1
|
Kobayashi G, Ito R, Taga M, Koyama K, Yano S, Endo T, Kai T, Yamamoto T, Hiratsuka T, Tsuruyama T. Proteomic profiling of FFPE specimens: Discovery of HNRNPA2/B1 and STT3B as biomarkers for determining formalin fixation durations. J Proteomics 2024; 301:105196. [PMID: 38723849 DOI: 10.1016/j.jprot.2024.105196] [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: 03/26/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
Recent advancements in proteomics technologies using formalin-fixed paraffin-embedded (FFPE) samples have significantly advanced biomarker discovery. Yet, the effects of varying sample preparation protocols on proteomic analyses remain poorly understood. We analyzed mouse liver FFPE samples that varied in fixatives, fixation duration, and storage temperature using LC/MS. We found that variations in fixation duration significantly affected the abundance of specific proteins, showing that HNRNPA2/B1 demonstrated a significant decrease in abundance in samples fixed for long periods, whereas STT3B exhibited a significant increase in abundance in samples fixed for long durations. These findings were supported by immunohistochemical analysis across liver, spleen, and lung tissues, demonstrating a significant decrease in the nuclear staining of HNRNPA2/B1 in long-duration acid formalin(AF)-fixed FFPE samples, and an increase in cytoplasmic staining of STT3B in long-duration neutral buffered formalin-fixed liver and lung tissues and granular staining in all long-duration AF-fixed FFPE tissue types. Similar trends were observed in the long-duration fixed HeLa cells. These results demonstrate that fixation duration critically affects the proteomic integrity of FFPE samples, emphasizing the urgent need for standardized fixation protocols to ensure consistent and reliable proteomic data. SIGNIFICANCE: The quality of FFPE samples is primarily influenced by the fixation and storage conditions. However, previous studies have mainly focused on their impact on nucleic acids and the extent to which different fixation conditions affect changes in proteins has not been evaluated. In addition, to our knowledge, proteomic research focusing on differences in formalin fixation conditions has not yet been conducted. Here, we analyzed FFPE samples with different formalin fixation and storage conditions using LC/MS and evaluated the impact of different fixation conditions on protein variations. Our study unequivocally established formalin fixation duration as a critical determinant of protein variation in FFPE specimens and successfully identified HNRNPA2/B1 and STT3B as potential biomarkers for predicting formalin fixation duration for the first time. The study findings open new avenues for quality assessment in biomedical research and diagnostics.
Collapse
Affiliation(s)
- Go Kobayashi
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Reiko Ito
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan; Department of Functions of Biological-defense Genome, Hiroshima University Graduate School, Hiroshima, Japan
| | - Masataka Taga
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Kazuaki Koyama
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Shiho Yano
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Tatsuya Endo
- Department of Physics, Graduate school of Science, Tohoku University, Miyagi, Japan
| | | | - Takushi Yamamoto
- Kyoto Applications Development Center, Analytical and Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Takuya Hiratsuka
- Department of Drug Discovery Medicine, Pathology Division, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan; Department of Functions of Biological-defense Genome, Hiroshima University Graduate School, Hiroshima, Japan; Department of Physics, Graduate school of Science, Tohoku University, Miyagi, Japan; Department of Drug Discovery Medicine, Pathology Division, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
2
|
Hiratsuka T, Ito S, Sakai R, Yokose T, Endo T, Daigo Y, Miyagi Y, Tsuruyama T. Proteome analysis of CD5-positive diffuse large B cell lymphoma FFPE tissue reveals downregulation of DDX3X, DNAJB1, and B cell receptor signaling pathway proteins including BTK and Immunoglobulins. Clin Proteomics 2023; 20:36. [PMID: 37705009 PMCID: PMC10498596 DOI: 10.1186/s12014-023-09422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/25/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND The molecular pathology of diffuse large B cell lymphoma (DLBCL) has been extensively studied. Among DLBCL subtypes, the prognosis of CD5-positive DLBCL is worse than that of CD5-negative DLBCL, considering the central nervous system relapse and poor response to R-CHOP therapy. However, the molecular mechanisms underlying the tumorigenesis and progression of CD5-positive DLBCL remain unknown. METHODS To identify molecular markers that can be targeted for treating DLBCL, a proteomic study was performed using liquid chromatography-mass spectrometry with chemically pretreated formalin-fixed paraffin-embedded specimens from CD5-positive (n = 5) and CD5-negative DLBCL patients (n = 6). RESULTS Twenty-one proteins showed significant downregulation in CD5-positive DLBCL compared to CD5-negative DLBCL. Principal component analysis of protein expression profiling in CD5-positive and CD5-negative DLBCL revealed that DNAJB1, DDX3X, and BTK, which is one of the B cell phenotypic proteins, were the most significantly downregulated proteins and served as biomarkers that distinguished both groups. Additionally, a set of immunoglobulins, including IgG4, exhibited significant downregulation. Immunohistochemistry analysis for BTK demonstrated reduced staining in CD5-positive DLBCL compared to CD5-negative DLBCL. CONCLUSIONS In conclusion, DNAJB1 and DDX3X, BTK, and a set of immunoglobulins are promising biomarkers. Probably, the suppression of BCR signaling is the unique phenotype of CD5-positive DLBCL. This formalin-fixed paraffin-embedded (FFPE)-based profiling may help to develop novel therapeutic molecularly targeted drugs for treating DLBCL.
Collapse
Affiliation(s)
- Takuya Hiratsuka
- Department of Drug Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rika Sakai
- Department of Oncology, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tatsuya Endo
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan
| | - Yataro Daigo
- Department of Medical Oncology, Cancer Center, and Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Physics, Graduate School of Science, Tohoku University, Sendai, Japan.
- Tazuke-Kofukai Medical Institute Kitano Hospital, Ogimachi, Osaka, Japan.
| |
Collapse
|
3
|
Moldovan R, Ichim VA, Beliș V. Recent perspectives on the early expression immunohistochemical markers in post-mortem recognition of myocardial infarction. Leg Med (Tokyo) 2023; 64:102293. [PMID: 37392575 DOI: 10.1016/j.legalmed.2023.102293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
Acute Myocardial Infarction (AMI) refers to the death of heart tissue in the absence ofperfusion. It is one of the top causes of death globally, particularly in middle andhigher-age groups. However, for the pathologist, the post-mortem macroscopic andmicroscopic diagnosis of early AMI remains challenging. In the early acute stage ofAMI, no microscopic visible signs of tissue alterations like necrosis and neutrophilinfiltration can be seen. In such a scenario, immunohistochemistry (IHC) accounts forthe most suitable and safest alternative to study early diagnostic cases by selectivelydetecting changes in the cell population. This systematic review focuses on themultiple causes/changes that lead to the privation of blood flow as well as tissuechanges induced by the absence of perfusion.We performed a systematic review of the last 10-15 years' publications that focused ondetecting immunohistochemical changes that appear in the cell population in case ofacute myocardial infarction. We found around 160 articles on AMI, which we narroweddown to 50 with the use of specific filters such as: "Acute Myocardial Infarction," "Ischemia," "Hypoxia," "Forensic," "Immunohistochemistry, and "Autopsy." The presentreview comprehensively highlights the current knowledge of specific IHC markers usedas gold standards during post-mortem investigation of acute myocardial infarction. Thepresent review comprehensively highlights the current knowledge of specific IHCmarkers used as gold standards during post-mortem investigation of acute myocardialinfarction, and some new potential immunohistochemical markers that can be used inthe early detection of myocardial infarction.
Collapse
Affiliation(s)
- Radu Moldovan
- Emergency County Hospital "Constantin Opris", Baia Mare, Department of Forensic Medicine, Street George Coșbuc 31, Baia Mare, Maramures, 430031, Romania.
| | - Vlad Andrei Ichim
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Department of Internal Medicine, Street Victor Babeș 8, Cluj-Napoca, Cluj, 400347, Romania.
| | - Vladimir Beliș
- University of Medicine and Pharmacy "Carol Davila" Bucharest Departament of Foresic Medicine, Street Bulevardul Eroii Sanitari 8, Bucharest, 050474, Romania.
| |
Collapse
|
4
|
Gao R, Li X. Extracellular Vesicles and Pathological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:17-31. [PMID: 37603270 DOI: 10.1007/978-981-99-1443-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Pathological cardiac hypertrophy is a well-recognized risk factor for cardiovascular diseases (CVDs). Although lots of efforts have been made to illustrate the underlying molecular mechanisms, many issues remain undiscovered. Recently, intercellular communication by delivering small molecules between different cell types in the progression of cardiac hypertrophy has been reported, including bioactive nucleic acids or proteins. These extracellular vesicles (EVs) may act in an autocrine or paracrine manner between cardiomyocytes and noncardiomyocytes to provoke or inhibit cardiac remodeling and hypertrophy. Besides, EVs can be used as novel diagnostic or prognostic biomarkers in cardiac hypertrophy and also may serve as potential therapeutic targets due to its biocompatible nature and low immunogenicity. In this chapter, we will first summarize the current knowledge about EVs from different cells in pathological cardiac hypertrophy. Then, we will focus on the value of EVs as therapeutic agents and biomarkers for pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Rongrong Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Zeng Z, Cai J, Liao Y, Sun S, Xie L. Progress in the effect of microRNA-21 on diseases via autophagy. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:936-941. [PMID: 36039591 PMCID: PMC10930284 DOI: 10.11817/j.issn.1672-7347.2022.210647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Indexed: 06/15/2023]
Abstract
Autophagy is a regulatory mechanism that packages damaged organelles, proteins, and pathogens to form vesicles and transports to lysosomes for degradation, enabling the recycle of useful components. Therefore, autophagy plays an important role in biological growth regulation and homeostasis. In the past two decades, growing evidence has shown that microRNA (miRNA) is closely related to autophagy. MiRNA-21 promotes or inhibits autophagy via regulating relevant pathways for different downstream target genes, and plays a role in tumors, ischemia-reperfusion injury, and other diseases.
Collapse
Affiliation(s)
- Zhengpeng Zeng
- Center of Health Management, Third Xiangya Hospital, Central South University, Changsha 410013.
- Department of Respiratory and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jinwen Cai
- Department of Respiratory and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yumei Liao
- Department of Respiratory and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shenghua Sun
- Department of Respiratory and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Lihua Xie
- Department of Respiratory and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
6
|
McLendon JM, Zhang X, Matasic DS, Kumar M, Koval OM, Grumbach IM, Sadayappan S, London B, Boudreau RL. Knockout of Sorbin And SH3 Domain Containing 2 (Sorbs2) in Cardiomyocytes Leads to Dilated Cardiomyopathy in Mice. J Am Heart Assoc 2022; 11:e025687. [PMID: 35730644 PMCID: PMC9333371 DOI: 10.1161/jaha.122.025687] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Sorbin and SH3 domain containing 2 (Sorbs2) protein is a cytoskeletal adaptor with an emerging role in cardiac biology and disease; yet, its potential relevance to adult‐onset cardiomyopathies remains underexplored. Sorbs2 global knockout mice display lethal arrhythmogenic cardiomyopathy; however, the causative mechanisms remain unclear. Herein, we examine Sorbs2 dysregulation in heart failure, characterize novel Sorbs2 cardiomyocyte‐specific knockout mice (Sorbs2‐cKO), and explore associations between Sorbs2 genetic variations and human cardiovascular disease. Methods and Results Bioinformatic analyses show myocardial Sorbs2 mRNA is consistently upregulated in humans with adult‐onset cardiomyopathies and in heart failure models. We generated Sorbs2‐cKO mice and report that they develop progressive systolic dysfunction and enlarged cardiac chambers, and they die with congestive heart failure at about 1 year old. After 3 months, Sorbs2‐cKO mice begin to show atrial enlargement and P‐wave anomalies, without dysregulation of action potential–associated ion channel and gap junction protein expressions. After 6 months, Sorbs2‐cKO mice exhibit impaired contractility in dobutamine‐treated hearts and skinned myofibers, without dysregulation of contractile protein expressions. From our comprehensive survey of potential mechanisms, we found that within 4 months, Sorbs2‐cKO hearts have defective microtubule polymerization and compensatory upregulation of structural cytoskeletal and adapter proteins, suggesting that this early intracellular structural remodeling is responsible for contractile dysfunction. Finally, we identified genetic variants that associate with decreased Sorbs2 expression and human cardiac phenotypes, including conduction abnormalities, atrial enlargement, and dilated cardiomyopathy, consistent with Sorbs2‐cKO mice phenotypes. Conclusions Our studies show that Sorbs2 is essential for maintaining structural integrity in cardiomyocytes, likely through strengthening the interactions between microtubules and other cytoskeletal proteins at cross‐link sites.
Collapse
Affiliation(s)
- Jared M McLendon
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Xiaoming Zhang
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Daniel S Matasic
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Department of Molecular Physiology and Biophysics University of Iowa Carver College of Medicine Iowa City IA
| | - Mohit Kumar
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Olha M Koval
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Isabella M Grumbach
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Sakthivel Sadayappan
- Department of Pharmacology and Systems Physiology University of Cincinnati OH.,Division of Cardiovascular Health and Disease Department of Internal Medicine Heart, Lung, and Vascular Institute University of Cincinnati OH
| | - Barry London
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| | - Ryan L Boudreau
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City IA.,Abboud Cardiovascular Research Center University of Iowa Carver College of Medicine Iowa City IA
| |
Collapse
|
7
|
Liu MM, Peng J, Guo YL, Zhu CG, Wu NQ, Xu RX, Dong Q, Cui CJ, Li JJ. SORBS2 as a molecular target for atherosclerosis in patients with familial hypercholesterolemia. J Transl Med 2022; 20:233. [PMID: 35590369 PMCID: PMC9118763 DOI: 10.1186/s12967-022-03381-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background Familial hypercholesterolemia (FH) is a metabolic disease in which patients are prone to develop premature atherosclerosis (AS). Sorbin and SH3 Domain Containing 2 (SORBS2) is known to play a role in coronary heart disease (CHD). However, the mechanism underlying SORBS2 involvement in the development of hypercholesterolemia remains unknown. Here, we investigated the effects of SORBS2 on inflammation and foam cell formation and its underlying mechanisms. Methods Using Bioinformatics analysis, we established that SORBS2 is upregulated in patients with FH. Circulating concentrations of SORBS2 were measured using ELISA kit (n = 30). The association between circulating SORBS2 levels and inflammatory factors or lipid indexes were conducted using Spearman correlation analysis. We further conducted in vitro experiments that the expression of SORBS2 were analyzed, and SORBS2 siRNA were transfected into oxidized LDL (OxLDL)-induced macrophages, followed by western blot and immunofluorescence. Results Circulating SORBS2 levels were positively associated with inflammatory factors and lipid indexes. We also observed that high in vitro expression of SORBS2 in OxLDL-induced macrophages. After SORBS2 silencing, Nod like receptor family pyrin domain-containing 3 protein(NLRP3)-Caspase1 activation and NF-κB activation were attenuated, and secretion of pro-inflammatory cytokines (IL-1β and IL-18) was decreased. Moreover, SORBS2 silencing blocked reactive oxygen species (ROS) production and lipid accumulation, and promoted cholesterol efflux through ABCG1-PPARγ pathway. Conclusions SORBS2 regulates lipid-induced inflammation and foam cell formation, and is a potential therapeutic target for hypercholesterolemia. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03381-z.
Collapse
Affiliation(s)
- Ming-Ming Liu
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Jia Peng
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Yuan-Lin Guo
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Cheng-Gang Zhu
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Na-Qiong Wu
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Rui-Xia Xu
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Qian Dong
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China
| | - Chuan-Jue Cui
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China.
| | - Jian-Jun Li
- Cardiovascular Metabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 BeiLiShi Road, XiCheng District, Beijing, China.
| |
Collapse
|
8
|
Liu X, Xu H, Xu H, Geng Q, Mak WH, Ling F, Su Z, Yang F, Zhang T, Chen J, Yang H, Wang J, Zhang X, Xu X, Jia H, Zhang Z, Liu X, Zhong S. New genetic variants associated with major adverse cardiovascular events in patients with acute coronary syndromes and treated with clopidogrel and aspirin. THE PHARMACOGENOMICS JOURNAL 2021; 21:664-672. [PMID: 34158603 PMCID: PMC8602039 DOI: 10.1038/s41397-021-00245-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 11/09/2022]
Abstract
Although a few studies have reported the effects of several polymorphisms on major adverse cardiovascular events (MACE) in patients with acute coronary syndromes (ACS) and those undergoing percutaneous coronary intervention (PCI), these genotypes account for only a small fraction of the variation and evidence is insufficient. This study aims to identify new genetic variants associated with MACE end point during the 18-month follow-up period by a two-stage large-scale sequencing data, including high-depth whole exome sequencing of 168 patients in the discovery cohort and high-depth targeted sequencing of 1793 patients in the replication cohort. We discovered eight new genotypes and their genes associated with MACE in patients with ACS, including MYOM2 (rs17064642), WDR24 (rs11640115), NECAB1 (rs74569896), EFR3A (rs4736529), AGAP3 (rs75750968), ZDHHC3 (rs3749187), ECHS1 (rs140410716), and KRTAP10-4 (rs201441480). Notably, the expressions of MYOM2 and ECHS1 are downregulated in both animal models and patients with phenotypes related to MACE. Importantly, we developed the first superior classifier for predicting 18-month MACE and achieved high predictive performance (AUC ranged between 0.92 and 0.94 for three machine-learning methods). Our findings shed light on the pathogenesis of cardiovascular outcomes and may help the clinician to make a decision on the therapeutic intervention for ACS patients.
Collapse
Affiliation(s)
- Xiaomin Liu
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Hanshi Xu
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Huaiqian Xu
- BGI-tech, BGI-Wuhan, Wuhan, 430075, Hubei, China
| | - Qingshan Geng
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P.R. China
| | - Wai-Ho Mak
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Zheng Su
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Fang Yang
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Tao Zhang
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Jiyan Chen
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P.R. China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, 310058, China
| | - Xiuqing Zhang
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen, 518083, China.,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China
| | - Zhiwei Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P.R. China
| | - Xiao Liu
- BGI-Shenzhen, Shenzhen, 518083, China. .,China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, 518083, China. .,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| | - Shilong Zhong
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P.R. China. .,Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P.R. China.
| |
Collapse
|
9
|
Di J, Yang M, Zhou H, Li M, Zhao J. MicroRNA-21-containing microvesicles from tubular epithelial cells promote cardiomyocyte hypertrophy. Ren Fail 2021; 43:391-400. [PMID: 33632070 PMCID: PMC7919913 DOI: 10.1080/0886022x.2021.1891098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Cardiomyocyte hypertrophy has been reported as one of the important mechanisms for cardiovascular disease (CVD) in patients with chronic kidney disease (CKD). MiroRNA-21(miR-21) was determined to play an important role in myocardial hypertrophy. However, the role of microvesicles (MVs) containing miR-21 in CKD-related cardiomyocyte hypertrophy remains largely unexplored. Methods Renal tubular epithelial cells were stimulated by transforming growth factor (TGF-β1), and the conditioned medium was extracted by differential centrifugation. Renal tubular epithelial cells were labeled with Dil-C18 dye and the recipient cardiomyocytes were observed by fluorescence microscope. MiR-21 level in MVs was detected by qRT-PCR, and the length and diameter of cardiomyocytes were measured by microscope. BCA protein kit and ANP kit were used to detect the content of cell protein and the level of ANP. MiR-21 inhibitor was transfected into cardiomyocytes to observe the effect of miR-21 on myocardial hypertrophy. Results TGF-β1 could induce donor renal tubular epithelial cells to produce MVs and delivered into cardiomyocytes, followed by the diameter, protein concentration and ANP content of cardiomyocytes significantly increased. Meanwhile, MiR-21 levels were markedly increased in MVs isolated from donor renal tubular epithelial cells and recipient cardiomyocytes. Pre-transfection of miR-21 inhibitors could inhibit MV-induced cardiomyocyte hypertrophy. Conclusion Tubular cells could secrete miR-21 by MVs and deliver it into recipient cardiomyocytes to induce cardiomyocyte hypertrophy. It might shed a new light on the mechanism and treatment of CKD-related cardiac dysfunction.
Collapse
Affiliation(s)
- Jia Di
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Min Yang
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Hua Zhou
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Min Li
- Department of Nephrology, The First People's Hospital of Changzhou, Changzhou, China
| | - Jiabi Zhao
- Department of Pathology, The Second People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
10
|
Quantitative Proteomic Analysis Using Formalin-Fixed, Paraffin-Embedded (FFPE) Human Cardiac Tissue. Methods Mol Biol 2021; 2261:525-533. [PMID: 33421012 DOI: 10.1007/978-1-0716-1186-9_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Clinical tissue archives represent an invaluable source of biological information. Formalin-fixed, paraffin-embedded (FFPE) tissue can be used for retrospective investigation of biomarkers of diseases and prognosis.Recently, the number of studies using proteome profiling of samples from clinical archives has markedly increased. However, the application of conventional quantitative proteomics technologies remains a challenge mainly due to the harsh fixation process resulting in protein cross-linking and protein degradation. In the present chapter, we demonstrate a protocol for label-free proteomic analysis of FFPE tissue prepared from human cardiac autopsies. The data presented here highlight the applicability and suitability of FFPE heart tissue for understanding the molecular mechanism of cardiac injury using a proteomics approach.
Collapse
|
11
|
Immunohistochemistry in the Detection of Early Myocardial Infarction: Systematic Review and Analysis of Limitations Because of Autolysis and Putrefaction. Appl Immunohistochem Mol Morphol 2020; 28:95-102. [PMID: 32044877 DOI: 10.1097/pai.0000000000000688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The postmortem diagnosis of acute myocardial infarction is one of the main problems in forensic practice, especially in cases in which death occurs soon after (from minutes to a few hours) the onset of the ischemic damage. Several authors have highlighted the possibility to overcome the limits of conventional histology in this diagnosis by utilizing immunohistochemistry. In the present research, we examined over 30 scientific studies and picked out over 20 main immunohistochemical antigens analyzed with a view to enabling the rapid diagnosis of early myocardial infarction. The aim of our review was to examine and summarize all the principal markers studied to date and also to consider their limitations, including protein alteration because of cadaveric autolysis and putrefaction.
Collapse
|
12
|
Proteomics in Deaths by Drowning: Diagnostic Efficacy of Apolipoprotein A1 and α-1Antitrypsin, Pilot Study. Diagnostics (Basel) 2020; 10:diagnostics10100747. [PMID: 32987960 PMCID: PMC7650832 DOI: 10.3390/diagnostics10100747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
Drowning is one of the leading causes of death worldwide. The pathophysiology of drowning is complex and, sometimes, interpretation of the circumstances of death in the autopsy becomes the main source of information in its diagnosis. New advances in medical research, such as proteomics, especially in forensic pathology, are still in the development. We proposed to investigate the application of Mass Spectrometry-based technologies, to identify differentially expressed proteins that may act as potential biomarkers in the postmortem diagnosis of drowning. We performed a pilot proteomic experiment with the inclusion of two drowned and two control forensic cases. After applying restrictive parameters, we identified apolipoprotein A1 (ApoA1) and α-1 antitrypsin as differentially expressed between the two diagnostic groups. A validation experiment, with the determination of both proteins in 25 forensic cases (16 drowned and 9 controls) was performed, and we corroborated ApoA1 higher values in the drowning group, whereas α-1 antitrypsin showed lower levels. After adjusting by confounder factors, both remained as predictive independent factors for diagnosis of drowning (p = 0.010 and p = 0.022, respectively). We constructed ROC curves for biomarkers’ levels attending at the origin of death and established an ApoA1 cut-off point of 100 mg/dL. Correct classification based on the diagnosis criteria was reached for 73.9% of the cases in a discriminant analysis. We propose apolipoprotein A1 (with our cutoff value for correct classification) and α-1 antitrypsin as valuable biomarkers of drowning. Our study, based on forensic cases, reveals our proteomic approach as a new complementary tool in the forensic diagnosis of drowning and, perhaps, in clinical future implications in drowned patients. However, this is a pilot approach, and future studies are necessary to consolidate our promising preliminary data.
Collapse
|
13
|
Azimzadeh O, Azizova T, Merl-Pham J, Blutke A, Moseeva M, Zubkova O, Anastasov N, Feuchtinger A, Hauck SM, Atkinson MJ, Tapio S. Chronic Occupational Exposure to Ionizing Radiation Induces Alterations in the Structure and Metabolism of the Heart: A Proteomic Analysis of Human Formalin-Fixed Paraffin-Embedded (FFPE) Cardiac Tissue. Int J Mol Sci 2020; 21:ijms21186832. [PMID: 32957660 PMCID: PMC7555548 DOI: 10.3390/ijms21186832] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/23/2022] Open
Abstract
Epidemiological studies on workers employed at the Mayak plutonium enrichment plant have demonstrated an association between external gamma ray exposure and an elevated risk of ischemic heart disease (IHD). In a previous study using fresh-frozen post mortem samples of the cardiac left ventricle of Mayak workers and non-irradiated controls, we observed radiation-induced alterations in the heart proteome, mainly downregulation of mitochondrial and structural proteins. As the control group available at that time was younger than the irradiated group, we could not exclude age as a confounding factor. To address this issue, we have now expanded our study to investigate additional samples using archival formalin-fixed paraffin-embedded (FFPE) tissue. Importantly, the control group studied here is older than the occupationally exposed (>500 mGy) group. Label-free quantitative proteomics analysis showed that proteins involved in the lipid metabolism, sirtuin signaling, mitochondrial function, cytoskeletal organization, and antioxidant defense were the most affected. A histopathological analysis elucidated large foci of fibrotic tissue, myocardial lipomatosis and lymphocytic infiltrations in the irradiated samples. These data highlight the suitability of FFPE material for proteomics analysis. The study confirms the previous results emphasizing the role of adverse metabolic changes in the radiation-associated IHD. Most importantly, it excludes age at the time of death as a confounding factor.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
- Correspondence: ; Tel.: +49-89-3187-3887
| | - Tamara Azizova
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Juliane Merl-Pham
- Helmholtz Zentrum München—German Research Centre for Environmental Health, Research Unit Protein Science, 80939 Munich, Germany; (J.M.-P.); (S.M.H.)
| | - Andreas Blutke
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Research Unit Analytical Pathology, 85764 Neuherberg, Germany; (A.B.); (A.F.)
| | - Maria Moseeva
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Olga Zubkova
- Southern Urals Biophysics Institute (SUBI), Russian Federation, 456780 Ozyorsk, Russia; (T.A.); (M.M.); (O.Z.)
| | - Natasa Anastasov
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
| | - Annette Feuchtinger
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Research Unit Analytical Pathology, 85764 Neuherberg, Germany; (A.B.); (A.F.)
| | - Stefanie M. Hauck
- Helmholtz Zentrum München—German Research Centre for Environmental Health, Research Unit Protein Science, 80939 Munich, Germany; (J.M.-P.); (S.M.H.)
| | - Michael J. Atkinson
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
- Chair of Radiation Biology, Technical University of Munich, 81675 Munich, Germany
| | - Soile Tapio
- Helmholtz Zentrum München—German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany; (N.A.); (M.J.A.); (S.T.)
| |
Collapse
|
14
|
Ding Y, Yang J, Chen P, Lu T, Jiao K, Tester DJ, Giudicessi JR, Jiang K, Ackerman MJ, Li Y, Wang DW, Lee H, Wang DW, Xu X. Knockout of SORBS2 Protein Disrupts the Structural Integrity of Intercalated Disc and Manifests Features of Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2020; 9:e017055. [PMID: 32808564 PMCID: PMC7660791 DOI: 10.1161/jaha.119.017055] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Background Sorbs2b (sorbin and SH3 domain-containing 2b) was recently identified as a cardiomyopathy gene from a zebrafish mutagenesis screen. However, cardiac functions of its mammalian ortholog remain elusive. Methods and Results We conducted a detailed expression and subcellular localization analysis of Sorbs2 ortholog in mice and a phenotypic characterization in Sorbs2 knockout mice. Sorbs2 is highly expressed in the mouse heart and encodes an adhesion junction/desmosome protein that is mainly localized to the intercalated disc. A mutation with near complete depletion of the Sorbs2 protein in mice results in phenotypes characteristic of human arrhythmogenic cardiomyopathy (ACM), including right ventricular dilation, right ventricular dysfunction, spontaneous ventricular tachycardia, and premature death. Sorbs2 is required to maintain the structural integrity of intercalated disc. Its absence resulted in profound cardiac electrical remodeling with impaired impulse conduction and action potential derangements. Targeted sequencing of human patients with ACM identified 2 rare splicing variants classified as likely pathogenic were in 2 unrelated individuals with ACM from a cohort of 59 patients with ACM. Conclusions The Sorbs2 knockout mouse manifests several key features reminiscent of human ACM. Although the candidacy of SORBS2 as a new ACM-susceptibility gene is supported by preliminary human genetics study, future validation in larger cohorts with ACM is needed.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Jingchun Yang
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Peng Chen
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tong Lu
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Kunli Jiao
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | | | | | - Kai Jiang
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Michael J. Ackerman
- Department of Cardiovascular Medicine (Division of Heart Rhythm Services)Mayo ClinicRochesterMN
- Pediatric and Adolescent Medicine (Division of Pediatric Cardiology)Mayo ClinicRochesterMN
- Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory)Mayo ClinicRochesterMN
| | - Yigang Li
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Dao Wu Wang
- State Key Laboratory of Reproductive MedicineClinical Center of Reproductive Medicine and Department of CardiologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - HoN‐chi Lee
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Dao Wen Wang
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaolei Xu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| |
Collapse
|
15
|
Qi Z, Wu D, Li M, Yan Z, Yang X, Ji N, Wang Y, Zhang J. The pluripotent role of exosomes in mediating non-coding RNA in ventricular remodeling after myocardial infarction. Life Sci 2020; 254:117761. [PMID: 32413403 DOI: 10.1016/j.lfs.2020.117761] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
With the increase of an aging population and the rising incidence of cardiovascular diseases, heart failure (HF) patients are on the rise every year. Myocardial infarction (MI) is the leading cause of HF in patients among cardiovascular diseases. In clinic, patients with MI are often assessed by biochemical indicators, electrocardiography, brain natriuretic peptide levels, myocardial enzymology, echocardiography and other means to predict the occurrence of HF and ventricular remodeling (VR). But there is still a lack of more accurate evaluation. VR is the basic mechanism of HF. In recent years, the molecular mechanism of VR has been studied mainly from the aspects of myocardial hypertrophy, myocardial fibrosis, inflammation, myocardial energy disorder, apoptosis, autophagy and pyroptosis. Exosomes are considered as the main mediators of intercellular information transmission. In addition, exosomes can promote the migration and transformation of intercellular RNAs, which are highly conserved non-coding RNAs. They can mediate the process of cell proliferation and differentiation of the target cell membrane. Exosomes have protective effects on VR after MI by inhibiting fibrosis, promoting angiogenesis and inhibiting inflammation and pyroptosis. We reviewed the specific protective mechanisms of exosomes for VR after MI. In addition, we discussed the formation of targeted exosomes and the role of non-coding RNAs in VR.
Collapse
Affiliation(s)
- Zhongwen Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China
| | - Dan Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoya Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Nan Ji
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yueyao Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China.
| |
Collapse
|
16
|
Hiratsuka T, Arakawa Y, Yajima Y, Kakimoto Y, Shima K, Yamazaki Y, Ikegami M, Yamamoto T, Fujiwake H, Fujimoto K, Yamada N, Tsuruyama T. Hierarchical Cluster and Region of Interest Analyses Based on Mass Spectrometry Imaging of Human Brain Tumours. Sci Rep 2020; 10:5757. [PMID: 32238824 PMCID: PMC7113320 DOI: 10.1038/s41598-020-62176-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/06/2020] [Indexed: 12/31/2022] Open
Abstract
Imaging mass spectrometry (IMS) has been rarely used to examine specimens of human brain tumours. In the current study, high quality brain tumour samples were selected by tissue observation. Further, IMS analysis was combined with a new hierarchical cluster analysis (IMS-HCA) and region of interest analysis (IMS-ROI). IMS-HCA was successful in creating groups consisting of similar signal distribution images of glial fibrillary acidic protein (GFAP) and related multiple proteins in primary brain tumours. This clustering data suggested the relation of GFAP and these identified proteins in the brain tumorigenesis. Also, high levels of histone proteins, haemoglobin subunit α, tubulins, and GFAP were identified in a metastatic brain tumour using IMS-ROI. Our results show that IMS-HCA and IMS-ROI are promising techniques for identifying biomarkers using brain tumour samples.
Collapse
Affiliation(s)
- Takuya Hiratsuka
- Department of Drug and Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Yoshiki Arakawa
- Department of Neural Surgery, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Yuka Yajima
- Department of Microbiology, Muroran Institute of Technology, Muroran, Hokkaido, 050-8585, Japan
| | - Yu Kakimoto
- Department of Forensic Medicine, Graduate School of Medicine, Tokai University School of Medicine, Isehara-Shimokasuya 143, Kanagawa, 259-1193, Japan
| | - Keisuke Shima
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Yuzo Yamazaki
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Masahiro Ikegami
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Takushi Yamamoto
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Hideshi Fujiwake
- Research Center, Shimadzu General Services, Inc., 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Koichi Fujimoto
- Department of Neural Surgery, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Norishige Yamada
- Clinical bioresource centre, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug and Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan. .,Clinical bioresource centre, Kyoto University Hospital, Kyoto, 606-8507, Japan.
| |
Collapse
|
17
|
Li C, Liu F, Liu S, Pan H, Du H, Huang J, Xie Y, Li Y, Zhao R, Wei Y. Elevated myocardial SORBS2 and the underlying implications in left ventricular noncompaction cardiomyopathy. EBioMedicine 2020; 53:102695. [PMID: 32143182 PMCID: PMC7058526 DOI: 10.1016/j.ebiom.2020.102695] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/19/2023] Open
Abstract
Background Left ventricular noncompaction cardiomyopathy (LVNC) is a hereditary heart disease characterized by an excessive trabecular meshwork of deep intertrabecular recesses within the ventricular myocardium. The guidelines for management of LVNC patients aim to improve quality of life by preventing cardiac heart failure. However, the mechanism underlying LVNC-associated heart failure remains poorly understood. Methods Using protein mass spectrometry analysis, we established that Sorbin And SH3 Domain Containing 2 (SORBS2) is up-regulated in LVNC hearts without changes to structure proteins. We conducted in vivo experiments wherein the heart tissues of wild-type mice were injected with an AAV9 vector to overexpress SORBS2, followed by analysis using echocardiography, T-tubule analysis and Ca2+ imaging to identify functional and morphological changes. In addition, we analyzed the function and structure of SORBS2 overexpressing human embryonic stem cell (hESC) derived cardiomyocytes (hESC-CM) via immunoblotting, immunohistochemistry, immunofluorescence, and confocal Ca2+ imaging. Findings LVNC myocardial tissues feature strongly elevated expression of SORBS2, microtubule densification and redistribution of Junctophilin 2 (JP2). SORBS2 interacts with β-tubulin, promoting its polymerization in 293T cells and hESC-derived CMs. In vivo, cardiac dysfunction, β-tubulin densification, JP2 translocation, T-tubule disorganization and Ca2+ handling dysfunction were observed in mice overexpressing SORBS2. Interpretation We identified a novel mechanism through which SORBS2 interacts with β-tubulin and promotes microtubule densification, eventually effecting JP2 distribution and T-tubule, potentially contributing to heart failure in LVNC disease. Fund This work was supported by a CAMS Initiative for Innovative Medicine grant (CAMS-I2M, 2016-I2M-1-015 to Y.J.Wei)
Collapse
Affiliation(s)
- Chunyan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Fan Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Haizhou Pan
- Children's Heart Center, the Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiwei Du
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Jian Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yuanyuan Xie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yanfen Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Ranxu Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China.
| |
Collapse
|
18
|
Pathak D, Srivastava AK, Padma MV, Gulati S, Rajeswari MR. Quantitative Proteomic and Network Analysis of Differentially Expressed Proteins in PBMC of Friedreich's Ataxia (FRDA) Patients. Front Neurosci 2019; 13:1054. [PMID: 31680804 PMCID: PMC6802492 DOI: 10.3389/fnins.2019.01054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/19/2019] [Indexed: 11/23/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by an expanded (GAA) trinucleotide repeat in the FXN gene. The extended repeats expansion results in reduced transcription and, thereby, decreased expression of the mitochondrial protein, frataxin. Given the ongoing drug trials, identification of reliable and easily accessible biomarkers for monitoring disease progression and therapeutic intervention is a foremost requirement. In this study, comparative proteomic profiling of PBMC proteins from FRDA patients and age- and gender-matched healthy controls was done using 2D-Differential in-Gel Electrophoresis (2D-DIGE). Protein–protein interaction (PPI) was analyzed using BioGRID and STRING pathway analysis tools. Using biological variance analysis (BVA) and LC/MS, we found eight differentially expressed proteins with fold change ≥1.5; p ≤ 0.05. Based on their cellular function, the identified proteins showed a strong pathological role in neuroinflammation, cardiomyopathy, compromised glucose metabolism, and iron transport, which are the major clinical manifestations of FRDA. Protein–protein network analysis of differentially expressed proteins with frataxin further supports their involvement in the pathophysiology of FRDA. Considering their crucial role in the cardiac and neurological complications, respectively, the two down-regulated proteins, actin α cardiac muscle 1 (ACTC1) and pyruvate dehydrogenase E1 component subunit β (PDHE1), are suggested as potential prognostic markers for FRDA.
Collapse
Affiliation(s)
- Deepti Pathak
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Achal Kumar Srivastava
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, New Delhi, India
| | - M V Padma
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Sheffali Gulati
- Department of Paediatrics, All India Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Moganty R Rajeswari
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, New Delhi, India
| |
Collapse
|
19
|
Zou L, Wang X, Guo Z, Sun H, Shao C, Yang Y, Sun W. Differential urinary proteomics analysis of myocardial infarction using iTRAQ quantification. Mol Med Rep 2019; 19:3972-3988. [PMID: 30942401 PMCID: PMC6471447 DOI: 10.3892/mmr.2019.10088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/06/2019] [Indexed: 11/06/2022] Open
Abstract
Myocardial infarction (MI) is a disease characterized by high morbidity and mortality rates. MI biomarkers are frequently used in clinical diagnosis; however, their specificity and sensitivity remain unsatisfactory. Urinary proteome is an easy, efficient and noninvasive source to examine biomarkers associated with various diseases. The present study, to the best of the authors' knowledge, is the first to examine the urinary proteome using the isobaric tags for relative and absolute quantitation (iTRAQ) technology to identify potential diagnostic biomarkers of MI. The urinary proteome was analyzed within 12 h following the first symptoms of early‑onset MI and at day 7 following percutaneous coronary intervention via iTRAQ labeling and two‑dimensional liquid chromatography‑tandem mass spectrometry. Candidate biomarkers were validated by multiple reaction monitoring (MRM) analysis. A total of 233 urinary proteins were differentially expressed. Gene enrichment analysis identified that the urinary proteome in patients with MI was associated with atherosclerosis, abnormal coagulation and abnormal cell metabolism. In total, 12 differentially expressed urinary proteins were validated by MRM analysis, five of which were associated with MI for the first time in the present study. Binary logistic regression analysis suggested that the combination of five urinary proteins (antithrombin‑III, complement C3, α‑1‑acid glycoprotein 1, serotransferrin and cathepsin Z) may be used to diagnose MI with 94% sensitivity and 93% specificity. In addition, the protein expression levels of three proteins were significantly restored to normal levels following surgical treatment. The verified candidate biomarkers may be used for the diagnosis of MI, and for monitoring the disease status and the effects of treatments for MI. The present results may facilitate future clinical applications of the urinary proteome to diagnose MI.
Collapse
Affiliation(s)
- Lili Zou
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Xubo Wang
- Department of Cardiology, The Fourth Hospital of Jilin University, Changchun, Jilin 130011, P.R. China
| | - Zhengguang Guo
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Haidan Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Chen Shao
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Yehong Yang
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Wei Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| |
Collapse
|
20
|
Neagu AN. Proteome Imaging: From Classic to Modern Mass Spectrometry-Based Molecular Histology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:55-98. [PMID: 31347042 DOI: 10.1007/978-3-030-15950-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to overcome the limitations of classic imaging in Histology during the actually era of multiomics, the multi-color "molecular microscope" by its emerging "molecular pictures" offers quantitative and spatial information about thousands of molecular profiles without labeling of potential targets. Healthy and diseased human tissues, as well as those of diverse invertebrate and vertebrate animal models, including genetically engineered species and cultured cells, can be easily analyzed by histology-directed MALDI imaging mass spectrometry. The aims of this review are to discuss a range of proteomic information emerging from MALDI mass spectrometry imaging comparative to classic histology, histochemistry and immunohistochemistry, with applications in biology and medicine, concerning the detection and distribution of structural proteins and biological active molecules, such as antimicrobial peptides and proteins, allergens, neurotransmitters and hormones, enzymes, growth factors, toxins and others. The molecular imaging is very well suited for discovery and validation of candidate protein biomarkers in neuroproteomics, oncoproteomics, aging and age-related diseases, parasitoproteomics, forensic, and ecotoxicology. Additionally, in situ proteome imaging may help to elucidate the physiological and pathological mechanisms involved in developmental biology, reproductive research, amyloidogenesis, tumorigenesis, wound healing, neural network regeneration, matrix mineralization, apoptosis and oxidative stress, pain tolerance, cell cycle and transformation under oncogenic stress, tumor heterogeneity, behavior and aggressiveness, drugs bioaccumulation and biotransformation, organism's reaction against environmental penetrating xenobiotics, immune signaling, assessment of integrity and functionality of tissue barriers, behavioral biology, and molecular origins of diseases. MALDI MSI is certainly a valuable tool for personalized medicine and "Eco-Evo-Devo" integrative biology in the current context of global environmental challenges.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania.
| |
Collapse
|
21
|
Cai H, Miao M, Wang Z. miR-214-3p promotes the proliferation, migration and invasion of osteosarcoma cells by targeting CADM1. Oncol Lett 2018; 16:2620-2628. [PMID: 30013657 PMCID: PMC6036594 DOI: 10.3892/ol.2018.8927] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 02/27/2018] [Indexed: 02/05/2023] Open
Abstract
Although osteosarcoma (OS) is the most common type of primary bone tumor in adolescents and young adults, its mechanism remains unclear. A previous study by the authors demonstrated that miR-214-3p was upregulated in OS patients. Therefore, the present study aimed to investigate the effect and molecular mechanism of miR-214-3p in OS cells. OS cell lines, U2OS and MNNG/HOS Cl#5, were transiently transfected with miR-214-3p mimics, a control mimic, miR-214-3p inhibitors and a control inhibitor. Subsequent assays revealed that elevated miR-214-3p promoted the proliferative, migratory and invasive abilities of OS cells, while the opposite effects were observed in cells that were transfected with miR-214-3p inhibitors. The interaction between miR-214-3p and cell adhesion molecule 1 (CADM1) 3'untranslated region (UTR) was verified by a dual luciferase assay, which indicated that the relative luciferase activity was decreased in 293T cells that were co-transfected with miR-214-3p mimic and psiCHECK2-CADM1-3'UTR compared with cells that were co-transfected with psiCHECK2-CADM1-3'UTR and control mimic. The knockdown of CADM1 using small-interfering RNA enhanced the proliferative, migratory and invasive abilities of OS cells. Furthermore, downregulated CADM1 expression increased the expression of phosphorylated P44/42 mitogen activated kinase (MAPK). In conclusion, miR-214-3p was able to directly target CADM1 and decrease its expression. This resulted in the activation of the P44/42 MAPK signaling pathway, and thereby promoted the proliferation, migration and invasion of OS cells.
Collapse
Affiliation(s)
- Haiqing Cai
- Pediatric Orthopedic Department, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200031, P.R. China
| | - Mingyuan Miao
- Pediatric Orthopedic Department, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200031, P.R. China
| | - Zhigang Wang
- Pediatric Orthopedic Department, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
22
|
Yajima Y, Hiratsuka T, Kakimoto Y, Ogawa S, Shima K, Yamazaki Y, Yoshikawa K, Tamaki K, Tsuruyama T. Region of Interest analysis using mass spectrometry imaging of mitochondrial and sarcomeric proteins in acute cardiac infarction tissue. Sci Rep 2018; 8:7493. [PMID: 29748547 PMCID: PMC5945593 DOI: 10.1038/s41598-018-25817-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022] Open
Abstract
Matrix-assisted laser desorption ionization image mass spectrometry (MALDI-IMS) has been developed for the identification of peptides in various tissues. The MALDI-IMS signal distribution patterns and quantification of the signal intensities of the regions of interest (ROI) with healthy regions were compared for identification of the disease specific biomarkers. We performed a new ROI analysis using the conventional t-test and data number independent Cohen’s d-value analysis. Using these techniques, we analysed heart tissues after acute myocardial infarction (AMI). As a result, IMS signals of mitochondrial adenosine triphosphate synthase alpha subunit (ATP5A), myosin-6/7(MYH6/7), aortic actin, and the myosin light chain 3 (MYL3) were identified in the infarcted region. In particular, the signals of MYH7 are significantly greater in the infarcted region using ROI analysis. ROI analysis using MALDI-IMS may be a promising technique for the identification of biomarkers for pathological studies that involve the comparison of diseased and control areas.
Collapse
Affiliation(s)
- Yuka Yajima
- Department of Microbiology, Muroran Institute of Technology, Muroran, Hokkaido, 050-8585, Japan
| | - Takuya Hiratsuka
- Department of Drug and Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan.
| | - Yu Kakimoto
- Department of Forensic Medicine, Graduate School of Medicine, Tokai University School of Medicine, Isehara-Shimokasuya 143, Kanagawa, 259-1193, Japan
| | - Shuichiro Ogawa
- Center for Anatomical, Pathological, and Forensic Medical Research, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Keisuke Shima
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Yuzo Yamazaki
- Kyoto Applications Development Center, Analytical & Measuring Instruments Division, Shimadzu Corporation, 1 Nishino-kyo-Kuwabara-cho, Kyoto, 604-8511, Japan
| | - Kenichi Yoshikawa
- Department of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Keiji Tamaki
- Department of Forensic Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug and Discovery Medicine, Pathology Division, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan.
| |
Collapse
|
23
|
Mondello C, Cardia L, Bartoloni G, Asmundo A, Ventura Spagnolo E. Immunohistochemical study on dystrophin expression in CAD-related sudden cardiac death: a marker of early myocardial ischaemia. Int J Legal Med 2018; 132:1333-1339. [PMID: 29732464 DOI: 10.1007/s00414-018-1843-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 04/04/2018] [Indexed: 01/26/2023]
Abstract
The aims of this study were to assess if dystrophin can be a tool for the forensic evaluation of sudden cardiac death due to coronary atherosclerotic disease (CAD) and particularly if it can be a marker of early myocardial ischaemia. Then in this investigation, the dystrophin was compared to C5b-9 and fibronectin to analyze if there are some differences in the expression of these proteins. Two groups of CAD-related sudden cardiac death, respectively the group 1 with gross and/or histological evidence and the group 2 with no specific histological signs of myocardial ischaemia were used. A third group formed by cases of acute mechanical asphyxiation was used as a control. The immunohistochemical staining by dystrophin, C5b-9 and fibronectin antibodies was performed. Loss of sarcolemmal dystrophin was observed in different degrees according to more or less significant histological evidence of myocardial ischaemia. Moreover, the comparison between loss of dystrophin expression and fibronectin positivity showed significant differences in group 2. The results suggested that dystrophin can be used in forensic diagnosis of CAD-related sudden cardiac death and as marker of early myocardial ischaemia.
Collapse
Affiliation(s)
- Cristina Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, via Consolare Valeria, 1, 98125, Messina, Italy
| | - Luigi Cardia
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria, 98125, Gazzi, Italy
| | - Giovanni Bartoloni
- Department of Anatomy, Diagnostic Pathology, Legal Medicine Hygiene and Public Health, University of Catania, Catania, Italy
| | - Alessio Asmundo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, via Consolare Valeria, 1, 98125, Messina, Italy
| | - Elvira Ventura Spagnolo
- Legal Medicine Section, Department for Health Promotion and Mother-Child Care, University of Palermo, Via del Vespro, 129, 90127, Palermo, Italy.
| |
Collapse
|
24
|
Miyagawa-Hayashino A, Yoshifuji H, Kitagori K, Ito S, Oku T, Hirayama Y, Salah A, Nakajima T, Kiso K, Yamada N, Haga H, Tsuruyama T. Increase of MZB1 in B cells in systemic lupus erythematosus: proteomic analysis of biopsied lymph nodes. Arthritis Res Ther 2018; 20:13. [PMID: 29382365 PMCID: PMC5791339 DOI: 10.1186/s13075-018-1511-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease in which dysregulation of B cells has been recognized. Here, we searched for potential biomarkers of SLE using liquid chromatography-tandem mass spectrometry (LC-MS). Methods Lymph nodes from SLE patients and controls were analyzed by LC-MS. To validate the identified molecules, immunoblotting and immunohistochemistry were performed and B cells from SLE patients were analyzed by quantitative RT-PCR. B-cell subsets from NZB/W F1 mice, which exhibit autoimmune disease resembling human SLE, were analyzed by flow cytometry. Endoplasmic reticulum (ER) stress was induced by tunicamycin and the serum concentration of anti-dsDNA antibodies was determined by ELISA. TUNEL methods and immunoblotting were used to assess the effect of tunicamycin. Results MZB1, which comprises part of a B-cell-specific ER chaperone complex and is a key player in antibody secretion, was one of the differentially expressed proteins identified by LC-MS and confirmed by immunoblotting. Immunohistochemically, larger numbers of MZB1+ cells were located mainly in interfollicular areas and scattered in germinal centers in specimens from SLE patients compared with those from controls. MZB1 colocalized with CD138+ plasma cells and IRTA1+ marginal zone B cells. MZB1 mRNA was increased by 2.1-fold in B cells of SLE patients with active disease (SLE Disease Activity Index 2000 ≥ 6) compared with controls. In aged NZB/W F1 mice, splenic marginal zone B cells and plasma cells showed elevated MZB1 levels. Tunicamycin induced apoptosis of MZB1+ cells in target organs, resulting in decreased serum anti-dsDNA antibody levels. Additionally, MZB1+ cells were increased in synovial tissue specimens from patients with rheumatoid arthritis. Conclusions MZB1 may be a potential therapeutic target in excessive antibody-secreting cells in SLE. Electronic supplementary material The online version of this article (10.1186/s13075-018-1511-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aya Miyagawa-Hayashino
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan. .,Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan. .,Present address: Department of Clinical Pathology, Kansai Medical University, Osaka, Japan.
| | - Hajime Yoshifuji
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kitagori
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Ito
- Bio Frontier Platform, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takuma Oku
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.,Research Portfolio & Science, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Japan
| | - Yoshitaka Hirayama
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.,Research Portfolio & Science, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Japan
| | - Adeeb Salah
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Toshiki Nakajima
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kaori Kiso
- Center for Anatomical, Pathological and Forensic Medical Research, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norishige Yamada
- Center for Anatomical, Pathological and Forensic Medical Research, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Center for Anatomical, Pathological and Forensic Medical Research, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
25
|
Aljakna A, Fracasso T, Sabatasso S. Molecular tissue changes in early myocardial ischemia: from pathophysiology to the identification of new diagnostic markers. Int J Legal Med 2018; 132:425-438. [DOI: 10.1007/s00414-017-1750-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
|
26
|
沈 安, 胡 水, 黄 敬, 郑 德, 胡 兆. [Screening and verification of plasma biomarkers for stable angina pectoris: a differential proteomic analysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1370-1374. [PMID: 29070468 PMCID: PMC6743951 DOI: 10.3969/j.issn.1673-4254.2017.10.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To compare and analyze the differentially expressed plasma proteome between patients with stable angina pectoris (SAP) and healthy donors to identify the biomarkers for early diagnosis of SAP. METHODS Plasma samples from 60 patients with SAP and 60 healthy controls were collected. Twenty samples (100 mL each) randomly selected from each group were pooled and after removing high-abundance proteins from the pooled plasma, two-dimensional gel electrophoresis (2DE) was performed to isolate the total proteins. The protein spots with more than 2 fold changes were selected after 2D analysis using software, and the differentially expressed proteins were identified by MALDI TOF/TOF mass spectrometer. ELISA was performed to detect hemoglobin subunit delta (HBD) levels in 40 randomly selected samples from each group for verification of the results of 2DE. RESULTS A total of 7 differentially expressed proteins were found in plasma samples from patients with SAP, including 3 up regulated proteins (serum albumin, hemoglobin subunit alpha and hemoglobin subunit delta,) and 4 down?regulated ones (apolipoprotein L1, apolipoprotein C3, apolipoprotein E and complement C4B). ELISA results showed that HBD level was increased in SAP plasma, which was consistent with the results of 2DE. CONCLUSION Patients with SAP have different plasma protein profiles from those of healthy controls, and HBD may serve as a potential specific biomarker for early diagnosis of SAP.
Collapse
Affiliation(s)
- 安娜 沈
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 水旺 胡
- 南方医科大学病理生理学教研室, 广东 广州 510515Department of Clinical Laboratory, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 敬 黄
- 南方医科大学第三附属医院 检验科, 广东 广州 510630Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - 德仲 郑
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 兆霆 胡
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
27
|
沈 安, 胡 水, 黄 敬, 郑 德, 胡 兆. [Screening and verification of plasma biomarkers for stable angina pectoris: a differential proteomic analysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1370-1374. [PMID: 29070468 PMCID: PMC6743951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To compare and analyze the differentially expressed plasma proteome between patients with stable angina pectoris (SAP) and healthy donors to identify the biomarkers for early diagnosis of SAP. METHODS Plasma samples from 60 patients with SAP and 60 healthy controls were collected. Twenty samples (100 mL each) randomly selected from each group were pooled and after removing high-abundance proteins from the pooled plasma, two-dimensional gel electrophoresis (2DE) was performed to isolate the total proteins. The protein spots with more than 2 fold changes were selected after 2D analysis using software, and the differentially expressed proteins were identified by MALDI TOF/TOF mass spectrometer. ELISA was performed to detect hemoglobin subunit delta (HBD) levels in 40 randomly selected samples from each group for verification of the results of 2DE. RESULTS A total of 7 differentially expressed proteins were found in plasma samples from patients with SAP, including 3 up regulated proteins (serum albumin, hemoglobin subunit alpha and hemoglobin subunit delta,) and 4 down?regulated ones (apolipoprotein L1, apolipoprotein C3, apolipoprotein E and complement C4B). ELISA results showed that HBD level was increased in SAP plasma, which was consistent with the results of 2DE. CONCLUSION Patients with SAP have different plasma protein profiles from those of healthy controls, and HBD may serve as a potential specific biomarker for early diagnosis of SAP.
Collapse
Affiliation(s)
- 安娜 沈
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 水旺 胡
- 南方医科大学病理生理学教研室, 广东 广州 510515Department of Clinical Laboratory, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 敬 黄
- 南方医科大学第三附属医院 检验科, 广东 广州 510630Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - 德仲 郑
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 兆霆 胡
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
28
|
Coelho Molck M, Simioni M, Vieira TP, Sgardioli IC, Monteiro FP, Souza J, Fett‐Conte AC, Félix TM, Monlléo IL, Gil‐da‐Silva‐Lopes VL. Genomic imbalances in syndromic congenital heart disease. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Molck MC, Simioni M, Paiva Vieira T, Sgardioli IC, Paoli Monteiro F, Souza J, Fett-Conte AC, Félix TM, Lopes Monlléo I, Gil-da-Silva-Lopes VL. Genomic imbalances in syndromic congenital heart disease. J Pediatr (Rio J) 2017; 93:497-507. [PMID: 28336264 DOI: 10.1016/j.jped.2016.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To identify pathogenic genomic imbalances in patients presenting congenital heart disease (CHD) with extra cardiac anomalies and exclusion of 22q11.2 deletion syndrome (22q11.2 DS). METHODS 78 patients negative for the 22q11.2 deletion, previously screened by fluorescence in situ hybridization (FISH) and/or multiplex ligation probe amplification (MLPA) were tested by chromosomal microarray analysis (CMA). RESULTS Clinically significant copy number variations (CNVs ≥300kb) were identified in 10% (8/78) of cases. In addition, potentially relevant CNVs were detected in two cases (993kb duplication in 15q21.1 and 706kb duplication in 2p22.3). Genes inside the CNV regions found in this study, such as IRX4, BMPR1A, SORBS2, ID2, ROCK2, E2F6, GATA4, SOX7, SEMAD6D, FBN1, and LTPB1 are known to participate in cardiac development and could be candidate genes for CHD. CONCLUSION These data showed that patients presenting CHD with extra cardiac anomalies and exclusion of 22q11.2 DS should be investigated by CMA. The present study emphasizes the possible role of CNVs in CHD.
Collapse
Affiliation(s)
- Miriam Coelho Molck
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética Médica, Campinas, SP, Brazil
| | - Milena Simioni
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética Médica, Campinas, SP, Brazil
| | - Társis Paiva Vieira
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética Médica, Campinas, SP, Brazil
| | | | - Fabíola Paoli Monteiro
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética Médica, Campinas, SP, Brazil
| | - Josiane Souza
- Centro de Atendimento Integral ao Fissurado Lábio Palatal (CAIF), Curitiba, PR, Brazil
| | - Agnes Cristina Fett-Conte
- Faculdade de Medicina de São José do Rio Preto, Departamento de Biologia Molecular, São José do Rio Preto, SP, Brazil
| | - Têmis Maria Félix
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Isabella Lopes Monlléo
- Universidade Federal de Alagoas (UFAL), Hospital Universitário, Faculdade de Medicina, Serviço de Genética Clínica, Maceió, AL, Brazil
| | | |
Collapse
|
30
|
Ceciliani F, Roccabianca P, Giudice C, Lecchi C. Application of post-genomic techniques in dog cancer research. MOLECULAR BIOSYSTEMS 2017; 12:2665-79. [PMID: 27345606 DOI: 10.1039/c6mb00227g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Omics techniques have been widely applied to veterinary science, although mostly on farm animal productions and infectious diseases. In canine oncology, on the contrary, the use of omics methodologies is still far behind. This review presents the most recent achievement in the application of postgenomic techniques, such as transcriptomics, proteomics, and metabolomics, to canine cancer research. The protocols to recover material suitable for omics analyses from formalin-fixed, paraffin-embedded tissues are presented, and omics applications for biomarker discovery and their potential for cancer diagnostics in veterinary medicine are highlighted.
Collapse
Affiliation(s)
- F Ceciliani
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - P Roccabianca
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - C Giudice
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - C Lecchi
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| |
Collapse
|
31
|
High Expression of Galectin-3 in Patients with IgG4-Related Disease: A Proteomic Approach. PATHOLOGY RESEARCH INTERNATIONAL 2017; 2017:9312142. [PMID: 28593065 PMCID: PMC5448067 DOI: 10.1155/2017/9312142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 12/24/2022]
Abstract
Objectives Immunoglobulin G4-related disease (IgG4-RD) is a multiorgan condition manifesting itself in different forms. This study aimed to investigate protein expression profiles and to find the possible biomarker for IgG4-RD by liquid chromatography mass spectrometry (LC-MS) using tissue sections in IgG4-RD patients. Methods Protein expression profiles in five IgG4-related pancreatitis and three normal pancreatic samples were compared using LC-MS and were validated by quantitative real-time PCR (qRT-PCR), immunoblotting, and immunohistochemistry. ELISA was employed in the serum of 20 patients with systemic IgG4-RD before and during steroid treatment. Results LC-MS indicated that the levels of 17 proteins were significantly higher and 12 others were significantly lower in IgG4-related pancreatitis patients compared to controls. Among these proteins, galectin-3 levels were 13-fold higher in IgG4-related pancreatitis (P < 0.01). These results were confirmed by immunoblotting and qRT-PCR. The average number of galectin-3 + cells in various organs of IgG4-RD patients, including salivary glands, lungs, and lymph nodes, was higher than in controls. Galectin-3 was detectable in macrophages, dendritic cells, and stromal myofibroblast-like cells, but not in lymphocytes by immunofluorescence staining. Serum galectin-3 levels were higher in patients with IgG4-RD compared with healthy donors and remained high during steroid therapy. Conclusion Galectin-3 was overexpressed in IgG4-RD and the levels were indirectly related to clinical activity.
Collapse
|
32
|
Role of microRNAs in sepsis. Inflamm Res 2017; 66:553-569. [DOI: 10.1007/s00011-017-1031-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 12/15/2022] Open
|
33
|
Mokou M, Lygirou V, Vlahou A, Mischak H. Proteomics in cardiovascular disease: recent progress and clinical implication and implementation. Expert Rev Proteomics 2017; 14:117-136. [DOI: 10.1080/14789450.2017.1274653] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marika Mokou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Vasiliki Lygirou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Harald Mischak
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Mosaiques Diagnostics, Hannover, Germany
| |
Collapse
|
34
|
Immunohistochemical detection of early myocardial infarction: a systematic review. Int J Legal Med 2016; 131:411-421. [PMID: 27885432 DOI: 10.1007/s00414-016-1494-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022]
Abstract
The postmortem diagnosis of early myocardial infarction is a challenge for forensic pathologists because the routine histology is neither specific. Many authors have suggested the use of the immunohistochemistry to fill the gaps in the histological diagnosis of early myocardial infarction. This review aims to analyse advances of immunohistochemical detection of early cardiac damage due to ischaemia. To this purpose, we reviewed experimental studies that investigated immunohistochemical markers and their estimated timing of expression. The review was performed according to specific inclusion and exclusion criteria, and a total of 23 studies assessing the immunohistochemical markers for the diagnosis and timing of early myocardial infarction were identified. The literature review highlights that the analysed markers are complement components, others being inflammatory mediators, cardiac cell proteins, plasma proteins, stress or hypoxia-induced factors and proteins associated with heart failure. All studies demonstrate the effectiveness of the tested markers in the early detection of myocardial infarction in both animal and human samples.
Collapse
|
35
|
Sepsis-associated cardiac dysfunction is controlled by small RNA molecules. J Mol Cell Cardiol 2016; 97:67-9. [DOI: 10.1016/j.yjmcc.2016.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/09/2016] [Indexed: 11/23/2022]
|
36
|
miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J Mol Cell Cardiol 2016; 94:43-53. [DOI: 10.1016/j.yjmcc.2016.03.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/23/2022]
|
37
|
Massengill MT, Ashraf HM, Chowdhury RR, Chrzanowski SM, Kar J, Warren SA, Walter GA, Zeng H, Kang BH, Anderson RH, Moss RL, Kasahara H. Acute heart failure with cardiomyocyte atrophy induced in adult mice by ablation of cardiac myosin light chain kinase. Cardiovasc Res 2016; 111:34-43. [PMID: 27025239 DOI: 10.1093/cvr/cvw069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/17/2016] [Indexed: 12/31/2022] Open
Abstract
AIMS Under pressure overload, initial adaptive hypertrophy of the heart is followed by cardiomyocyte elongation, reduced contractile force, and failure. The mechanisms governing the transition to failure are not fully understood. Pressure overload reduced cardiac myosin light chain kinase (cMLCK) by ∼80% within 1 week and persists. Knockdown of cMLCK in cardiomyocytes resulted in reduced cardiac contractility and sarcomere disorganization. Thus, we hypothesized that acute reduction of cMLCK may be causative for reduced contractility and cardiomyocyte remodelling during the transition from compensated to decompensated cardiac hypertrophy. METHODS AND RESULTS To mimic acute cMLCK reduction in adult hearts, the floxed-Mylk3 gene that encodes cMLCK was inducibly ablated in Mylk3(flox/flox)/merCremer mice (Mylk3-KO), and compared with two control mice (Mylk3(flox/flox) and Mylk3(+/+)/merCremer) following tamoxifen injection (50 mg/kg/day, 2 consecutive days). In Mylk3-KO mice, reduction of cMLCK protein was evident by 4 days, with a decline to below the level of detection by 6 days. By 7 days, these mice exhibited heart failure, with reduction of fractional shortening compared with those in two control groups (19.8 vs. 28.0% and 27.7%). Severely convoluted cardiomyocytes with sarcomeric disorganization, wavy fibres, and cell death were demonstrated in Mylk3-KO mice. The cardiomyocytes were also unable to thicken adaptively to pressure overload. CONCLUSION Our results, using a new mouse model mimicking an acute reduction of cMLCK, suggest that cMLCK plays a pivotal role in the transition from compensated to decompensated hypertrophy via sarcomeric disorganization.
Collapse
Affiliation(s)
- Michael T Massengill
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Hassan M Ashraf
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Rajib R Chowdhury
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Stephen M Chrzanowski
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Jeena Kar
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Sonisha A Warren
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| | - Huadong Zeng
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, University of Florida, Gainesville, FL, USA
| | - Byung-Ho Kang
- Electron Microscopy and Bio-imaging Laboratory, University of Florida, Gainesville, FL, USA
| | | | - Richard L Moss
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, 1600 SW Archer Rd, M543, Gainesville, FL 32610-0274, USA
| |
Collapse
|
38
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac Extracellular Vesicles in Normal and Infarcted Heart. Int J Mol Sci 2016; 17:ijms17010063. [PMID: 26742038 PMCID: PMC4730308 DOI: 10.3390/ijms17010063] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/10/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
Heart is a complex assembly of many cell types constituting myocardium, endocardium and epicardium that intensively communicate to each other in order to maintain the proper cardiac function. There are many types of intercellular intracardiac signals, with a prominent role of extracellular vesicles (EVs), such as exosomes and microvesicles, for long-distant delivering of complex messages. Cardiomyocytes release EVs, whose content could significantly vary depending on the stimulus. In stress, such as hypoxia, inflammation or injury, cardiomyocytes increase secretion of EVs. In hypoxic conditions, cardiac EVs are enriched with angiogenic and prosurvival factors. In acute myocardial infarction (AMI), damaged cardiac muscle cells produce EVs with increased content of angiogenic, anti-apoptotic, mitogenic and growth factors in order to induce repair and healing of the infarcted myocardium. Exosomal microRNAs play a central role in cardiac regeneration. In AMI, circulating cardiac EVs abundantly contain cardiac-specific miRNAs that serve as indicators of cardiac damage and have a big diagnostic potential as AMI biomarkers. Cardioprotective and regenerative properties of exosomes derived from cardiac and non-cardiac stem/progenitor cells are very helpful to be used in cell-free cardiotherapy and regeneration of post-infarct myocardium.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia.
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia.
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 143025 Moscow, Russia.
- Department of Biophysics, Biological Faculty, Moscow State University, 119991 Moscow, Russia.
| | - Yuri V Bobryshev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia.
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
- School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
39
|
Affiliation(s)
- Connie Wu
- From the Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA (C.W.); and the Early Career Committee of the American Heart Association Functional Genomics and Translational Biology Council, Dallas, TX (P.A.)
| | - Pankaj Arora
- From the Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA (C.W.); and the Early Career Committee of the American Heart Association Functional Genomics and Translational Biology Council, Dallas, TX (P.A.).
| |
Collapse
|
40
|
Kakimoto Y, Kamiguchi H, Ochiai E, Satoh F, Osawa M. MicroRNA Stability in Postmortem FFPE Tissues: Quantitative Analysis Using Autoptic Samples from Acute Myocardial Infarction Patients. PLoS One 2015; 10:e0129338. [PMID: 26046358 PMCID: PMC4457786 DOI: 10.1371/journal.pone.0129338] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/08/2015] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are very short (18–24 nucleotides) nucleic acids that are expressed in a number of biological tissues and have been shown to be more resistant to extreme temperatures and pH compared to longer RNA molecules, like mRNAs. As miRNAs contribute to diverse biological process and respond to various kinds of cellular stress, their utility as diagnostic biomarkers and/or therapeutic targets has recently been explored. Here, we have evaluated the usefulness of miRNA quantification during postmortem examination of cardiac tissue from acute myocardial infarction (AMI) patients. Cardiac tissue was collected within one week of the patient’s death and either frozen (19 samples) or fixed in formalin for up to three years (36 samples). RNA integrity was evaluated with an electropherogram, and it appears that longer RNAs are fragmented after death in the long-term fixed samples. Quantitative PCR was also performed for seven miRNAs and three other small RNAs in order to determine the appropriate controls for our postmortem analysis. Our data indicate that miR-191 and miR-26b are more suitable than the other types of small RNA molecules as they are stably detected after death and long-term fixation. Further, we also applied our quantitation method, using these endogenous controls, to evaluate the expression of three previously identified miRNA biomarkers, miR-1, miR-208b, and miR-499a, in formalin-fixed tissues from AMI patients. Although miR-1 and miR-208b decreased (1.4-fold) and increased (1.2-fold), respectively, in the AMI samples compared to the controls, the significance of these changes was limited by our sample size. In contrast, the relative level of miR-499a was significantly decreased in the AMI samples (2.1-fold). This study highlights the stability of miRNAs after death and long-term fixation, validating their use as reliable biomarkers for AMI during postmortem examination.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroshi Kamiguchi
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Eriko Ochiai
- Department of Forensic Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Fumiko Satoh
- Department of Forensic Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Motoki Osawa
- Department of Forensic Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
41
|
Lee J, Joo EJ, Lim HJ, Park JM, Lee KY, Park A, Seok A, Lee H, Kang HG. Proteomic analysis of serum from patients with major depressive disorder to compare their depressive and remission statuses. Psychiatry Investig 2015; 12:249-59. [PMID: 25866527 PMCID: PMC4390597 DOI: 10.4306/pi.2015.12.2.249] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/03/2014] [Accepted: 12/26/2014] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Currently, there are a few biological markers to aid in the diagnosis and treatment of depression. However, it is not sufficient for diagnosis. We attempted to identify differentially expressed proteins during depressive moods as putative diagnostic biomarkers by using quantitative proteomic analysis of serum. METHODS Blood samples were collected twice from five patients with major depressive disorder (MDD) at depressive status before treatment and at remission status during treatment. Samples were individually analyzed by liquid chromatography-tandem mass spectrometry for protein profiling. Differentially expressed proteins were analyzed by label-free quantification. Enzyme-linked immunosorbent assay (ELISA) results and receiver-operating characteristic (ROC) curves were used to validate the differentially expressed proteins. For validation, 8 patients with MDD including 3 additional patients and 8 matched normal controls were analyzed. RESULTS The quantitative proteomic studies identified 10 proteins that were consistently upregulated or downregulated in 5 MDD patients. ELISA yielded results consistent with the proteomic analysis for 3 proteins. Expression levels were significantly different between normal controls and MDD patients. The 3 proteins were ceruloplasmin, inter-alpha-trypsin inhibitor heavy chain H4 and complement component 1qC, which were upregulated during the depressive status. The depressive status could be distinguished from the euthymic status from the ROC curves for these proteins, and this discrimination was enhanced when all 3 proteins were analyzed together. CONCLUSION This is the first proteomic study in MDD patients to compare intra-individual differences dependent on mood. This technique could be a useful approach to identify MDD biomarkers, but requires additional proteomic studies for validation.
Collapse
Affiliation(s)
- Jiyeong Lee
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, Republic of Korea
| | - Eun-Jeong Joo
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Hee-Joung Lim
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jong-Moon Park
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Kyu Young Lee
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Arum Park
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, Republic of Korea
| | - AeEun Seok
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, Republic of Korea
| | - HooKeun Lee
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Hee-Gyoo Kang
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon, Republic of Korea
- Institute for Senior Industry, Eulji University, Seongnam, Republic of Korea
- Department of Biomedical Laboratory Science, Graduate School of Health Science, Eulji University, Seongnam, Republic of Korea
| |
Collapse
|
42
|
Schechter MA, Hsieh MKH, Njoroge LW, Thompson JW, Soderblom EJ, Feger BJ, Troupes CD, Hershberger KA, Ilkayeva OR, Nagel WL, Landinez GP, Shah KM, Burns VA, Santacruz L, Hirschey MD, Foster MW, Milano CA, Moseley MA, Piacentino V, Bowles DE. Phosphoproteomic profiling of human myocardial tissues distinguishes ischemic from non-ischemic end stage heart failure. PLoS One 2014; 9:e104157. [PMID: 25117565 PMCID: PMC4130503 DOI: 10.1371/journal.pone.0104157] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/06/2014] [Indexed: 12/31/2022] Open
Abstract
The molecular differences between ischemic (IF) and non-ischemic (NIF) heart failure are poorly defined. A better understanding of the molecular differences between these two heart failure etiologies may lead to the development of more effective heart failure therapeutics. In this study extensive proteomic and phosphoproteomic profiles of myocardial tissue from patients diagnosed with IF or NIF were assembled and compared. Proteins extracted from left ventricular sections were proteolyzed and phosphopeptides were enriched using titanium dioxide resin. Gel- and label-free nanoscale capillary liquid chromatography coupled to high resolution accuracy mass tandem mass spectrometry allowed for the quantification of 4,436 peptides (corresponding to 450 proteins) and 823 phosphopeptides (corresponding to 400 proteins) from the unenriched and phospho-enriched fractions, respectively. Protein abundance did not distinguish NIF from IF. In contrast, 37 peptides (corresponding to 26 proteins) exhibited a ≥ 2-fold alteration in phosphorylation state (p<0.05) when comparing IF and NIF. The degree of protein phosphorylation at these 37 sites was specifically dependent upon the heart failure etiology examined. Proteins exhibiting phosphorylation alterations were grouped into functional categories: transcriptional activation/RNA processing; cytoskeleton structure/function; molecular chaperones; cell adhesion/signaling; apoptosis; and energetic/metabolism. Phosphoproteomic analysis demonstrated profound post-translational differences in proteins that are involved in multiple cellular processes between different heart failure phenotypes. Understanding the roles these phosphorylation alterations play in the development of NIF and IF has the potential to generate etiology-specific heart failure therapeutics, which could be more effective than current therapeutics in addressing the growing concern of heart failure.
Collapse
Affiliation(s)
- Matthew A. Schechter
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael K. H. Hsieh
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Linda W. Njoroge
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - J. Will Thompson
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erik J. Soderblom
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bryan J. Feger
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Constantine D. Troupes
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Kathleen A. Hershberger
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Olga R. Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Whitney L. Nagel
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Gina P. Landinez
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Kishan M. Shah
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Virginia A. Burns
- Duke Translational Research Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lucia Santacruz
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew D. Hirschey
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care, Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Carmelo A. Milano
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - M. Arthur Moseley
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Valentino Piacentino
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Dawn E. Bowles
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
43
|
Shen X, Young R, Canty JM, Qu J. Quantitative proteomics in cardiovascular research: global and targeted strategies. Proteomics Clin Appl 2014; 8:488-505. [PMID: 24920501 DOI: 10.1002/prca.201400014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/02/2014] [Accepted: 06/06/2014] [Indexed: 11/05/2022]
Abstract
Extensive technical advances in the past decade have substantially expanded quantitative proteomics in cardiovascular research. This has great promise for elucidating the mechanisms of cardiovascular diseases and the discovery of cardiac biomarkers used for diagnosis and treatment evaluation. Global and targeted proteomics are the two major avenues of quantitative proteomics. While global approaches enable unbiased discovery of altered proteins via relative quantification at the proteome level, targeted techniques provide higher sensitivity and accuracy, and are capable of multiplexed absolute quantification in numerous clinical/biological samples. While promising, technical challenges need to be overcome to enable full utilization of these techniques in cardiovascular medicine. Here, we discuss recent advances in quantitative proteomics and summarize applications in cardiovascular research with an emphasis on biomarker discovery and elucidating molecular mechanisms of disease. We propose the integration of global and targeted strategies as a high-throughput pipeline for cardiovascular proteomics. Targeted approaches enable rapid, extensive validation of biomarker candidates discovered by global proteomics. These approaches provide a promising alternative to immunoassays and other low-throughput means currently used for limited validation.
Collapse
Affiliation(s)
- Xiaomeng Shen
- Department of Biochemistry, University at Buffalo, Buffalo, NY, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| | | | | | | |
Collapse
|
44
|
Vona B, Nanda I, Neuner C, Schröder J, Kalscheuer VM, Shehata-Dieler W, Haaf T. Terminal chromosome 4q deletion syndrome in an infant with hearing impairment and moderate syndromic features: review of literature. BMC MEDICAL GENETICS 2014; 15:72. [PMID: 24962056 PMCID: PMC4077152 DOI: 10.1186/1471-2350-15-72] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/23/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Terminal deletions of chromosome 4q are associated with a broad spectrum of phenotypes including cardiac, craniofacial, digital, and cognitive impairment. The rarity of this syndrome renders genotype-phenotype correlation difficult, which is further complicated by the widely different phenotypes observed in patients sharing similar deletion intervals. CASE PRESENTATION Herein, we describe a boy with congenital hearing impairment and a variety of moderate syndromic features that prompted SNP array analysis disclosing a heterozygous 6.9 Mb deletion in the 4q35.1q35.2 region, which emerged de novo in the maternal germ line. CONCLUSION In addition to the index patient, we review 35 cases from the literature and DECIPHER database to attempt genotype-phenotype correlations for a syndrome with great phenotypic variability. We delineate intervals with recurrent phenotypic overlap, particularly for cleft palate, congenital heart defect, intellectual disability, and autism spectrum disorder. Broad phenotypic presentation of the terminal 4q deletion syndrome is consistent with incomplete penetrance of the individual symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas Haaf
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
45
|
Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, Just A, Remke J, Zimmer K, Zeug A, Ponimaskin E, Schmiedl A, Yin X, Mayr M, Halder R, Fischer A, Engelhardt S, Wei Y, Schober A, Fiedler J, Thum T. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest 2014; 124:2136-46. [PMID: 24743145 DOI: 10.1172/jci70577] [Citation(s) in RCA: 761] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 02/20/2014] [Indexed: 12/20/2022] Open
Abstract
In response to stress, the heart undergoes extensive cardiac remodeling that results in cardiac fibrosis and pathological growth of cardiomyocytes (hypertrophy), which contribute to heart failure. Alterations in microRNA (miRNA) levels are associated with dysfunctional gene expression profiles associated with many cardiovascular disease conditions; however, miRNAs have emerged recently as paracrine signaling mediators. Thus, we investigated a potential paracrine miRNA crosstalk between cardiac fibroblasts and cardiomyocytes and found that cardiac fibroblasts secrete miRNA-enriched exosomes. Surprisingly, evaluation of the miRNA content of cardiac fibroblast-derived exosomes revealed a relatively high abundance of many miRNA passenger strands ("star" miRNAs), which normally undergo intracellular degradation. Using confocal imaging and coculture assays, we identified fibroblast exosomal-derived miR-21_3p (miR-21*) as a potent paracrine-acting RNA molecule that induces cardiomyocyte hypertrophy. Proteome profiling identified sorbin and SH3 domain-containing protein 2 (SORBS2) and PDZ and LIM domain 5 (PDLIM5) as miR-21* targets, and silencing SORBS2 or PDLIM5 in cardiomyocytes induced hypertrophy. Pharmacological inhibition of miR-21* in a mouse model of Ang II-induced cardiac hypertrophy attenuated pathology. These findings demonstrate that cardiac fibroblasts secrete star miRNA-enriched exosomes and identify fibroblast-derived miR-21* as a paracrine signaling mediator of cardiomyocyte hypertrophy that has potential as a therapeutic target.
Collapse
|
46
|
Indolfi C, Curcio A. Stargazing microRNA maps a new miR-21 star for cardiac hypertrophy. J Clin Invest 2014; 124:1896-8. [PMID: 24743143 DOI: 10.1172/jci75801] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Left ventricular hypertrophy is an initial compensatory mechanism in response to cardiac stress that can degenerate into heart failure and sudden cardiac death. Recent studies have shown that microRNAs (miRs) regulate several aspects of cardiovascular diseases. In this issue of the JCI, Bang and colleagues identified an exosome-mediated communication mechanism between cardiac fibroblasts and cardiomyocytes. Specifically, cardiac fibroblasts secrete miR-enriched exosomes, which are subsequently taken up by cardiomyocytes, in which they alter gene expression. In particular, a passenger strand miR, miR-21*, was identified as a potent paracrine factor that induces cardiomyocyte hypertrophy when shuttled through exosomes. These advanced comprehensive analyses represent a major step forward in our understanding of cardiovascular physiopathology, providing a promising adjunctive target for possible therapeutic approaches, namely the miR-mediated paracrine signaling network.
Collapse
|