.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Desheng Qi
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Milin Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Binbin Meng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaolei Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhihong Zuo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Zhanwen Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Wenxuan Zou
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhonghua Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| |
Collapse
|
4
|
Lozano-Edo S, Roselló-Lletí E, Sánchez-Lázaro I, Tarazón E, Portolés M, Ezzitouny M, Lopez-Vilella R, Arnau MA, Almenar L, Martínez-Dolz L. Cardiac Allograft Rejection Induces Changes in Nucleocytoplasmic Transport: RANGAP1 as a Potential Non-Invasive Biomarker. J Pers Med 2022; 12:jpm12060913. [PMID: 35743697 PMCID: PMC9225640 DOI: 10.3390/jpm12060913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 11/16/2022] Open
Abstract
The non-invasive diagnosis of acute cellular rejection (ACR) is a major challenge. We performed a molecular study analyzing the predictive capacity of serum RanGTPase AP1 (RANGAP1) for diagnosing ACR during the first year after heart transplantation (HT). We included the serum samples of 75 consecutive HT patients, extracted after clinical stability, to determine the RANGAP1 levels through ELISA. In addition, various clinical, analytical, and echocardiographic variables, as well as endomyocardial biopsy results, were collected. RANGAP1 levels were higher in patients who developed ACR (median 63.15 ng/mL; (inter-quartile range (IQR), 36.61–105.69) vs. 35.33 ng/mL (IQR, 19.18–64.59); p = 0.02). Receiver operating characteristic (ROC) curve analysis confirmed that RANGAP1 differentiated between patients with and without ACR (area under curve (AUC), 0.70; p = 0.02), and a RANGAP1 level exceeding the cut-off point (≥90 ng/mL) was identified as a risk factor for the development of ACR (OR, 6.8; p = 0.006). Two independent predictors of ACR identified in this study were higher RANGAP1 and N-terminal pro-brain natriuretic peptide levels. The analysis of the ROC curve of the model showed a significant AUC of 0.77, p = 0.001. Our findings suggest that RANGAP1 quantification facilitates risk prediction for the occurrence of ACR and could be considered as a novel non-invasive biomarker of ACR.
Collapse
Affiliation(s)
- Silvia Lozano-Edo
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Correspondence:
| | - Esther Roselló-Lletí
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Estefanía Tarazón
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Manuel Portolés
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Maryem Ezzitouny
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
| | - Raquel Lopez-Vilella
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
| | - Miguel Angel Arnau
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
| | - Luis Almenar
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.E.); (R.L.-V.); (M.A.A.); (L.A.); (L.M.-D.)
- Clinical and Translational Research Group in Cardiology, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain; (E.R.-L.); (E.T.); (M.P.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
5
|
Electron Microscopy Reveals Evidence of Perinuclear Clustering of Mitochondria in Cardiac Biopsy-Proven Allograft Rejection. J Pers Med 2022; 12:jpm12020296. [PMID: 35207783 PMCID: PMC8878136 DOI: 10.3390/jpm12020296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 12/18/2022] Open
Abstract
Acute cellular rejection is a major complication in heart transplantation. We focus on the analysis of new ultrastructural findings in cardiac biopsy rejection based on mitochondrial intracellular organization. This study includes heart transplanted patients from a single center who were referred for endomyocardial biopsies as a scheduled routine screening. Participants were divided into two groups: patients transplanted without allograft rejection (Grade 0R), and patients with biopsy-proven allograft rejection (Grade ≥ 2R). Using electronic microscopy, we detected a significant increase in the volume density of mitochondria (p < 0.0001) and dense bodies (p < 0.01) in the rejection group. The most relevant finding was the presence of local accumulations of mitochondria close to the nuclear envelope, pressing and molding the morphology of this membrane in all rejection samples (100%). We identified this perinuclear clustering of mitochondria phenomenon in a 68 ± 27% of the total cardiac nucleus observed from rejection samples. We did not observe this phenomenon in any non-rejection samples, reflecting excellent sensitivity and specificity. We have identified a specific phenomenon affecting the architecture of the nuclear membrane—perinuclear clustering of mitochondria—in endomyocardial biopsies from patients with cardiac rejection. This ultrastructural approach might complement and improve the diagnosis of rejection.
Collapse
|
6
|
Ezzitouny M, Roselló-Lletí E, Portolés M, Sánchez-Lázaro I, Arnau-Vives MÁ, Tarazón E, Gil-Cayuela C, Lozano-Edo S, López-Vilella R, Almenar-Bonet L, Martínez-Dolz L. Value of SERCA2a as a Biomarker for the Identification of Patients with Heart Failure Requiring Circulatory Support. J Pers Med 2021; 11:jpm11111122. [PMID: 34834474 PMCID: PMC8622248 DOI: 10.3390/jpm11111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 01/03/2023] Open
Abstract
Background: Heart failure (HF) alters the nucleo-cytoplasmic transport of cardiomyocytes and reduces SERCA2a levels, essential for intracellular calcium homeostasis. We consider in this study whether the molecules involved in these processes can differentiate those patients with advanced HF and the need for mechanical circulatory support (MCS) as a bridge to recovery or urgent heart transplantation from those who are clinically stable and who are transplanted in an elective code. Material and method: Blood samples from 29 patients with advanced HF were analysed by ELISA, and the plasma levels of Importin5, Nucleoporin153 kDa, RanGTPase-Activating Protein 1 and sarcoplasmic reticulum Ca2+ ATPase were compared between patients requiring MCS and those patients without a MCS need prior to heart transplantation. Results: SERCA2a showed significantly lower levels in patients who had MCS compared to those who did not require it (0.501 ± 0.530 ng/mL vs. 1.123 ± 0.661 ng/mL; p = 0.01). A SERCA2a cut-off point of 0.84 ng/mL (AUC 0.812 ± 0.085, 95% CI: 0.646–0.979; p = 0.004) provided a 92% sensitivity, 62% specificity, 91% negative predictive value and 67% positive predictive value. Conclusions: In this cohort, patients with advanced HF and a need for MCS have shown significantly lower levels of SERCA2a as compared to stable patients without a need for MCS prior to heart transplantation. This is a small study with preliminary findings, and larger-powered dedicated studies are required to confirm and validate these results.
Collapse
Affiliation(s)
- Meryem Ezzitouny
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
- Correspondence: ; Fax: +34-96-124-58-49
| | - Esther Roselló-Lletí
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Manuel Portolés
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Miguel Ángel Arnau-Vives
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Estefanía Tarazón
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Carolina Gil-Cayuela
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
| | - Silvia Lozano-Edo
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
| | - Raquel López-Vilella
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
| | - Luis Almenar-Bonet
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| | - Luis Martínez-Dolz
- Heart Failure and Transplant Unit, Cardiology Department, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain; (I.S.-L.); (M.Á.A.-V.); (S.L.-E.); (R.L.-V.); (L.A.-B.); (L.M.-D.)
- Myocardial Dysfunction and Heart Transplant Group, Health Research Institute La Fe, 46026 Valencia, Spain; (E.R.-L.); (M.P.); (E.T.); (C.G.-C.)
- Center for Biomedical Research Network on Cardiovascular Diseases (Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares: CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
7
|
Lozano-Edo S, Sánchez-Lázaro I, Almenar-Bonet L, Portolés M, Ezzitouny M, Tarazón E, Roselló-Lleti E, Lopez-Vilella R, Martínez-Dolz L. Alterations in the Nucleocytoplasmic Transport in Heart Transplant Rejection. Transplant Proc 2021; 53:2718-2720. [PMID: 34674882 DOI: 10.1016/j.transproceed.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Nucleocytoplasmic transport is a crucial process for cell function. Previous studies have observed alterations in different molecules involved in it, relating them to ventricular function. However, there are no published data evaluating possible differences in the expression of these molecules in heart transplantation (HT) recipients. Our objective is to evaluate whether its levels are related to the appearance of cellular rejection (CR) during the first year after HT. METHODS A prospective clinical cohort that included patients undergoing HT between January 2017 and January 2019 (n = 46). Blood samples for the analysis of importin 5 (IMP5), nucleoporin 153 (Nup153); RAN-GTPaseAP1 (RanGAP1), and sarcoplasmic reticulum calcium ATPase (ATP-aseCaTransp) were collected approximately 2 months post-HT. The levels obtained were correlated with the incidence of at least moderate CR during the first year of follow-up. RESULTS Results showed that 17.39% of the patients had at least moderate CR during the first year of follow-up. Higher levels of IMP5, Nup153, and RanGAP1 were observed in this group. This difference was statistically significant in the case of Nup153 and RanGAP1 (15.94 ± 14.00 vs 28.62 ± 23.61, P = .048; 21.95 ± 15.97 vs 40.90 ± 27.16, P = .026, respectively); there was an opposite trend in the ATP-aseCaTransp case. CONCLUSION Patients with at least a moderate degree of CR during follow-up showed higher serum levels of IMP5, Nup153, and RanGAP1. The prognostic usefulness of the determination of these biomarkers and whether their elevation during follow-up would facilitate early, noninvasive identification of patients with CR remains to be clarified.
Collapse
Affiliation(s)
- Silvia Lozano-Edo
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain.
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| | - Luis Almenar-Bonet
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| | - Manuel Portolés
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| | - Maryem Ezzitouny
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Estefanía Tarazón
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| | - Esther Roselló-Lleti
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| | - Raquel Lopez-Vilella
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain; Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain; Consorcio Centro de Investigación Biomédica en Red, M.P (CIBERCV), Madrid, Spain
| |
Collapse
|
8
|
García-Manzanares M, Tarazón E, Ortega A, Gil-Cayuela C, Martínez-Dolz L, González-Juanatey JR, Lago F, Portolés M, Roselló-Lletí E, Rivera M. XPO1 Gene Therapy Attenuates Cardiac Dysfunction in Rats with Chronic Induced Myocardial Infarction. J Cardiovasc Transl Res 2020; 13:593-600. [PMID: 31768947 PMCID: PMC7423868 DOI: 10.1007/s12265-019-09932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Transcriptomic signature of XPO1 was highly expressed and inversely related to left ventricular function in ischemic cardiomyopathy patients. We hypothesized that treatment with AAV9-shXPO1 attenuates left ventricular dysfunction and remodeling in a myocardial infarction rat model. We induced myocardial infarction by coronary ligation in Sprague-Dawley rats (n = 10), which received AAV9-shXPO1 (n = 5) or placebo AAV9-scramble (n = 5) treatment. Serial echocardiographic assessment was performed throughout the study. After myocardial infarction, AAV9-shXPO1-treated rats showed partial recovery of left ventricular fractional shortening (16.8 ± 2.8 vs 24.6 ± 4.1%, P < 0.05) and a maintained left ventricular dimension (6.17 ± 0.95 vs 4.70 ± 0.93 mm, P < 0.05), which was not observed in non-treated rats. Furthermore, lower levels of EXP-1 (P < 0.05) and lower collagen fibers and fibrosis in cardiac tissue were observed. However, no differences were found in the IL-6 or TNFR1 plasma levels of the myocardium of AAV9-shXPO1 rats. AAV9-shXPO1 administration attenuates cardiac dysfunction and remodeling in rats after myocardial infarction, producing the gene silencing of XPO1.
Collapse
Affiliation(s)
- María García-Manzanares
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Estefanía Tarazón
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ana Ortega
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Carolina Gil-Cayuela
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Luis Martínez-Dolz
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University Hospital La Fe, Valencia, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Esther Roselló-Lletí
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Miguel Rivera
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
9
|
Ezzitouny M, Sánchez-Lázaro I, Rivera M, Portolés-Sanz M, Roselló-Lletí E, Gil-Cayuela C, Almenar-Bonet L, López-Vilella R, Ferré-Vallverdú M, Sanz-Sánchez J, Cerveró-Rubio A, Jiménez-Aguilella JJ, Pérez-Roselló V, Donoso-Trenado V, Arenas-Martín P, Lozano-Edo S, Jover-Pastor P, Martínez-Dolz L. Molecular Alterations of Nucleocytoplasmic Transport in Patients on the Heart Transplantation Waiting List and Its Correlation With the Severity and Etiology of Heart Failure. Transplant Proc 2019; 51:369-371. [PMID: 30879543 DOI: 10.1016/j.transproceed.2018.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/26/2018] [Accepted: 12/09/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To evaluate whether the levels of some molecules implicated in nucleocytoplasmic transport in human cardiomyocytes are related to the severity of heart failure (HF) in patients on the heart transplantation (HT) waiting list, and to determine whether there is a differential pattern of molecular alteration between ischemic cardiomyopathy (ICM) and non-ischemic dilated cardiomyopathy (DCM). METHODS Sixty-three blood samples collected before HT were analyzed to identify the levels of IMPORTIN5 (IMP5); IMPORTINalpha2; ATPaseCaTransp (ATPCa); NUCLEOPORIN153kDa (Nup153); NUCLEOPORIN160kDa (Nup160); RANGTPaseAP1 (RanGAP1) and EXPORTIN4 (EXP4). These data were then compared between patients with advanced HF with or without the need for ventricular support with extracorporeal membrane oxygenation (ECMO) as a bridge for HT, as well as between patients with non-ischemic DCM and patients with ICM. RESULTS Thirty-three patients had ICM, 26 had non-ischemic DCM, and 4 had heart disease. Seventeen patients required ventricular assistance as a bridge to HT. The levels of ATPCa, RanGAP1, and IMP5 were significantly higher in patients with ECMO, while EXP4 was significantly higher in patients without ECMO. Patients with DCM showed higher levels of IMP5, RanGAP1, and Nup153 than those with ICM. CONCLUSION Patients with advanced HF in critical condition (with ECMO as a bridge for HT) presented with significantly higher levels of ATPCa, RanGAP1, and IMP5, while patients with DCM had significantly higher levels of RanGAP1, IMP5, and Nup153. It remains to be clarified whether the determination of these molecules would facilitate the early identification of this group or if their alteration occurs as consequence of circulatory support with ECMO.
Collapse
Affiliation(s)
- M Ezzitouny
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain.
| | - I Sánchez-Lázaro
- Heart Failure and Transplant Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - M Rivera
- Health Research Institute La Fe, Valencia, Spain
| | | | | | | | - L Almenar-Bonet
- Heart Failure and Transplant Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - R López-Vilella
- Heart Failure and Transplant Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - M Ferré-Vallverdú
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - J Sanz-Sánchez
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - A Cerveró-Rubio
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | | | - V Pérez-Roselló
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - V Donoso-Trenado
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - P Arenas-Martín
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - S Lozano-Edo
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - P Jover-Pastor
- Cardiology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - L Martínez-Dolz
- Heart Failure and Transplant Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| |
Collapse
|
10
|
Moonlighting nuclear pore proteins: tissue-specific nucleoporin function in health and disease. Histochem Cell Biol 2018; 150:593-605. [PMID: 30361777 DOI: 10.1007/s00418-018-1748-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2018] [Indexed: 12/14/2022]
Abstract
The nuclear pore complex is the main transportation hub for exchange between the cytoplasm and the nucleus. It is built from nucleoporins that form distinct subcomplexes to establish this huge protein complex in the nuclear envelope. Malfunctioning of nucleoporins is well known in human malignancies, such as gene fusions of NUP214 and NUP98 in hematological neoplasms and overexpression of NUP88 in a variety of human cancers. In the past decade, the incremental utilization of next-generation sequencing has unraveled mutations in nucleoporin genes in the context of an increasing number of hereditary diseases, often in a tissue-specific manner. It emerges that, on one hand, the central nervous system and the heart are particularly sensitive to mutations in nucleoporin genes. On the other hand, nucleoporins forming the scaffold structure of the nuclear pore complex are eminently mutation-prone. These novel and exciting associations between nucleoporins and human diseases emphasize the need to shed light on these unanticipated tissue-specific roles of nucleoporins that may go well beyond their role in nucleocytoplasmic transport. In this review, the current insights into altered nucleoporin function associated with human hereditary disorders will be discussed.
Collapse
|
11
|
Preston CC, Wyles SP, Reyes S, Storm EC, Eckloff BW, Faustino RS. NUP155 insufficiency recalibrates a pluripotent transcriptome with network remodeling of a cardiogenic signaling module. BMC SYSTEMS BIOLOGY 2018; 12:62. [PMID: 29848314 PMCID: PMC5977756 DOI: 10.1186/s12918-018-0590-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/24/2018] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atrial fibrillation is a cardiac disease driven by numerous idiopathic etiologies. NUP155 is a nuclear pore complex protein that has been identified as a clinical driver of atrial fibrillation, yet the precise mechanism is unknown. The present study employs a systems biology algorithm to identify effects of NUP155 disruption on cardiogenicity in a model of stem cell-derived differentiation. METHODS Embryonic stem (ES) cell lines (n = 5) with truncated NUP155 were cultured in parallel with wild type (WT) ES cells (n = 5), and then harvested for RNAseq. Samples were run on an Illumina HiSeq 2000. Reads were analyzed using Strand NGS, Cytoscape, DAVID and Ingenuity Pathways Analysis to deconvolute the NUP155-disrupted transcriptome. Network topological analysis identified key features that controlled framework architecture and functional enrichment. RESULTS In NUP155 truncated ES cells, significant expression changes were detected in 326 genes compared to WT. These genes segregated into clusters that enriched for specific gene ontologies. Deconvolution of the collective framework into discrete sub-networks identified a module with the highest score that enriched for Cardiovascular System Development, and revealed NTRK1/TRKA and SRSF2/SC35 as critical hubs within this cardiogenic module. CONCLUSIONS The strategy of pluripotent transcriptome deconvolution used in the current study identified a novel association of NUP155 with potential drivers of arrhythmogenic AF. Here, NUP155 regulates cardioplasticity of a sub-network embedded within a larger framework of genome integrity, and exemplifies how transcriptome cardiogenicity in an embryonic stem cell genome is recalibrated by nucleoporin dysfunction.
Collapse
Affiliation(s)
- Claudia C. Preston
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N, Sioux Falls, SD 57104 USA
| | - Saranya P. Wyles
- Department of Dermatology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Santiago Reyes
- Department of Surgery, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Emily C. Storm
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N, Sioux Falls, SD 57104 USA
| | - Bruce W. Eckloff
- Medical Genome Facility, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Randolph S. Faustino
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N, Sioux Falls, SD 57104 USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD 57105 USA
| |
Collapse
|
12
|
Nagai H, Satomi T, Abiru A, Miyamoto K, Nagasawa K, Maruyama M, Yamamoto S, Kikuchi K, Fuse H, Noda M, Tsujihata Y. Antihypertrophic Effects of Small Molecules that Maintain Mitochondrial ATP Levels Under Hypoxia. EBioMedicine 2017; 24:147-158. [PMID: 28942281 PMCID: PMC5652136 DOI: 10.1016/j.ebiom.2017.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022] Open
Abstract
Since impaired mitochondrial ATP production in cardiomyocytes is thought to lead to heart failure, a drug that protects mitochondria and improves ATP production under disease conditions would be an attractive treatment option. In this study, we identified small-molecule drugs, including the anti-parasitic agent, ivermectin, that maintain mitochondrial ATP levels under hypoxia in cardiomyocytes. Mechanistically, transcriptomic analysis and gene silencing experiments revealed that ivermectin increased mitochondrial ATP production by inducing Cox6a2, a subunit of the mitochondrial respiratory chain. Furthermore, ivermectin inhibited the hypertrophic response of human induced pluripotent stem cell-derived cardiomyocytes. Pharmacological inhibition of importin β, one of the targets of ivermectin, exhibited protection against mitochondrial ATP decline and cardiomyocyte hypertrophy. These findings indicate that maintaining mitochondrial ATP under hypoxia may prevent hypertrophy and improve cardiac function, providing therapeutic options for mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hiroaki Nagai
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan.
| | - Tomoko Satomi
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Akiko Abiru
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazumasa Miyamoto
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Koji Nagasawa
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Minoru Maruyama
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Satoshi Yamamoto
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Kuniko Kikuchi
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiromitsu Fuse
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Masakuni Noda
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Yoshiyuki Tsujihata
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
13
|
Dewenter M, von der Lieth A, Katus HA, Backs J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ Res 2017; 121:1000-1020. [DOI: 10.1161/circresaha.117.310355] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium (Ca
2+
) is a universal regulator of various cellular functions. In cardiomyocytes, Ca
2+
is the central element of excitation–contraction coupling, but also impacts diverse signaling cascades and influences the regulation of gene expression, referred to as excitation–transcription coupling. Disturbances in cellular Ca
2+
-handling and alterations in Ca
2+
-dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with several excitation–transcription coupling pathways shown to be critically involved in structural and functional remodeling processes. Thus, targeting Ca
2+
-dependent transcriptional pathways might offer broad therapeutic potential. In this article, we (1) review cytosolic and nuclear Ca
2+
dynamics in cardiomyocytes with respect to their impact on Ca
2+
-dependent signaling, (2) give an overview on Ca
2+
-dependent transcriptional pathways in cardiomyocytes, and (3) discuss implications of excitation–transcription coupling in the diseased heart.
Collapse
Affiliation(s)
- Matthias Dewenter
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Albert von der Lieth
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Hugo A. Katus
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Johannes Backs
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| |
Collapse
|
14
|
Dynamic mislocalizations of nuclear pore complex proteins after focal cerebral ischemia in rat. J Neurosci Res 2016; 95:1745-1759. [DOI: 10.1002/jnr.24005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/17/2016] [Accepted: 12/02/2016] [Indexed: 12/15/2022]
|
15
|
Faustino RS, Behfar A, Groenendyk J, Wyles SP, Niederlander N, Reyes S, Puceat M, Michalak M, Terzic A, Perez-Terzic C. Calreticulin secures calcium-dependent nuclear pore competency required for cardiogenesis. J Mol Cell Cardiol 2016; 92:63-74. [PMID: 26826378 DOI: 10.1016/j.yjmcc.2016.01.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 01/20/2016] [Accepted: 01/26/2016] [Indexed: 12/24/2022]
Abstract
Calreticulin deficiency causes myocardial developmental defects that culminate in an embryonic lethal phenotype. Recent studies have linked loss of this calcium binding chaperone to failure in myofibrillogenesis through an as yet undefined mechanism. The purpose of the present study was to identify cellular processes corrupted by calreticulin deficiency that precipitate dysregulation of cardiac myofibrillogenesis related to acquisition of cardiac phenotype. In an embryonic stem cell knockout model, calreticulin deficit (crt(-/-)) compromised nucleocytoplasmic transport of nuclear localization signal-dependent and independent pathways, disrupting nuclear import of the cardiac transcription factor MEF2C. The expression of nucleoporins and associated nuclear transport proteins in derived crt(-/-) cardiomyocytes revealed an abnormal nuclear pore complex (NPC) configuration. Altered protein content in crt(-/-) cells resulted in remodeled NPC architecture that caused decreased pore diameter and diminished probability of central channel occupancy versus wild type counterparts. Ionophore treatment of impaired calcium handling in crt(-/-) cells corrected nuclear pore microarchitecture and rescued nuclear import resulting in normalized myofibrillogenesis. Thus, calreticulin deficiency alters nuclear pore function and structure, impeding myofibrillogenesis in nascent cardiomyocytes through a calcium dependent mechanism. This essential role of calreticulin in nucleocytoplasmic communication competency ties its regulatory action with proficiency of cardiac myofibrillogenesis essential for proper cardiac development.
Collapse
Affiliation(s)
- Randolph S Faustino
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Atta Behfar
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Saranya P Wyles
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Nicolas Niederlander
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Santiago Reyes
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andre Terzic
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Carmen Perez-Terzic
- Center for Regenerative Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Rehabilitation Research Center, Rochester, MN, USA.
| |
Collapse
|
16
|
|
17
|
Chahine MN, Mioulane M, Sikkel MB, O'Gara P, Dos Remedios CG, Pierce GN, Lyon AR, Földes G, Harding SE. Nuclear pore rearrangements and nuclear trafficking in cardiomyocytes from rat and human failing hearts. Cardiovasc Res 2014; 105:31-43. [PMID: 25341891 PMCID: PMC4277256 DOI: 10.1093/cvr/cvu218] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aims During cardiac hypertrophy, cardiomyocytes (CMs) increase in the size and expression of cytoskeletal proteins while reactivating a foetal gene programme. The process is proposed to be dependent on increased nuclear export and, since nuclear pore trafficking has limited capacity, a linked decrease in import. Our objective was to investigate the role of nuclear import and export in control of hypertrophy in rat and human heart failure (HF). Methods and results In myocardial tissue and isolated CMs from patients with dilated cardiomyopathy, nuclear size was increased; Nucleoporin p62, cytoplasmic RanBP1, and nuclear translocation of importins (α and β) were decreased while Exportin-1 was increased. CM from a rat HF model 16 weeks after myocardial infarction (MI) reproduced these nuclear changes. Nuclear import, determined by the rate of uptake of nuclear localization sequence (NLS)-tagged fluorescent substrate, was also decreased and this change was observed from 4 weeks after MI, before HF has developed. Treatment of isolated rat CMs with phenylephrine (PE) for 48 h produced similar cell and nuclear size increases, nuclear import and export protein rearrangement, and NLS substrate uptake decrease through p38 MAPK and HDAC-dependent pathways. The change in NLS substrate uptake occurred within 15 min of PE exposure. Inhibition of nuclear export with leptomycin B reversed established nuclear changes in PE-treated rat CMs and decreased NLS substrate uptake and cell/nuclear size in human CMs. Conclusions Nuclear transport changes related to increased export and decreased import are an early event in hypertrophic development. Hypertrophy can be prevented, or even reversed, by targeting import/export, which may open new therapeutic opportunities.
Collapse
Affiliation(s)
| | | | | | | | | | - Grant N Pierce
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Alexander R Lyon
- NHLI, Imperial College, London, UK NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | | | | |
Collapse
|
18
|
Ortega A, Roselló-Lletí E, Tarazón E, Molina-Navarro MM, Martínez-Dolz L, González-Juanatey JR, Lago F, Montoro-Mateos JD, Salvador A, Rivera M, Portolés M. Endoplasmic reticulum stress induces different molecular structural alterations in human dilated and ischemic cardiomyopathy. PLoS One 2014; 9:e107635. [PMID: 25226522 PMCID: PMC4166610 DOI: 10.1371/journal.pone.0107635] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 08/20/2014] [Indexed: 12/19/2022] Open
Abstract
Background The endoplasmic reticulum (ER) is a multifunctional organelle responsible for the synthesis and folding of proteins as well as for signalling and calcium storage, that has been linked to the contraction-relaxation process. Perturbations of its homeostasis activate a stress response in diseases such as heart failure (HF). To elucidate the alterations in ER molecular components, we analyze the levels of ER stress and structure proteins in human dilated (DCM) and ischemic (ICM) cardiomyopathies, and its relationship with patient's functional status. Methods and Results We examined 52 explanted human hearts from DCM (n = 21) and ICM (n = 21) subjects and 10 non-failing hearts as controls. Our results showed specific changes in stress (IRE1, p<0.05; p-IRE1, p<0.05) and structural (Reticulon 1, p<0.01) protein levels. The stress proteins GRP78, XBP1 and ATF6 as well as the structural proteins RRBP1, kinectin, and Nogo A and B, were upregulated in both DCM and ICM patients. Immunofluorescence results were concordant with quantified Western blot levels. Moreover, we show a novel relationship between stress and structural proteins. RRBP1, involved in procollagen synthesis and remodeling, was related with left ventricular function. Conclusions In the present study, we report the existence of alterations in ER stress response and shaping proteins. We show a plausible effect of the ER stress on ER structure in a suitable sample of DCM and ICM subjects. Patients with higher values of RRBP1 had worse left ventricular function.
Collapse
Affiliation(s)
- Ana Ortega
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Estefanía Tarazón
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | | | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | | | - Antonio Salvador
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
- * E-mail:
| |
Collapse
|
19
|
Molina-Navarro MM, Triviño JC, Martínez-Dolz L, Lago F, González-Juanatey JR, Portolés M, Rivera M. Functional networks of nucleocytoplasmic transport-related genes differentiate ischemic and dilated cardiomyopathies. A new therapeutic opportunity. PLoS One 2014; 9:e104709. [PMID: 25137373 PMCID: PMC4138080 DOI: 10.1371/journal.pone.0104709] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/10/2014] [Indexed: 01/31/2023] Open
Abstract
Heart failure provokes alterations in the expression of nucleocytoplasmic transport-related genes. To elucidate the nucleocytoplasmic transport-linked functional network underlying the two major causes of heart failure, ischemic cardiomyopathy (ICM) and dilated cardiomyopathy (DCM), we examined global transcriptome profiles of left ventricular myocardium tissue samples from 31 patients (ICM, n = 10; DCM, n = 13) undergoing heart transplantation and control donors (CNT, n = 8) using RNA-Sequencing and GeneMANIA. Comparative profiling of ICM versus control and DCM versus control showed 1081 and 2440 differentially expressed genes, respectively (>1.29-fold; P<0.05). GeneMANIA revealed differentially regulated functional networks specific to ICM and DCM. In comparison with CNT, differential expression was seen in 9 and 12 nucleocytoplasmic transport-related genes in ICM and DCM groups, respectively. DDX3X, KPNA2, and PTK2B were related to ICM, while SMURF2, NUP153, IPO5, RANBP3, NOXA1, and RHOJ were involved in DCM pathogenesis. Furthermore, the two pathologies shared 6 altered genes: XPO1, ARL4, NFKB2, FHL3, RANBP2, and RHOU showing an identical trend in expression in both ICM and DCM. Notably, the core of the derived functional networks composed of nucleocytoplasmic transport-related genes (XPO1, RANBP2, NUP153, IPO5, KPNA2, and RANBP3) branched into several pathways with downregulated genes. Moreover, we identified genes whose expression levels correlated with left ventricular mass index and left ventricular function parameters in HF patients. Collectively, our study provides a clear distinction between the two pathologies at the transcriptome level and opens up new possibilities to search for appropriate therapeutic targets for ICM and DCM.
Collapse
Affiliation(s)
| | | | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Jose Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| |
Collapse
|
20
|
Kimura M, Imamoto N. Biological significance of the importin-β family-dependent nucleocytoplasmic transport pathways. Traffic 2014; 15:727-48. [PMID: 24766099 DOI: 10.1111/tra.12174] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/22/2014] [Accepted: 04/22/2014] [Indexed: 12/19/2022]
Abstract
Importin-β family proteins (Imp-βs) are nucleocytoplasmic transport receptors (NTRs) that import and export proteins and RNAs through the nuclear pores. The family consists of 14-20 members depending on the biological species, and each member transports a specific group of cargoes. Thus, the Imp-βs mediate multiple, parallel transport pathways that can be regulated separately. In fact, the spatiotemporally differential expressions and the functional regulations of Imp-βs have been reported. Additionally, the biological significance of each pathway has been characterized by linking the function of a member of Imp-βs to a cellular consequence. Connecting these concepts, the regulation of the transport pathways conceivably induces alterations in the cellular physiological states. However, few studies have linked the regulation of an importin-β family NTR to an induced cellular response and the corresponding cargoes, despite the significance of this linkage in comprehending the biological relevance of the transport pathways. This review of recent reports on the regulation and biological functions of the Imp-βs highlights the significance of the transport pathways in physiological contexts and points out the possibility that the identification of yet unknown specific cargoes will reinforce the importance of transport regulation.
Collapse
Affiliation(s)
- Makoto Kimura
- Cellular Dynamics Laboratory, RIKEN, Hirosawa 2-1, Wako, Saitama, 351-0198, Japan
| | | |
Collapse
|
21
|
Differential gene expression of cardiac ion channels in human dilated cardiomyopathy. PLoS One 2013; 8:e79792. [PMID: 24339868 PMCID: PMC3855055 DOI: 10.1371/journal.pone.0079792] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 09/25/2013] [Indexed: 11/23/2022] Open
Abstract
Background Dilated cardiomyopathy (DCM) is characterized by idiopathic dilation and systolic contractile dysfunction of the cardiac chambers. The present work aimed to study the alterations in gene expression of ion channels involved in cardiomyocyte function. Methods and Results Microarray profiling using the Affymetrix Human Gene® 1.0 ST array was performed using 17 RNA samples, 12 from DCM patients undergoing cardiac transplantation and 5 control donors (CNT). The analysis focused on 7 cardiac ion channel genes, since this category has not been previously studied in human DCM. SCN2B was upregulated, while KCNJ5, KCNJ8, CLIC2, CLCN3, CACNB2, and CACNA1C were downregulated. The RT-qPCR (21 DCM and 8 CNT samples) validated the gene expression of SCN2B (p < 0.0001), KCNJ5 (p < 0.05), KCNJ8 (p < 0.05), CLIC2 (p < 0.05), and CACNB2 (p < 0.05). Furthermore, we performed an IPA analysis and we found a functional relationship between the different ion channels studied in this work. Conclusion This study shows a differential expression of ion channel genes involved in cardiac contraction in DCM that might partly underlie the changes in left ventricular function observed in these patients. These results could be the basis for new genetic therapeutic approaches.
Collapse
|
22
|
Meinel S, Ruhs S, Schumann K, Strätz N, Trenkmann K, Schreier B, Grosse I, Keilwagen J, Gekle M, Grossmann C. Mineralocorticoid receptor interaction with SP1 generates a new response element for pathophysiologically relevant gene expression. Nucleic Acids Res 2013; 41:8045-60. [PMID: 23821666 PMCID: PMC3783164 DOI: 10.1093/nar/gkt581] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mineralocorticoid receptor (MR) is a ligand-induced transcription factor belonging to the steroid receptor family and involved in water-electrolyte homeostasis, blood pressure regulation, inflammation and fibrosis in the renocardiovascular system. The MR shares a common hormone-response-element with the glucocorticoid receptor but nevertheless elicits MR-specific effects including enhanced epidermal growth factor receptor (EGFR) expression via unknown mechanisms. The EGFR is a receptor tyrosine kinase that leads to activation of MAP kinases, but that can also function as a signal transducer for other signaling pathways. In the present study, we mechanistically investigate the interaction between a newly discovered MR- but not glucocorticoid receptor- responsive-element (=MRE1) of the EGFR promoter, specificity protein 1 (SP1) and MR to gain general insights into MR-specificity. Biological relevance of the interaction for EGFR expression and consequently for different signaling pathways in general is demonstrated in human, rat and murine vascular smooth muscle cells and cells of EGFR knockout mice. A genome-wide promoter search for identical binding regions followed by quantitative PCR validation suggests that the identified MR-SP1-MRE1 interaction might be applicable to other genes. Overall, a novel principle of MR-specific gene expression is explored that applies to the pathophysiologically relevant expression of the EGFR and potentially also to other genes.
Collapse
Affiliation(s)
- Sandra Meinel
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, 06112 Halle/Saale, Germany, Institut für Informatik, Universität Halle-Wittenberg, 06120 Halle/Saale, Germany and Abteilung Molekulare Genetik, Leibniz-Institut für Pflanzengenetik und Kulturpflanzenforschung (IPK), 06466 Gatersleben, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Molina-Navarro MM, Roselló-Lletí E, Tarazón E, Ortega A, Sánchez-Izquierdo D, Lago F, González-Juanatey JR, García-Pavía P, Salvador A, Montero JA, Portolés M, Rivera M. Heart failure entails significant changes in human nucleocytoplasmic transport gene expression. Int J Cardiol 2013; 168:2837-43. [PMID: 23651824 DOI: 10.1016/j.ijcard.2013.03.192] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/26/2013] [Accepted: 03/30/2013] [Indexed: 01/04/2023]
Abstract
BACKGROUND Heart failure (HF) induces alterations in nucleocytoplasmic transport, which is essential to the cardiomyocyte biology. The objective of this study was to analyze the changes in gene expression in human HF, particularly focusing on nucleocytoplasmic transport-related genes. METHODS AND RESULTS 29 RNA heart samples from dilated cardiomyopathy (DCM, n = 12) and ischemic cardiomyopathy (ICM, n = 12) patients undergoing heart transplantation and control donors (CNT, n = 5) were extracted to perform a microarray profiling using Affymetrix Human Gene® 1.0 ST arrays. We focused on the study of 5 nucleocytoplasmic transport-related genes, since this functional category has not previously been studied in HF. XPO1, GABPB2, and RANBP17 were upregulated, while KALRN was downregulated in both DCM and ICM, and XPO5 only in DCM. Validation of the results by RT-qPCR increasing the total heart samples up to 41 showed a high degree of consistency with microarray results. Moreover, we observed a strong relationship between the XPO1 mRNA and robust left ventricular function parameters in ICM: left ventricular end-systolic (r = 0.81, p<0.0001) and end-diastolic diameters (r = 0.80, p<0.0001), and ejection fraction (r = -0.57, p<0.05). CONCLUSIONS We show that the expression of nucleocytoplasmic transport-related genes is altered in HF. Furthermore, XPO1 mRNA level is closely related with robust left ventricular function parameters in ICM patients. These changes may help to distinguish DCM and ICM in HF at the level of the transcriptome and provide a base for novel therapeutic approaches.
Collapse
|
24
|
Roselló-Lletí E, Alonso J, Cortés R, Almenar L, Martínez-Dolz L, Sánchez-Lázaro I, Lago F, Azorín I, Juanatey JRG, Portolés M, Rivera M. Cardiac protein changes in ischaemic and dilated cardiomyopathy: a proteomic study of human left ventricular tissue. J Cell Mol Med 2013; 16:2471-86. [PMID: 22435364 PMCID: PMC3823441 DOI: 10.1111/j.1582-4934.2012.01565.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The development of heart failure (HF) is characterized by progressive alteration of left ventricle structure and function. Previous works on proteomic analysis in cardiac tissue from patients with HF remain scant. The purpose of our study was to use a proteomic approach to investigate variations in protein expression of left ventricle tissue from patients with ischaemic (ICM) and dilated cardiomyopathy (DCM). Twenty-four explanted human hearts, 12 from patients with ICM and 12 with DCM undergoing cardiac transplantation and six non-diseased donor hearts (CNT) were analysed by 2DE. Proteins of interest were identified by mass spectrometry and validated by Western blotting and immunofluorescence. We encountered 35 differentially regulated spots in the comparison CNT versus ICM, 33 in CNT versus DCM, and 34 in ICM versus DCM. We identified glyceraldehyde 3-phophate dehydrogenase up-regulation in both ICM and DCM, and alpha-crystallin B down-regulation in both ICM and DCM. Heat shock 70 protein 1 was up-regulated only in ICM. Ten of the eleven differentially regulated proteins common to both aetiologies are interconnected as a part of a same network. In summary, we have shown by proteomics analysis that HF is associated with changes in proteins involved in the cellular stress response, respiratory chain and cardiac metabolism. Although we found altered expression of eleven proteins common to both ischaemic and dilated aetiology, we also observed different proteins altered in both groups. Furthermore, we obtained that seven of these eleven proteins are involved in cell death and apoptosis processes, and therefore in HF progression.
Collapse
Affiliation(s)
- Esther Roselló-Lletí
- Cardiocirculatory Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tarazón E, Rivera M, Roselló-Lletí E, Molina-Navarro MM, Sánchez-Lázaro IJ, España F, Montero JA, Lago F, González-Juanatey JR, Portolés M. Heart failure induces significant changes in nuclear pore complex of human cardiomyocytes. PLoS One 2012; 7:e48957. [PMID: 23152829 PMCID: PMC3495918 DOI: 10.1371/journal.pone.0048957] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 10/03/2012] [Indexed: 11/30/2022] Open
Abstract
Aims The objectives of this study were to analyse the effect of heart failure (HF) on several proteins of nuclear pore complex (NPC) and their relationship with the human ventricular function. Methods and Results A total of 88 human heart samples from ischemic (ICM, n = 52) and dilated (DCM, n = 36) patients undergoing heart transplant and control donors (CNT, n = 9) were analyzed by Western blot. Subcellular distribution of nucleoporins was analysed by fluorescence and immunocytochemistry. When we compared protein levels according to etiology, ICM showed significant higher levels of NDC1 (65%, p<0.0001), Nup160 (88%, p<0.0001) and Nup153 (137%, p = 0.004) than those of the CNT levels. Furthermore, DCM group showed significant differences for NDC1 (41%, p<0.0001), Nup160 (65%, p<0.0001), Nup153 (155%, p = 0.006) and Nup93 (88%, p<0.0001) compared with CNT. However, Nup155 and translocated promoter region (TPR) did not show significant differences in their levels in any etiology. Regarding the distribution of these proteins in cell nucleus, only NDC1 showed differences in HF. In addition, in the pathological group we obtained good relationship between the ventricular function parameters (LVEDD and LVESD) and Nup160 (r = −0382, p = 0.004; r = −0.290, p = 0.033; respectively). Conclusions This study shows alterations in specific proteins (NDC1, Nup160, Nup153 and Nup93) that compose NPC in ischaemic and dilated human heart. These changes, related to ventricular function, could be accompanied by alterations in the nucleocytoplasmic transport. Therefore, our findings may be the basis for a new approach to HF management.
Collapse
Affiliation(s)
- Estefanía Tarazón
- Cardiocirculatory Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | | | - Ignacio José Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Cardiology Department, Hospital Universitario La Fe, Valencia, Spain
| | - Francisco España
- Biochemistry Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | | | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Cell Biology and Pathology Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
- * E-mail:
| |
Collapse
|
26
|
García-Rúa V, Otero MF, Lear PV, Rodríguez-Penas D, Feijóo-Bandín S, Noguera-Moreno T, Calaza M, Álvarez-Barredo M, Mosquera-Leal A, Parrington J, Brugada J, Portolés M, Rivera M, González-Juanatey JR, Lago F. Increased expression of fatty-acid and calcium metabolism genes in failing human heart. PLoS One 2012; 7:e37505. [PMID: 22701570 PMCID: PMC3368932 DOI: 10.1371/journal.pone.0037505] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/19/2012] [Indexed: 01/08/2023] Open
Abstract
Background Heart failure (HF) involves alterations in metabolism, but little is known about cardiomyopathy-(CM)-specific or diabetes-independent alterations in gene expression of proteins involved in fatty-acid (FA) uptake and oxidation or in calcium-(Ca2+)-handling in the human heart. Methods RT-qPCR was used to quantify mRNA expression and immunoblotting to confirm protein expression in left-ventricular myocardium from patients with HF (n = 36) without diabetes mellitus of ischaemic (ICM, n = 16) or dilated (DCM, n = 20) cardiomyopathy aetiology, and non-diseased donors (CTL, n = 6). Results Significant increases in mRNA of genes regulating FA uptake (CD36) and intracellular transport (Heart-FA-Binding Protein (HFABP)) were observed in HF patients vs CTL. Significance was maintained in DCM and confirmed at protein level, but not in ICM. mRNA was higher in DCM than ICM for peroxisome-proliferator-activated-receptor-alpha (PPARA), PPAR-gamma coactivator-1-alpha (PGC1A) and CD36, and confirmed at the protein level for PPARA and CD36. Transcript and protein expression of Ca2+-handling genes (Two-Pore-Channel 1 (TPCN1), Two-Pore-Channel 2 (TPCN2), and Inositol 1,4,5-triphosphate Receptor type-1 (IP3R1)) increased in HF patients relative to CTL. Increases remained significant for TPCN2 in all groups but for TPCN1 only in DCM. There were correlations between FA metabolism and Ca2+-handling genes expression. In ICM there were six correlations, all distinct from those found in CTL. In DCM there were also six (all also different from those found in CTL): three were common to and three distinct from ICM. Conclusion DCM-specific increases were found in expression of several genes that regulate FA metabolism, which might help in the design of aetiology-specific metabolic therapies in HF. Ca2+-handling genes TPCN1 and TPCN2 also showed increased expression in HF, while HF- and CM-specific positive correlations were found among several FA and Ca2+-handling genes.
Collapse
Affiliation(s)
- Vanessa García-Rúa
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Manuel Francisco Otero
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
- Department of Clinical Chemistry, University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Pamela Virginia Lear
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Diego Rodríguez-Penas
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Sandra Feijóo-Bandín
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Teresa Noguera-Moreno
- Unit of Biostatistical Research, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Calaza
- Laboratory 10, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - María Álvarez-Barredo
- Department of Cardiology, University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Ana Mosquera-Leal
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - John Parrington
- Department of Pharmacology, Oxford University, Oxford, United Kingdom
| | - Josep Brugada
- Cardiology Department, Thorax Institute, Hospital Clinic, Barcelona, Spain
| | | | | | - José Ramón González-Juanatey
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
- Department of Cardiology, University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
| | - Francisca Lago
- Laboratory of Cellular and Molecular Cardiology, Santiago Institute of Biomedical Research (IDIS), University of Santiago de Compostela Clinical Hospital (CHUS), Santiago de Compostela, Spain
- * E-mail:
| |
Collapse
|
27
|
Differences in MEF2 and NFAT transcriptional pathways according to human heart failure aetiology. PLoS One 2012; 7:e30915. [PMID: 22363514 PMCID: PMC3281902 DOI: 10.1371/journal.pone.0030915] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 12/29/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Ca(2+) handling machinery modulates the activation of cardiac transcription pathways involved in heart failure (HF). The present study investigated the effect of HF aetiology on Ca(+2) handling proteins and NFAT1, MEF2C and GATA4 (transcription factors) in the same cardiac tissue. METHODOLOGY AND PRINCIPAL FINDINGS A total of 83 hearts from ischemic (ICM, n = 43) and dilated (DCM, n = 31) patients undergoing heart transplantation and controls (CNT, n = 9) were analyzed by western blotting. Subcellular distribution was analyzed by fluorescence and electron microscopy. When we compared Ca(+2) handling proteins according to HF aetiology, ICM showed higher levels of calmodulin (24%, p<0.01), calcineurin (26%, p<0.01) and Ca(2+)/Calmodulin-dependent kinase II (CaMKIIδ(b) nuclear isoform 62%, p<0.001) than the CNT group. However, these proteins in DCM did not significantly increase. Furthermore, ICM showed a significant elevation in MEF2C (33%, p<0.01), and GATA4 (49%, p<0.05); also NFAT1 (66%, p<0.001) was increased, producing the resultant translocation of this transcriptional factor into the nuclei. These results were supported by fluorescence and electron microscopy analysis. Whereas, DCM only had a significant increase in GATA4 (52%, p<0.05). Correlations between NFAT1 and MEF2C in both groups (ICM r = 0.38 and DCM r = 0.59, p<0.05 and p<0.01, respectively) were found; only ICM showed a correlation between GATA4 and NFAT1 (r = 0.37, p<0.05). CONCLUSIONS/SIGNIFICANCE This study shows an increase of Ca(2+) handling machinery synthesis and their cardiac transcription pathways in HF, being more markedly increased in ICM. Furthermore, there is a significant association between MEF2, NFAT1 and GATA4. These proteins could be therapeutic targets to improve myocardial function.
Collapse
|
28
|
Roselló-Lletí E, Rivera M, Cortés R, Azorín I, Sirera R, Martínez-Dolz L, Hove L, Cinca J, Lago F, González-Juanatey JR, Salvador A, Portolés M. Influence of heart failure on nucleolar organization and protein expression in human hearts. Biochem Biophys Res Commun 2012; 418:222-8. [PMID: 22244875 DOI: 10.1016/j.bbrc.2011.12.151] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 12/31/2011] [Indexed: 10/14/2022]
Abstract
We investigate for the first time the influence of heart failure (HF) on nucleolar organization and proteins in patients with ischemic (ICM) or dilated cardiomyopathy (DCM). A total of 71 human hearts from ICM (n=38) and DCM (n=27) patients, undergoing heart transplantation and control donors (n=6), were analysed by western-blotting, RT-PCR and cell biology methods. When we compared protein levels according to HF etiology, nucleolin was increased in both ICM (117%, p<0.05) and DCM (141%, p<0.01). Moreover, mRNA expression were also upregulated in ICM (1.46-fold, p<0.05) and DCM (1.70-fold, p<0.05. Immunofluorescence studies showed that the highest intensity of nucleolin was into nucleolus (p<0.0001), and it was increased in pathological hearts (p<0.0001). Ultrastructure analysis by electron microscopy showed an increase in the nucleus and nucleolus size in ICM (17%, p<0.05 and 131%, p<0.001) and DCM (56%, p<0.01 and 69%, p<0.01). Nucleolar organization was influenced by HF irrespective of etiology, increasing fibrillar centers (p<0.001), perinucleolar chromatin (p<0.01) and dense fibrillar components (p<0.01). Finally, left ventricular function parameters were related with nucleolin levels in ischemic hearts (p<0.0001). The present study demonstrates that HF influences on morphology and organization of nucleolar components, revealing changes in the expression and in the levels of nucleolin protein.
Collapse
Affiliation(s)
- Esther Roselló-Lletí
- Cardiocirculatory Unit, Research Center, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
D'Angelo MA, Gomez-Cavazos JS, Mei A, Lackner DH, Hetzer MW. A change in nuclear pore complex composition regulates cell differentiation. Dev Cell 2012; 22:446-58. [PMID: 22264802 DOI: 10.1016/j.devcel.2011.11.021] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 10/06/2011] [Accepted: 11/14/2011] [Indexed: 12/11/2022]
Abstract
Nuclear pore complexes (NPCs) are built from ∼30 different proteins called nucleoporins or Nups. Previous studies have shown that several Nups exhibit cell-type-specific expression and that mutations in NPC components result in tissue-specific diseases. Here we show that a specific change in NPC composition is required for both myogenic and neuronal differentiation. The transmembrane nucleoporin Nup210 is absent in proliferating myoblasts and embryonic stem cells (ESCs) but becomes expressed and incorporated into NPCs during cell differentiation. Preventing Nup210 production by RNAi blocks myogenesis and the differentiation of ESCs into neuroprogenitors. We found that the addition of Nup210 to NPCs does not affect nuclear transport but is required for the induction of genes that are essential for cell differentiation. Our results identify a single change in NPC composition as an essential step in cell differentiation and establish a role for Nup210 in gene expression regulation and cell fate determination.
Collapse
Affiliation(s)
- Maximiliano A D'Angelo
- Salk Institute for Biological Studies, Molecular and Cell Biology Laboratory, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
30
|
David-Watine B. Silencing nuclear pore protein Tpr elicits a senescent-like phenotype in cancer cells. PLoS One 2011; 6:e22423. [PMID: 21811608 PMCID: PMC3139644 DOI: 10.1371/journal.pone.0022423] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/22/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Tpr is a large coiled-coil protein located in the nuclear basket of the nuclear pore complex for which many different functions were proposed from yeast to human. METHODOLOGY/PRINCIPAL FINDINGS Here we show that depletion of Tpr by RNA interference triggers G0-G1 arrest and ultimately induces a senescent-like phenotype dependent on the presence of p53. We also found that Tpr depletion impairs the NES [nuclear export sequence]-dependent nuclear export of proteins and causes partial co-depletion of Nup153. In addition Tpr depletion impacts on level and function of the SUMO-protease SENP2 thus affecting SUMOylation regulation at the nuclear pore and overall SUMOylation in the cell. CONCLUSIONS Our data for the first time provide evidence that a nuclear pore component plays a role in controlling cellular senescence. Our findings also point to new roles for Tpr in the regulation of SUMO-1 conjugation at the nuclear pore and directly confirm Tpr involvement in the nuclear export of NES-proteins.
Collapse
Affiliation(s)
- Brigitte David-Watine
- Institut Pasteur, CNRS URA2582, Groupe E3 Biologie Cellulaire du Noyau, Paris, France.
| |
Collapse
|
31
|
Hall MN, Corbett AH, Pavlath GK. Regulation of nucleocytoplasmic transport in skeletal muscle. Curr Top Dev Biol 2011; 96:273-302. [PMID: 21621074 DOI: 10.1016/b978-0-12-385940-2.00010-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Proper skeletal muscle function is dependent on spatial and temporal control of gene expression in multinucleated myofibers. In addition, satellite cells, which are tissue-specific stem cells that contribute critically to repair and maintenance of skeletal muscle, are also required for normal muscle physiology. Gene expression in both myofibers and satellite cells is dependent upon nuclear proteins that require facilitated nuclear transport. A unique challenge for myofibers is controlling the transcriptional activity of hundreds of nuclei in a common cytoplasm yet achieving nuclear selectivity in transcription at specific locations such as neuromuscular synapses and myotendinous junctions. Nucleocytoplasmic transport of macromolecular cargoes is regulated by a complex interplay among various components of the nuclear transport machinery, namely nuclear pore complexes, nuclear envelope proteins, and various soluble transport receptors. The focus of this review is to highlight what is known about the nuclear transport machinery and its regulation in skeletal muscle and to consider the unique challenges that multinucleated muscle cells as well as satellite cells encounter in regulating nucleocytoplasmic transport during cell differentiation and tissue adaptation. Understanding how regulated nucleocytoplasmic transport controls gene expression in skeletal muscle may lead to further insights into the mechanisms contributing to muscle growth and maintenance throughout the lifespan of an individual.
Collapse
Affiliation(s)
- Monica N Hall
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
32
|
Jamali T, Jamali Y, Mehrbod M, Mofrad MRK. Nuclear pore complex: biochemistry and biophysics of nucleocytoplasmic transport in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:233-86. [PMID: 21414590 DOI: 10.1016/b978-0-12-386043-9.00006-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nuclear pore complexes (NPCs) are the gateways connecting the nucleoplasm and cytoplasm. This structures are composed of over 30 different proteins and 60-125 MDa of mass depending on type of species. NPCs are bilateral pathways that selectively control the passage of macromolecules into and out of the nucleus. Molecules smaller than 40 kDa diffuse through the NPC passively while larger molecules require facilitated transport provided by their attachment to karyopherins. Kinetic studies have shown that approximately 1000 translocations occur per second per NPC. Maintaining its high selectivity while allowing for rapid translocation makes the NPC an efficient chemical nanomachine. In this review, we approach the NPC function via a structural viewpoint. Putting together different pieces of this puzzle, this chapter confers an overall insight into what molecular processes are engaged in import/export of active cargos across the NPC and how different transporters regulate nucleocytoplasmic transport. In the end, the correlation of several diseases and disorders with the NPC structural defects and dysfunctions is discussed.
Collapse
Affiliation(s)
- T Jamali
- Department of Bioengineering, University of California, Berkeley, California, USA
| | | | | | | |
Collapse
|