1
|
Wirth L, Erny E, Krane M, Lahm H, Hein L, Gilsbach R, Lother A. Gene expression networks in endothelial cells from failing human hearts. Am J Physiol Heart Circ Physiol 2024; 327:H573-H581. [PMID: 39028282 DOI: 10.1152/ajpheart.00425.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Chronic heart failure is associated with adverse remodeling of the heart that is typically characterized by cardiomyocyte hypertrophy. This requires the formation of new capillaries to maintain oxygen supply. Insufficient angiogenesis promotes the transition from compensated hypertrophy into heart failure. The aim of this study was to identify angiogenesis-related gene networks and corresponding regulatory hubs in endothelial cells from failing human hearts. We isolated left ventricular endothelial cells from patients with advanced heart failure undergoing left ventricular assist device surgery (n = 15) and healthy organ donors (n = 2) and performed RNA sequencing. Subgroup analysis revealed no impact of comorbidities on gene expression. In a weighted gene coexpression network analysis, we found 26 gene clusters, of which 9 clusters showed a significant positive or negative correlation with the presence of heart failure. We identified the transcription factors CASZ1 (castor zinc finger 1), ZNF523 (zinc finger protein 523), and NFE2L1 (nuclear factor erythroid 2-related factor 1) as hub genes of a cluster related to angiogenesis. Knockdown of CASZ1, ZNF523, or NFE2L1 in human umbilical vein endothelial cells led to a downregulation of genes from the respective cluster, including CD34 and platelet-derived growth factor-β, confirming their regulatory function. In conclusion, we assessed gene networks in endothelial cells and identified transcription factors CASZ1, ZNF532, and NFE2L1 as potential regulators of angiogenesis in failing human hearts. Our study provides insights into the transcriptional regulation of angiogenesis beyond the classical vascular endothelial growth factor signaling pathway.NEW & NOTEWORTHY Gene coexpression network analysis defined 26 gene clusters expressed in endothelial cells from failing human hearts. Transcription factors CASZ1, ZNF523, and NFE2L1 were identified as hub genes of a cluster related to angiogenesis. Knockdown of CASZ1, ZNF523, or NFE2L1 in human umbilical vein endothelial cells led to a downregulation of genes from the respective cluster, confirming their regulatory function. This provides insights into the transcriptional regulation of angiogenesis in heart failure beyond classical signaling pathways.
Collapse
Affiliation(s)
- Luisa Wirth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elias Erny
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, Institute Insure, German Heart Center Munich, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Division of Cardiac Surgery, Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, United States
- German Center for Cardiovascular Research (DZHK) - Partner Site Munich Heart Alliance, Munich, Germany
| | - Harald Lahm
- Department of Cardiovascular Surgery, Institute Insure, German Heart Center Munich, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Interdisciplinary Medical Intensive Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Zhu Y, Chen Y, Zu Y. Leveraging a neutrophil-derived PCD signature to predict and stratify patients with acute myocardial infarction: from AI prediction to biological interpretation. J Transl Med 2024; 22:612. [PMID: 38956669 PMCID: PMC11221097 DOI: 10.1186/s12967-024-05415-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Programmed cell death (PCD) has recently been implicated in modulating the removal of neutrophils recruited in acute myocardial infarction (AMI). Nonetheless, the clinical significance and biological mechanism of neutrophil-related PCD remain unexplored. METHODS We employed an integrative machine learning-based computational framework to generate a predictive neutrophil-derived PCD signature (NPCDS) within five independent microarray cohorts from the peripheral blood of AMI patients. Non-negative matrix factorization was leveraged to develop an NPCDS-based AMI subtype. To elucidate the biological mechanism underlying NPCDS, we implemented single-cell transcriptomics on Cd45+ cells isolated from the murine heart of experimental AMI. We finally conducted a Mendelian randomization (MR) study and molecular docking to investigate the therapeutic value of NPCDS on AMI. RESULTS We reported the robust and superior performance of NPCDS in AMI prediction, which contributed to an optimal combination of random forest and stepwise regression fitted on nine neutrophil-related PCD genes (MDM2, PTK2B, MYH9, IVNS1ABP, MAPK14, GNS, MYD88, TLR2, CFLAR). Two divergent NPCDS-based subtypes of AMI were revealed, in which subtype 1 was characterized as inflammation-activated with more vibrant neutrophil activities, whereas subtype 2 demonstrated the opposite. Mechanically, we unveiled the expression dynamics of NPCDS to regulate neutrophil transformation from a pro-inflammatory phase to an anti-inflammatory phase in AMI. We uncovered a significant causal association between genetic predisposition towards MDM2 expression and the risk of AMI. We also found that lidoflazine, isotetrandrine, and cepharanthine could stably target MDM2. CONCLUSION Altogether, NPCDS offers significant implications for prediction, stratification, and therapeutic management for AMI.
Collapse
Affiliation(s)
- Yihao Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Yuxi Chen
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China.
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China.
- Marine Biomedical Science and Technology Innovation Platform of Lin-Gang Special Area, Shanghai, 201306, People's Republic of China.
| |
Collapse
|
3
|
Qi T, Zhang J, Zhang K, Zhang W, Song Y, Lian K, Kan C, Han F, Hou N, Sun X. Unraveling the role of the FHL family in cardiac diseases: Mechanisms, implications, and future directions. Biochem Biophys Res Commun 2024; 694:149468. [PMID: 38183876 DOI: 10.1016/j.bbrc.2024.149468] [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: 11/07/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Heart diseases are a major cause of morbidity and mortality worldwide. Understanding the molecular mechanisms underlying these diseases is essential for the development of effective diagnostic and therapeutic strategies. The FHL family consists of five members: FHL1, FHL2, FHL3, FHL4, and FHL5/Act. These members exhibit different expression patterns in various tissues including the heart. FHL family proteins are implicated in cardiac remodeling, regulation of metabolic enzymes, and cardiac biomechanical stress perception. A large number of studies have explored the link between FHL family proteins and cardiac disease, skeletal muscle disease, and ovarian metabolism, but a comprehensive and in-depth understanding of the specific molecular mechanisms targeting FHL on cardiac disease is lacking. The aim of this review is to explore the structure and function of FHL family members, to comprehensively elucidate the mechanisms by which they regulate the heart, and to explore in depth the changes in FHL family members observed in different cardiac disorders, as well as the effects of mutations in FHL proteins on heart health.
Collapse
Affiliation(s)
- Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Lian
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
4
|
Li Y, Bertozzi A, Mann MRW, Kühn B. Interdependent changes of nuclear lamins, nuclear pore complexes, and ploidy regulate cellular regeneration and stress response in the heart. Nucleus 2023; 14:2246310. [PMID: 37606283 PMCID: PMC10446781 DOI: 10.1080/19491034.2023.2246310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
In adult mammals, many heart muscle cells (cardiomyocytes) are polyploid, do not proliferate (post-mitotic), and, consequently, cannot contribute to heart regeneration. In contrast, fetal and neonatal heart muscle cells are diploid, proliferate, and contribute to heart regeneration. We have identified interdependent changes of the nuclear lamina, nuclear pore complexes, and DNA-content (ploidy) in heart muscle cell maturation. These results offer new perspectives on how cells alter their nuclear transport and, with that, their gene regulation in response to extracellular signals. We present how changes of the nuclear lamina alter nuclear pore complexes in heart muscle cells. The consequences of these changes for cellular regeneration and stress response in the heart are discussed.
Collapse
Affiliation(s)
- Yao Li
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alberto Bertozzi
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mellissa RW Mann
- Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Bernhard Kühn
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Normalization strategy for selection of reference genes for RT-qPCR analysis in left ventricles of failing human hearts. BMC Cardiovasc Disord 2022; 22:180. [PMID: 35439923 PMCID: PMC9019989 DOI: 10.1186/s12872-022-02614-9] [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: 08/27/2021] [Accepted: 03/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background Quantitative RT-PCR is a valuable tool for assessing the gene expression in different human tissues, particularly due to its exceptional sensitivity, accuracy and reliability. However, the choice of adequate control for normalization is a crucial step, greatly affecting the results of all subsequent analyses. So far, only a few studies were focused on the selection of optimal reference genes in left ventricles of failing human hearts, leading to several disparities in experimental results focused on differential gene expression in this area. Therefore, the main objective of this study was to identify a set of suitable reference genes in normal and failing left ventricle tissues, which could increase the reliability of RT-qPCR-based studies in the future. Methods We analyzed the expression of 15 commonly used housekeeping genes (ACTB, B2M, GAPDH, GUSB, HMBS, HPRT1, IPO8, PGK1, POLR2A, PPIA, RPLP0, TBP, TFRC, UBC and YWHAZ) in left ventricles of normal and failed hearts with two-step approach. In the first step, we excluded genes which are variantly expressed using ANOVA-based statistical method. Afterwards, the remaining genes were analyzed using geNorm, NormFinder and BestKeeper algorithms, together with delta Cq method. Finally, the geometric mean of gene rankings across all methods was calculated. Results Our analysis identified IPO8 and POLR2A as the most stably expressed genes, whereas ACTB and B2M were found to be expressed variantly, suggesting a potential role of these genes in the pathophysiological processes in failing human hearts. Discussion/conclusion Using our two-step approach, we identified and validated two reference genes expressed invariantly in left ventricles of both healthy and failing human hearts, as well as provided a guideline for the selection of reference genes in studies comparing gene expression in these types of tissues. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02614-9.
Collapse
|
6
|
van der Pijl RJ, Domenighetti AA, Sheikh F, Ehler E, Ottenheijm CAC, Lange S. The titin N2B and N2A regions: biomechanical and metabolic signaling hubs in cross-striated muscles. Biophys Rev 2021; 13:653-677. [PMID: 34745373 PMCID: PMC8553726 DOI: 10.1007/s12551-021-00836-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Muscle specific signaling has been shown to originate from myofilaments and their associated cellular structures, including the sarcomeres, costameres or the cardiac intercalated disc. Two signaling hubs that play important biomechanical roles for cardiac and/or skeletal muscle physiology are the N2B and N2A regions in the giant protein titin. Prominent proteins associated with these regions in titin are chaperones Hsp90 and αB-crystallin, members of the four-and-a-half LIM (FHL) and muscle ankyrin repeat protein (Ankrd) families, as well as thin filament-associated proteins, such as myopalladin. This review highlights biological roles and properties of the titin N2B and N2A regions in health and disease. Special emphasis is placed on functions of Ankrd and FHL proteins as mechanosensors that modulate muscle-specific signaling and muscle growth. This region of the sarcomere also emerged as a hotspot for the modulation of passive muscle mechanics through altered titin phosphorylation and splicing, as well as tethering mechanisms that link titin to the thin filament system.
Collapse
Affiliation(s)
| | - Andrea A. Domenighetti
- Shirley Ryan AbilityLab, Chicago, IL USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL USA
| | - Farah Sheikh
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Coen A. C. Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ USA
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Stephan Lange
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
8
|
Dong K, He X, Su H, Fulton DJR, Zhou J. Genomic analysis of circular RNAs in heart. BMC Med Genomics 2020; 13:167. [PMID: 33160353 PMCID: PMC7648966 DOI: 10.1186/s12920-020-00817-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Heart failure is a leading cause of human morbidity and mortality. Circular RNAs (circRNAs) are a newly discovered class of RNA that have been found to have important physiological and pathological roles. In the current study, we de novo analyzed existing whole transcriptome data from 5 normal and 5 dilated cardiomyopathy (DCM) human heart samples and compared the results with circRNAs that have been previously reported in human, mouse and rat hearts. RESULTS Our analysis identifies a list of cardiac circRNAs that are reliably detected in multiple studies. We have also defined the top 30 most abundant circRNAs in healthy human hearts which include some with previously unrecognized cardiac roles such as circHIPK3_11 and circTULP4_1. We further found that many circRNAs are dysregulated in DCM, particularly transcripts originating from DCM-related gene loci, such as TTN and RYR2. In addition, we predict the potential of cardiac circRNAs to sponge miRNAs that have reported roles in heart disease. We found that circALMS1_6 has the highest potential to bind miR-133, a microRNA that can regulate cardiac remodeling. Interestingly, we detected a novel class of circRNAs, referred to as read-though (rt)-circRNAs which are produced from exons of two different neighboring genes. Specifically, rt-circRNAs from SCAF8 and TIAM2 were observed to be dysregulated in DCM and these rt-circRNAs have the potential to sponge multiple heart disease-related miRNAs. CONCLUSIONS In summary, this study provides a valuable resource for exploring the function of circRNAs in human heart disease and establishes a functional paradigm for identifying novel circRNAs in other tissues.
Collapse
Affiliation(s)
- Kunzhe Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Xiangqin He
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Huabo Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA.,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - David J R Fulton
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA.,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
9
|
Deutsch JL, Heath JL. MLLT10 in benign and malignant hematopoiesis. Exp Hematol 2020; 87:1-12. [PMID: 32569758 DOI: 10.1016/j.exphem.2020.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 01/01/2023]
Abstract
Non-random chromosomal translocations involving the putative transcription factor Mixed Lineage Leukemia Translocated to 10 (MLLT10, also known as AF10) are commonly observed in both acute myeloid and lymphoid leukemias and are indicative of a poor prognosis. Despite the well-described actions of oncogenic MLLT10 fusion proteins, the role of wild-type MLLT10 in hematopoiesis is not well characterized. The protein structure and several interacting partners have been described and provide indications as to the potential functions of MLLT10. This review examines these aspects of MLLT10, contextualizing its function in benign and malignant hematopoiesis.
Collapse
Affiliation(s)
- Jamie L Deutsch
- Department of Pediatrics, University of Vermont, Burlington, VT
| | - Jessica L Heath
- Department of Pediatrics, University of Vermont, Burlington, VT; Department of Biochemistry, University of Vermont, Burlington, VT 05405; University of Vermont Cancer Center, Burlington, VT.
| |
Collapse
|
10
|
Burdine RD, Preston CC, Leonard RJ, Bradley TA, Faustino RS. Nucleoporins in cardiovascular disease. J Mol Cell Cardiol 2020; 141:43-52. [PMID: 32209327 DOI: 10.1016/j.yjmcc.2020.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 01/01/2023]
Abstract
Cardiovascular disease is a pressing health problem with significant global health, societal, and financial burdens. Understanding the molecular basis of polygenic cardiac pathology is thus essential to devising novel approaches for management and treatment. Recent identification of uncharacterized regulatory functions for a class of nuclear envelope proteins called nucleoporins offers the opportunity to understand novel putative mechanisms of cardiac disease development and progression. Consistent reports of nucleoporin deregulation associated with ischemic and dilated cardiomyopathies, arrhythmias and valvular disorders suggests that nucleoporin impairment may be a significant but understudied variable in cardiopathologic disorders. This review discusses and converges existing literature regarding nuclear pore complex proteins and their association with cardiac pathologies, and proposes a role for nucleoporins as facilitators of cardiac disease.
Collapse
Affiliation(s)
- Ryan D Burdine
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America; School of Health Sciences, University of South Dakota, 414 E Clark St, Vermillion, SD 57069, United States of America
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Riley J Leonard
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Tyler A Bradley
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America; Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22(nd) Street, Sioux Falls, SD 57105, United States of America.
| |
Collapse
|
11
|
Modulation of ADAR mRNA expression in patients with congenital heart defects. PLoS One 2019; 14:e0200968. [PMID: 31039163 PMCID: PMC6490900 DOI: 10.1371/journal.pone.0200968] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 04/05/2019] [Indexed: 12/26/2022] Open
Abstract
Adenosine (A) to inosine (I) RNA editing is a hydrolytic deamination reaction catalyzed by the adenosine deaminase (ADAR) enzyme acting on double-stranded RNA. This posttranscriptional process diversifies a plethora of transcripts, including coding and noncoding RNAs. Interestingly, few studies have been carried out to determine the role of RNA editing in vascular disease. The aim of this study was to determine the potential role of ADARs in congenital heart disease. Strong downregulation of ADAR2 and increase in ADAR1 expression was observed in blood samples from congenital heart disease (CHD) patients. The decrease in expression of ADAR2 was in line with its downregulation in ventricular tissues of dilated cardiomyopathy patients. To further decipher the plausible regulatory pathway of ADAR2 with respect to heart physiology, miRNA profiling of ADAR2 was performed on tissues from ADAR2-/- mouse hearts. Downregulation of miRNAs (miR-29b, miR-405, and miR-19) associated with cardiomyopathy and cardiac fibrosis was observed. Moreover, the upregulation of miR-29b targets COL1A2 and IGF1, indicated that ADAR2 might be involved in cardiac myopathy. The ADAR2 target vascular development associated protein-coding gene filamin B (FLNB) was selected. The editing levels of FLNB were dramatically reduced in ADAR2-/- mice; however, no observable changes in FLNB expression were noted in ADAR2-/- mice compared to wild-type mice. This study proposes that sufficient ADAR2 enzyme activity might play a vital role in preventing cardiovascular defects.
Collapse
|
12
|
Sweet ME, Cocciolo A, Slavov D, Jones KL, Sweet JR, Graw SL, Reece TB, Ambardekar AV, Bristow MR, Mestroni L, Taylor MRG. Transcriptome analysis of human heart failure reveals dysregulated cell adhesion in dilated cardiomyopathy and activated immune pathways in ischemic heart failure. BMC Genomics 2018; 19:812. [PMID: 30419824 PMCID: PMC6233272 DOI: 10.1186/s12864-018-5213-9] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/31/2018] [Indexed: 01/17/2023] Open
Abstract
Background Current heart failure (HF) treatment is based on targeting symptoms and left ventricle dysfunction severity, relying on a common HF pathway paradigm to justify common treatments for HF patients. This common strategy may belie an incomplete understanding of heterogeneous underlying mechanisms and could be a barrier to more precise treatments. We hypothesized we could use RNA-sequencing (RNA-seq) in human heart tissue to delineate HF etiology-specific gene expression signatures. Results RNA-seq from 64 human left ventricular samples: 37 dilated (DCM), 13 ischemic (ICM), and 14 non-failing (NF). Using a multi-analytic approach including covariate adjustment for age and sex, differentially expressed genes (DEGs) were identified characterizing HF and disease-specific expression. Pathway analysis investigated enrichment for biologically relevant pathways and functions. DCM vs NF and ICM vs NF had shared HF-DEGs that were enriched for the fetal gene program and mitochondrial dysfunction. DCM-specific DEGs were enriched for cell-cell and cell-matrix adhesion pathways. ICM-specific DEGs were enriched for cytoskeletal and immune pathway activation. Using the ICM and DCM DEG signatures from our data we were able to correctly classify the phenotypes of 24/31 ICM and 32/36 DCM samples from publicly available replication datasets. Conclusions Our results demonstrate the commonality of mitochondrial dysfunction in end-stage HF but more importantly reveal key etiology-specific signatures. Dysfunctional cell-cell and cell-matrix adhesion signatures typified DCM whereas signals related to immune and fibrotic responses were seen in ICM. These findings suggest that transcriptome signatures may distinguish end-stage heart failure, shedding light on underlying biological differences between ICM and DCM. Electronic supplementary material The online version of this article (10.1186/s12864-018-5213-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mary E Sweet
- Human Medical Genetics and Genomics, University of Colorado, Aurora, CO, USA
| | - Andrea Cocciolo
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - Dobromir Slavov
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado, Aurora, CO, USA
| | - Joseph R Sweet
- Department of Statistics, E. & J. Gallo, Modesto, CA, USA
| | - Sharon L Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - T Brett Reece
- Department of Cardiothoracic Surgery, University of Colorado Hospital, Aurora, CO, USA
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Michael R Bristow
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Luisa Mestroni
- Human Medical Genetics and Genomics, University of Colorado, Aurora, CO, USA.,Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - Matthew R G Taylor
- Human Medical Genetics and Genomics, University of Colorado, Aurora, CO, USA. .,Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
13
|
Kwon HK, Jeong H, Hwang D, Park ZY. Comparative proteomic analysis of mouse models of pathological and physiological cardiac hypertrophy, with selection of biomarkers of pathological hypertrophy by integrative Proteogenomics. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2018; 1866:S1570-9639(18)30118-3. [PMID: 30048702 DOI: 10.1016/j.bbapap.2018.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/13/2018] [Accepted: 07/20/2018] [Indexed: 12/21/2022]
Abstract
To determine fundamental characteristics of pathological cardiac hypertrophy, protein expression profiles in two widely accepted models of cardiac hypertrophy (swimming-trained mouse for physiological hypertrophy and pressure-overload-induced mouse for pathological hypertrophy) were compared using a label-free quantitative proteomics approach. Among 3955 proteins (19,235 peptides, false-discovery rate < 0.01) identified in these models, 486 were differentially expressed with a log2 fold difference ≥ 0.58, or were detected in only one hypertrophy model (each protein from 4 technical replicates, p < .05). Analysis of gene ontology biological processes and KEGG pathways identified cellular processes enriched in one or both hypertrophy models. Processes unique to pathological hypertrophy were compared with processes previously identified in cardiac-hypertrophy models. Individual proteins with differential expression in processes unique to pathological hypertrophy were further confirmed using the results of previous targeted functional analysis studies. Using a proteogenomic approach combining transcriptomic and proteomic analyses, similar patterns of differential expression were observed for 23 proteins and corresponding genes associated with pathological hypertrophy. A total of 11 proteins were selected as early-stage pathological-hypertrophy biomarker candidates, and the results of western blotting for five of these proteins in independent samples confirmed the patterns of differential expression in mouse models of pathological and physiological cardiac hypertrophy.
Collapse
Affiliation(s)
- Hye Kyeong Kwon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyobin Jeong
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea; Center for Plant Aging Research, Institute for Basic Science, DGIST, Daegu 42988, Republic of Korea; School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Daehee Hwang
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea; Center for Plant Aging Research, Institute for Basic Science, DGIST, Daegu 42988, Republic of Korea
| | - Zee-Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
14
|
Isaza C, Rosas JF, Lorenzo E, Marrero A, Ortiz C, Ortiz MR, Perez L, Cabrera‐Ríos M. Biological signaling pathways and potential mathematical network representations: biological discovery through optimization. Cancer Med 2018; 7:1875-1895. [PMID: 29635835 PMCID: PMC5943441 DOI: 10.1002/cam4.1301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 01/04/2023] Open
Abstract
Establishing the role that different genes play in the development of cancer is a daunting task. A step toward this end is the detection of genes that are important in the illness from high-throughput biological experiments. Furthermore, it is safe to say that it is highly unlikely that these show expression changes independently, even with a list of potentially important genes. A biological signaling pathway is a more plausible underlying mechanism as favored in the literature. This work attempts to build a mathematical network problem through the analysis of microarray experiments. A preselection of genes is carried out with a multiple criteria optimization framework previously published by our research group . Afterward, application of the Traveling Salesperson Problem and Minimum Spanning Tree network optimization models are proposed to identify potential signaling pathways via the most correlated path among the genes of interest. Biological evidencing is provided to assess the effectiveness of the proposed methods. The capability of our analysis strategy is also demonstrated through the undertaking of meta-analysis studies. Three important aspects are novel in this work: (1) our joint analyses of different groups of lung cancer states reveal new correlations, biologically evidenced, and previously undocumented; (2) computation of the correlation coefficients from expression differences leads to an effective use of network optimization methods; and (3) the methods yield mathematically optimal correlation structures: no other configuration is better correlated using the available information.
Collapse
Affiliation(s)
- Clara Isaza
- Public Health ProgramPonce Health Sciences UniversityPonce00732‐7004Puerto Rico
| | - Juan F. Rosas
- Industrial Engineering DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Enery Lorenzo
- Industrial Engineering DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Arlette Marrero
- Biology DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Cristina Ortiz
- Biology DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Michael R. Ortiz
- Biology DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Lynn Perez
- Biology DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| | - Mauricio Cabrera‐Ríos
- Industrial Engineering DepartmentThe Applied Optimization GroupUniversity of Puerto Rico‐MayagüezMayagüez00681‐9043Puerto Rico
| |
Collapse
|
15
|
A transcriptomic study of myogenic differentiation under the overexpression of PPARγ by RNA-Seq. Sci Rep 2017; 7:15308. [PMID: 29127356 PMCID: PMC5681552 DOI: 10.1038/s41598-017-14275-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022] Open
Abstract
To study the cellular and molecular function of peroxisome proliferator-activated receptor γ (PPARγ) in skeletal muscle differentiation, we have generated inducible gain-of-function to overexpress PPARγ in C2C12 myoblasts. In order to identify PPARγ targets, RNA sequencing (RNA-seq) was used to evaluate and quantify the transcriptomes and expression patterns during myogenic differentiation under the overexpression of PPARγ. The formation of myotubes and the expression of muscle-specific myogenic genes such as MyoD and MyoG may be inhibited by PPARγ overexpression. Multiple genes and pathways were significantly involved in this process, including 11 genes such as Fndc9 and Slc14a1 with fundamental change of regulation modes, 9 genes of which were validated by the data of qRT-PCR. Our studies demonstrate that PPARγ would play critical roles on myoblasts differentiation, mediating crosstalk among several pathways and transcription factors. Our data is available in the Gene Expression Omnibus (GEO) database with the accession number as GSE99399.
Collapse
|
16
|
Molecular Characterization of Pediatric Restrictive Cardiomyopathy from Integrative Genomics. Sci Rep 2017; 7:39276. [PMID: 28098235 PMCID: PMC5241776 DOI: 10.1038/srep39276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pediatric restrictive cardiomyopathy (RCM) is a genetically heterogeneous heart disease with limited therapeutic options. RCM cases are largely idiopathic; however, even within families with a known genetic cause for cardiomyopathy, there is striking variability in disease severity. Although accumulating evidence implicates both gene expression and alternative splicing in development of dilated cardiomyopathy (DCM), there have been no detailed molecular characterizations of underlying pathways dysregulated in RCM. RNA-Seq on a cohort of pediatric RCM patients compared to other forms of adult cardiomyopathy and controls identified transcriptional differences highly common to the cardiomyopathies, as well as those unique to RCM. Transcripts selectively induced in RCM include many known and novel G-protein coupled receptors linked to calcium handling and contractile regulation. In-depth comparisons of alternative splicing revealed splicing events shared among cardiomyopathy subtypes, as well as those linked solely to RCM. Genes identified with altered alternative splicing implicate RBM20, a DCM splicing factor, as a potential mediator of alternative splicing in RCM. We present the first comprehensive report on molecular pathways dysregulated in pediatric RCM including unique/shared pathways identified compared to other cardiomyopathy subtypes and demonstrate that disruption of alternative splicing patterns in pediatric RCM occurs in the inverse direction as DCM.
Collapse
|
17
|
Transcriptional profiling of rat skeletal muscle hypertrophy under restriction of blood flow. Gene 2016; 594:229-237. [PMID: 27613141 DOI: 10.1016/j.gene.2016.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 08/16/2016] [Accepted: 09/05/2016] [Indexed: 01/03/2023]
Abstract
Blood flow restriction (BFR) under low-intensity resistance training (LIRT) can produce similar effects upon muscles to that of high-intensity resistance training (HIRT) while overcoming many of the restrictions to HIRT that occurs in a clinical setting. However, the potential molecular mechanisms of BFR induced muscle hypertrophy remain largely unknown. Here, using a BFR rat model, we aim to better elucidate the mechanisms regulating muscle hypertrophy as induced by BFR and reveal possible clinical therapeutic targets for atrophy cases. We performed genome wide screening with microarray analysis to identify unique differentially expressed genes during rat muscle hypertrophy. We then successfully separated the differentially expressed genes from BRF treated soleus samples by comparing the Affymetrix rat Genome U34 2.0 array with the control. Using qRT-PCR and immunohistochemistry (IHC) we also analyzed other related differentially expressed genes. Results suggested that muscle hypertrophy induced by BFR is essentially regulated by the rate of protein turnover. Specifically, PI3K/AKT and MAPK pathways act as positive regulators in controlling protein synthesis where ubiquitin-proteasome acts as a negative regulator. This represents the first general genome wide level investigation of the gene expression profile in the rat soleus after BFR treatment. This may aid our understanding of the molecular mechanisms regulating and controlling muscle hypertrophy and provide support to the BFR strategies aiming to prevent muscle atrophy in a clinical setting.
Collapse
|
18
|
Zhang H, Yu Z, He J, Hua B, Zhang G. Identification of the molecular mechanisms underlying dilated cardiomyopathy via bioinformatic analysis of gene expression profiles. Exp Ther Med 2016; 13:273-279. [PMID: 28123501 PMCID: PMC5245080 DOI: 10.3892/etm.2016.3953] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/21/2016] [Indexed: 12/29/2022] Open
Abstract
In the present study, gene expression profiles of patients with dilated cardiomyopathy (DCM) were re-analyzed with bioinformatics tools to investigate the molecular mechanisms underlying DCM. Gene expression dataset GSE3585 was downloaded from Gene Expression Omnibus, which included seven heart biopsy samples obtained from patients with DCM and five healthy controls. Differential analysis was performed using a Limma package in R to screen for differentially expressed genes (DEGs). Functional enrichment analysis was subsequently conducted for DEGs using the Database for Annotation, Visualization and Integration Discovery. A protein-protein interaction (PPI) network was constructed using information from Search Tool for the Retrieval of Interacting Genes software. A total of 89 DEGs were identified in the patients with DCM, including 67 upregulated and 22 downregulated genes. Functional enrichment analysis demonstrated that the downregulated genes predominantly encoded chromosomal proteins and transport-related proteins, which were significantly associated with the biological processes of ‘nucleosome assembly’, ‘chromatin assembly’, ‘protein-DNA complex assembly’, ‘nucleosome organization’ and ‘DNA packaging’ (H1 histone family member 0, histone cluster 1 H1c, histone cluster 1 H2bd and H2A histone family member Z). The upregulated genes detected in the present study encoded secreted proteins or phosphotransferase, which were associated with biological processes including ‘cell adhesion’ [connective tissue growth factor (CTGF)], ‘skeletal system development’ [CTGF and insulin-like growth factor binding protein 3 (IGFBP3)], ‘muscle organ development’ (SMAD7) and ‘regulation of cell migration’ [SMAD7, IGFBP3 and insulin receptor (INSR)]. Notably, signal transducer and activator of transcription 3, SMAD7, INSR, CTGF, exportin 1, IGFBP3 and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha were hub nodes with the higher degree in the PPI network. Therefore, the results of the present study suggested that DEGs may alter the biological processes of ‘nucleosome formation’, ‘cell adhesion’, ‘skeletal system development’, ‘muscle organ development’ and ‘regulation of cell migration’ in the development of DCM.
Collapse
Affiliation(s)
- Hu Zhang
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhuo Yu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jianchao He
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Baotong Hua
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Guiming Zhang
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
19
|
Nanni S, Re A, Ripoli C, Gowran A, Nigro P, D’Amario D, Amodeo A, Crea F, Grassi C, Pontecorvi A, Farsetti A, Colussi C. The nuclear pore protein Nup153 associates with chromatin and regulates cardiac gene expression in dystrophicmdxhearts. Cardiovasc Res 2016; 112:555-567. [DOI: 10.1093/cvr/cvw204] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 08/13/2016] [Indexed: 11/14/2022] Open
|
20
|
DeAguero JL, McKown EN, Zhang L, Keirsey J, Fischer EG, Samedi VG, Canan BD, Kilic A, Janssen PML, Delfín DA. Altered protein levels in the isolated extracellular matrix of failing human hearts with dilated cardiomyopathy. Cardiovasc Pathol 2016; 26:12-20. [PMID: 27837684 DOI: 10.1016/j.carpath.2016.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/23/2016] [Accepted: 10/10/2016] [Indexed: 01/09/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is associated with extensive pathological cardiac remodeling and involves numerous changes in the protein expression profile of the extracellular matrix of the heart. We obtained seven human, end-stage, failing hearts with DCM (DCM-failing) and nine human, nonfailing donor hearts and compared their extracellular matrix protein profiles. We first showed that the DCM-failing hearts had indeed undergone extensive remodeling of the left ventricle myocardium relative to nonfailing hearts. We then isolated the extracellular matrix from a subset of these hearts and performed a proteomic analysis on the isolated matrices. We found that the levels of 26 structural proteins were altered in the DCM-failing isolated cardiac extracellular matrix compared to nonfailing isolated cardiac extracellular matrix. Overall, most of the extracellular matrix proteins showed reduced levels in the DCM-failing hearts, while all of the contractile proteins showed increased levels. There was a mixture of increased and decreased levels of cytoskeletal and nuclear transport proteins. Using immunoprobing, we verified that collagen IV (α2 and α6 isoforms), zyxin, and myomesin protein levels were reduced in the DCM-failing hearts. We expect that these data will add to the understanding of the pathology associated with heart failure with DCM.
Collapse
Affiliation(s)
- Joshua L DeAguero
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Elizabeth N McKown
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Liwen Zhang
- The Ohio State University Mass Spectrometry and Proteomics Facility, Campus Chemical Instrument Center, 460 W. 12th Ave., Room 250 Biomedical Research Tower, Columbus, OH 43210, USA.
| | - Jeremy Keirsey
- The Ohio State University Mass Spectrometry and Proteomics Facility, Campus Chemical Instrument Center, 460 W. 12th Ave., Room 250 Biomedical Research Tower, Columbus, OH 43210, USA.
| | - Edgar G Fischer
- The University of New Mexico School of Medicine, Department of Pathology, MSC08 4640, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Von G Samedi
- The University of New Mexico School of Medicine, Department of Pathology, MSC08 4640, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Benjamin D Canan
- The Ohio State University College of Medicine, Department of Physiology and Cell Biology and the Davis Heart Lung Research Institute, 200 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210.
| | - Ahmet Kilic
- The Ohio State University College of Medicine, Department of Surgery and the Davis Heart Lung Research Institute, Richard M. Ross Heart Hospital, 452 West 10th Ave., Columbus, OH 43210.
| | - Paul M L Janssen
- The Ohio State University College of Medicine, Department of Physiology and Cell Biology and the Davis Heart Lung Research Institute, 200 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210.
| | - Dawn A Delfín
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
21
|
Cao S, Zhou Q, Chen JL, Hu B, Guo RQ. The differences in left atrial function between ischemic and idiopathic dilated cardiomyopathy patients: A two-dimensional speckle tracking imaging study. JOURNAL OF CLINICAL ULTRASOUND : JCU 2016; 44:437-445. [PMID: 26990443 DOI: 10.1002/jcu.22352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 02/01/2016] [Accepted: 02/08/2016] [Indexed: 06/05/2023]
Abstract
PURPOSE To evaluate left atrial (LA) function in patients with ischemic (ICM) or idiopathic dilated (DCM) cardiomyopathy via two-dimensional speckle-tracking imaging. METHODS We measured the LA maximum volume, minimum volume, and volume before the atrial systole, and calculated total emptying volume, expansion index, active emptying volume, and fraction. We measured strain and strain rate during systole and late diastole using two-dimensional speckle-tracking imaging, and analyzed correlations between variables. RESULTS We found no significant differences in LA size, left ventricle (LV) end-diastole diameter, LV ejection fraction (EF), E/A, E/e', deceleration time of the E wave, and effective mitral regurgitant orifice area between the DCM and the ICM group. However, the LA expansion index, active EF, systolic and late diastolic strain, and strain rate were lower in the ICM group (p < 0.05). The expansion index and active EF were positively correlated with the systolic strain rate and the absolute value of the late diastolic strain rate, respectively. CONCLUSIONS LA basic echocardiographic variables did not reflect the differences between ICM and DCM patients, but the systolic and late diastolic strain, as well as the strain rate, were lower in DCM patients. Two-dimensional speckle-tracking imaging is a promising method to differentiate these patients. © 2016 Wiley Periodicals, Inc. J Clin Ultrasound 44:437-445, 2016.
Collapse
Affiliation(s)
- Sheng Cao
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Zhou
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jin-Ling Chen
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bo Hu
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui-Qiang Guo
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
22
|
Karyopherin Alpha 2 Promotes the Inflammatory Response in Rat Pancreatic Acinar Cells Via Facilitating NF-κB Activation. Dig Dis Sci 2016; 61:747-57. [PMID: 26526450 DOI: 10.1007/s10620-015-3948-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 10/24/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Activation of the transcription factor NF-κB and expression of pro-inflammatory mediators have been considered as major events of acute pancreatitis (AP). Karyopherin alpha 2 (KPNA2), a member of the importin α family, reportedly modulates p65 subcellular localization. AIM This study aimed to investigate the expression and possible functions of KPNA2 in the AP cell and animal model, focusing on its association with NF-κB activation. METHODS An AP cell model was established with the cerulein-stimulated AR42J and isolated rat pancreatic acinar cells. The AP rat model was induced by the intraperitoneal injection of cerulein. The secretion of TNF-α, IL-6, and LDH was detected by ELISA kits and the production of NO using nitric oxide kit. Expression of KPNA2 was measured by RT-PCR and Western blot. Expression levels of IKKα, phosphorylation of p65, and total p65 were detected by Western blot. Co-localization of KPNA2 with p65 was observed by immunofluorescence assay. To determine the biological functions of KPNA2 in cerulein-induced inflammatory response, RNA interference was employed to knockdown KPNA2 expression in AR42J and isolated pancreatic acini cells. RESULTS Cerulein stimulated KPNA2 expression and IL-6, TNF-α, NO, and LDH production in rat pancreatic acinar cells. Cerulein triggered the phosphorylation and nuclear translocation of NF-κB p65 subunit, indicating the NF-κB activation. The co-localization and nuclear accumulation of KPNA2 and p65 were detected in cerulein-treated cells. Knocking down KPNA2 hindered cerulein-induced nuclear transportation of p65 and alleviated the subsequent inflammatory response in rat pancreatic acinar cells. Additionally, KPNA2 expression was significantly up-regulated in cerulein-induced AP rat model. CONCLUSIONS KPNA2-facilitated p65 nuclear translocation promotes NF-κB activation and inflammation in acute pancreatitis.
Collapse
|
23
|
Lee YS, Kim JK, Ryu SW, Bae SJ, Kwon K, Noh YH, Kim SY. Integrative meta-analysis of multiple gene expression profiles in acquired gemcitabine-resistant cancer cell lines to identify novel therapeutic biomarkers. Asian Pac J Cancer Prev 2016; 16:2793-800. [PMID: 25854364 DOI: 10.7314/apjcp.2015.16.7.2793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In molecular-targeted cancer therapy, acquired resistance to gemcitabine is a major clinical problem that reduces its effectiveness, resulting in recurrence and metastasis of cancers. In spite of great efforts to reveal the overall mechanism of acquired gemcitabine resistance, no definitive genetic factors have been identified that are absolutely responsible for the resistance process. Therefore, we performed a cross-platform meta-analysis of three publically available microarray datasets for cancer cell lines with acquired gemcitabine resistance, using the R-based RankProd algorithm, and were able to identify a total of 158 differentially expressed genes (DEGs; 76 up- and 82 down-regulated) that are potentially involved in acquired resistance to gemcitabine. Indeed, the top 20 up- and down-regulated DEGs are largely associated with a common process of carcinogenesis in many cells. For the top 50 up- and down-regulated DEGs, we conducted integrated analyses of a gene regulatory network, a gene co-expression network, and a protein-protein interaction network. The identified DEGs were functionally enriched via Gene Ontology hierarchy and Kyoto Encyclopedia of Genes and Genomes pathway analyses. By systemic combinational analysis of the three molecular networks, we could condense the total number of DEGs to final seven genes. Notably, GJA1, LEF1, and CCND2 were contained within the lists of the top 20 up- or down-regulated DEGs. Our study represents a comprehensive overview of the gene expression patterns associated with acquired gemcitabine resistance and theoretical support for further clinical therapeutic studies.
Collapse
Affiliation(s)
- Young Seok Lee
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, Republic of Korea E-mail :
| | | | | | | | | | | | | |
Collapse
|
24
|
Basheer WA, Harris BS, Mentrup HL, Abreha M, Thames EL, Lea JB, Swing DA, Copeland NG, Jenkins NA, Price RL, Matesic LE. Cardiomyocyte-specific overexpression of the ubiquitin ligase Wwp1 contributes to reduction in Connexin 43 and arrhythmogenesis. J Mol Cell Cardiol 2015; 88:1-13. [PMID: 26386426 DOI: 10.1016/j.yjmcc.2015.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/30/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022]
Abstract
Gap junctions (GJ) are intercellular channels composed of connexin subunits that play a critical role in a diverse number of cellular processes in all tissue types. In the heart, GJs mediate electrical coupling between cardiomyocytes and display mislocalization and/or downregulation in cardiac disease (a process known as GJ remodeling), producing an arrhythmogenic substrate. The main constituent of GJs in the ventricular myocardium is Connexin 43 (Cx43), an integral membrane protein that is rapidly turned over and shows decreased expression or function with age. We hypothesized that Wwp1, an ubiquitin ligase whose expression in known to increase in aging-related pathologies, may regulate Cx43 in vivo by targeting it for ubiquitylation and degradation and yield tissue-specific Cx43 loss of function phenotypes. When Wwp1 was globally overexpressed in mice under the control of a β-actin promoter, the highest induction of Wwp1 expression was observed in the heart which was associated with a 90% reduction in cardiac Cx43 protein levels, left ventricular hypertrophy (LVH), and the development of lethal ventricular arrhythmias around 8weeks of age. This phenotype was completely penetrant in two independent founder lines. Cardiomyocyte-specific overexpression of Wwp1 confirmed that this phenotype was cell autonomous and delineated Cx43-dependent and -independent roles for Wwp1 in arrhythmogenesis and LVH, respectively. Using a cell-based system, it was determined that Wwp1 co-immunoprecipitates with and ubiquitylates Cx43, causing a decrease in the steady state levels of Cx43 protein. These findings offer new mechanistic insights into the regulation of Cx43 which may be exploitable in various gap junctionopathies.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Connexin 43/genetics
- Connexin 43/metabolism
- Disease Models, Animal
- Female
- Gap Junctions/metabolism
- Gap Junctions/pathology
- Gene Expression Regulation
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Male
- Mice
- Mice, Transgenic
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenotype
- Promoter Regions, Genetic
- Proteasome Endopeptidase Complex/metabolism
- Protein Stability
- Proteolysis
- Signal Transduction
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitination
Collapse
Affiliation(s)
- Wassim A Basheer
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Brett S Harris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Heather L Mentrup
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Measho Abreha
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Elizabeth L Thames
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Jessica B Lea
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Deborah A Swing
- Mouse Cancer Genetics Program, The National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Neal G Copeland
- Mouse Cancer Genetics Program, The National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Nancy A Jenkins
- Mouse Cancer Genetics Program, The National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Robert L Price
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Lydia E Matesic
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
25
|
Lee YS, Hwang SG, Kim JK, Park TH, Kim YR, Myeong HS, Choi JD, Kwon K, Jang CS, Ro YT, Noh YH, Kim SY. Identification of novel therapeutic target genes in acquired lapatinib-resistant breast cancer by integrative meta-analysis. Tumour Biol 2015; 37:2285-97. [PMID: 26361955 DOI: 10.1007/s13277-015-4033-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/02/2015] [Indexed: 12/11/2022] Open
Abstract
Acquired resistance to lapatinib is a highly problematic clinical barrier that has to be overcome for a successful cancer treatment. Despite efforts to determine the mechanisms underlying acquired lapatinib resistance (ALR), no definitive genetic factors have been reported to be solely responsible for the acquired resistance in breast cancer. Therefore, we performed a cross-platform meta-analysis of three publically available microarray datasets related to breast cancer with ALR, using the R-based RankProd package. From the meta-analysis, we were able to identify a total of 990 differentially expressed genes (DEGs, 406 upregulated, 584 downregulated) that are potentially associated with ALR. Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the DEGs showed that "response to organic substance" and "p53 signaling pathway" may be largely involved in ALR process. Of these, many of the top 50 upregulated and downregulated DEGs were found in oncogenesis of various tumors and cancers. For the top 50 DEGs, we constructed the gene coexpression and protein-protein interaction networks from a huge database of well-known molecular interactions. By integrative analysis of two systemic networks, we condensed the total number of DEGs to six common genes (LGALS1, PRSS23, PTRF, FHL2, TOB1, and SOCS2). Furthermore, these genes were confirmed in functional module eigens obtained from the weighted gene correlation network analysis of total DEGs in the microarray datasets ("GSE16179" and "GSE52707"). Our integrative meta-analysis could provide a comprehensive perspective into complex mechanisms underlying ALR in breast cancer and a theoretical support for further chemotherapeutic studies.
Collapse
Affiliation(s)
- Young Seok Lee
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sun Goo Hwang
- Plant Genomics Laboratory, Department of Applied Plant Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Jin Ki Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Tae Hwan Park
- Department of Plastic and Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Young Rae Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Ho Sung Myeong
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jong Duck Choi
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Kang Kwon
- School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Cheol Seong Jang
- Plant Genomics Laboratory, Department of Applied Plant Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Young Tae Ro
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Yun Hee Noh
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea.
| |
Collapse
|
26
|
|