1
|
Shitara Y, Konno R, Yoshihara M, Kashima K, Ito A, Mukai T, Kimoto G, Kakiuchi S, Ishikawa M, Kakihara T, Nagamatsu T, Takahashi N, Fujishiro J, Kawakami E, Ohara O, Kawashima Y, Watanabe E. Host-derived protein profiles of human neonatal meconium across gestational ages. Nat Commun 2024; 15:5543. [PMID: 39019879 PMCID: PMC11255260 DOI: 10.1038/s41467-024-49805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/19/2024] [Indexed: 07/19/2024] Open
Abstract
Meconium, a non-invasive biomaterial reflecting prenatal substance accumulation, could provide valuable insights into neonatal health. However, the comprehensive protein profile of meconium across gestational ages remains unclear. Here, we conducted an extensive proteomic analysis of first meconium from 259 newborns across varied gestational ages to delineate protein composition and elucidate its relevance to neonatal diseases. The first meconium samples were collected, with the majority obtained before feeding, and the mean time for the first meconium passage from the anus was 11.9 ± 9.47 h. Our analysis revealed 5370 host-derived meconium proteins, which varied depending on sex and gestational age. Specifically, meconium from preterm infants exhibited elevated concentrations of proteins associated with the extracellular matrix. Additionally, the protein profiles of meconium also exhibited unique variations depending on both specific diseases, including gastrointestinal diseases, congenital heart diseases, and maternal conditions. Furthermore, we developed a machine learning model to predict gestational ages using meconium proteins. Our model suggests that newborns with gastrointestinal diseases and congenital heart diseases may have immature gastrointestinal systems. These findings highlight the intricate relationship between clinical parameters and meconium protein composition, offering potential for a novel approach to assess neonatal gastrointestinal health.
Collapse
Affiliation(s)
- Yoshihiko Shitara
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryo Konno
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Masahito Yoshihara
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, Japan
| | - Kohei Kashima
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Ito
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Mukai
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Goh Kimoto
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satsuki Kakiuchi
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaki Ishikawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Tomo Kakihara
- Department of Pediatric Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Naoto Takahashi
- Department of Pediatrics, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Fujishiro
- Department of Pediatric Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiryo Kawakami
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Kanagawa, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan.
| | - Eiichiro Watanabe
- Department of Pediatric Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Surgery, Gunma Children's Medical Center, Gunma, Japan.
| |
Collapse
|
2
|
Sasset L, Manzo OL, Zhang Y, Marino A, Rubinelli L, Riemma MA, Chalasani MLS, Dasoveanu DC, Roviezzo F, Jankauskas SS, Santulli G, Bucci MR, Lu TT, Di Lorenzo A. Nogo-A reduces ceramide de novo biosynthesis to protect from heart failure. Cardiovasc Res 2023; 119:506-519. [PMID: 35815623 PMCID: PMC10226746 DOI: 10.1093/cvr/cvac108] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Maria Antonietta Riemma
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Madhavi Latha S Chalasani
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dragos C Dasoveanu
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fiorentina Roviezzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Stanislovas S Jankauskas
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Theresa T Lu
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
3
|
Li X, Le HT, Sato F, Kang TH, Makishima M, Zhong L, Liu Y, Guo L, Bhawal UK. Dec1 deficiency protects the heart from fibrosis, inflammation, and myocardial cell apoptosis in a mouse model of cardiac hypertrophy. Biochem Biophys Res Commun 2020; 532:513-519. [PMID: 32896382 DOI: 10.1016/j.bbrc.2020.08.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022]
Abstract
Cardiac inflammation and fibrosis triggered by left ventricular pressure overload are the major causes of heart dysfunction. Differentiated embryonic chondrocyte gene 1 (Dec1) is a basic helix-loop-helix transcription factor that is comprehensively involved in inflammation and tissue fibrosis, but its role in cardiac hypertrophy remains unclear. This study explored the effects of Dec1 on cardiac fibrosis, inflammation, and apoptosis in hypertrophic conditions. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy in wild-type (WT) mice and in Dec1 knock out (KO) mice for 4 weeks. Using the TAC mouse model, prominent differences in cardiac hypertrophy at the morphological, functional, and molecular levels were delineated by Masson's Trichrome and TUNEL staining, immunohistochemistry, RT-PCR and Western Blot. DNA microarray and microRNA (miRNA) array analyses were carried out to identify gene and miRNA expression patterns. Dec1KO mice exhibited a more severe hypertrophic heart, whereas WT mice showed a more pronounced perivascular fibrosis after TAC at 4 weeks. The Dec1 deficiency promoted M2 phenotype macrophages. Dec1KO TAC mice showed fewer apoptotic cells than WT TAC mice. APEX1, WNT16, FGF10 and MMP-10 were differentially expressed according to DNA microarray analysis and expression levels of those genes and the corresponding miRNAs (miR-295, miR-200 b, miR-130a, miR-92a) showed the same trends. Furthermore, luciferase reporter assay confirmed that FGF10 is the direct target gene of miR-130. In conclusion, a Dec1 deficiency protects the heart from perivascular fibrosis, regulates M1/M2 macrophage polarization and reduces cell apoptosis, which may provide a novel insight for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.
| | - Hue Thi Le
- Department of Physiology, Hanoi Medical University, Hanoi, Viet Nam.
| | - Fuyuki Sato
- Pathology Division, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Tong Ho Kang
- Graduate School of Biotechnology, Kyung Hee University, Republic of Korea.
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University, School of Medicine, Tokyo, Japan.
| | - Liangjun Zhong
- Department of Stomatology, Hangzhou Normal University, Hangzhou, China.
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.
| | - Lijia Guo
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Ujjal K Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan.
| |
Collapse
|
4
|
Chen L, Guan J, Wei Q, Yuan Z, Zhang M. Potential role of "omics" technique in prenatal diagnosis of congenital heart defects. Clin Chim Acta 2018; 482:185-190. [PMID: 29649453 DOI: 10.1016/j.cca.2018.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 02/08/2023]
Abstract
Congenital heart defect (CHD) is one of the most common birth defects and is the leading cause of neonatal death. Currently, there are no biomarkers available for prenatal diagnosis of CHD. Clinical strategies to diagnose CHD mostly depend on fetal echocardiography. Recent advances in "omics" techniques have opened up new possibilities for biomarker discoveries. In this review, we discuss recent advances in prenatal detection of CHD using biomarkers obtained by "omics" approaches, including genomics, proteomics, metabolomics, and others. There is great potential in obtaining various kinds of parameters using "omics" studies to facilitate early and accurate diagnosis of CHD.
Collapse
Affiliation(s)
- Lizhu Chen
- Department of Ultrasound, Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Johnny Guan
- Department of Urology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qiuju Wei
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Mo Zhang
- Department of Urology, Shengjing Hospital, China Medical University, Shenyang 110004, China..
| |
Collapse
|
5
|
Huang Q, Xi G, Alamdar A, Zhang J, Shen H. Comparative proteomic analysis reveals heart toxicity induced by chronic arsenic exposure in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 229:210-218. [PMID: 28599205 DOI: 10.1016/j.envpol.2017.05.077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/12/2017] [Accepted: 05/27/2017] [Indexed: 06/07/2023]
Abstract
Arsenic is a widespread metalloid in the environment, which poses a broad spectrum of adverse effects on human health. However, a global view of arsenic-induced heart toxicity is still lacking, and the underlying molecular mechanisms remain unclear. By performing a comparative quantitative proteomic analysis, the present study aims to investigate the alterations of proteome profile in rat heart after long-term exposure to arsenic. As a result, we found that the abundance of 81 proteins were significantly altered by arsenic treatment (35 up-regulated and 46 down-regulated). Among these, 33 proteins were specifically associated with cardiovascular system development and function, including heart development, heart morphology, cardiac contraction and dilation, and other cardiovascular functions. It is further proposed that the aberrant regulation of 14 proteins induced by arsenic would disturb cardiac contraction and relaxation, impair heart morphogenesis and development, and induce thrombosis in rats, which is mediated by the Akt/p38 MAPK signaling pathway. Overall, these findings will augment our knowledge of the involved mechanisms and develop useful biomarkers for cardiotoxicity induced by environmental arsenic exposure.
Collapse
Affiliation(s)
- Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Guochen Xi
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Ambreen Alamdar
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Jie Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Heqing Shen
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China.
| |
Collapse
|
6
|
Ding Y, Gao BB, Zhou L, Ye XH, Li H, Lai L, Huang JY. Clinical implications of plasma Nogo-A levels in patients with coronary heart disease. Arch Med Sci 2017; 13:771-777. [PMID: 28721144 PMCID: PMC5510510 DOI: 10.5114/aoms.2016.58713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/28/2015] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Nogo-A is an important neurite growth-regulatory protein in the adult and developing nervous system. Recently, increasing evidence has shown that Nogo-A plays important roles in cardiac development and may act as a potential indicator for heart failure. In addition, increased oxidative stress has been found in individuals with cardiovascular diseases. However, not much is known regarding the expression levels of Nogo-A and reactive oxygen species (ROS) in patients with coronary heart disease (CHD). Therefore, we sought to investigate the relationship between Nogo-A, ROS levels and CHD. MATERIAL AND METHODS The plasma Nogo-A and ROS concentrations of 122 acute coronary syndrome (ACS), 101 unstable angina pectoris (UAP), and 21 acute myocardial infarction (AMI) patients and 56 healthy controls were measured by enzyme-linked immunosorbent assay (ELISA). We further generated a receiver operating characteristic (ROC) curve to assess the diagnostic accuracy of Nogo-A and ROS in CHD. RESULTS The Nogo-A and ROS levels were significantly higher in patients with CHD than those in healthy controls. In addition, multivariate logistic regression analysis revealed that the level of Nogo-A (odds ratio (OR) = 1.624, 95% confidence interval: 1.125-2.293, p = 0.009) is a risk factor for prediction of CHD. Nogo-A has diagnostic value, with an optimal threshold of 5.466 ng/ml for maximized diagnostic performance (59% sensitivity and 78.6% specificity, area under curve, p < 0.05). However, ROS concentration is not a risk factor for prediction of CHD (OR = 0.999, 95% confidence interval: 0.997-1.001, p = 0.320). CONCLUSIONS Increased plasma Nogo-A level may be associated with CHD.
Collapse
Affiliation(s)
- Yu Ding
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Bei-Bei Gao
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Liang Zhou
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Xian-Hua Ye
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Hong Li
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Lei Lai
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Jin-Yu Huang
- Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| |
Collapse
|
7
|
Zhang Y, Huang Y, Cantalupo A, Azevedo PS, Siragusa M, Bielawski J, Giordano FJ, Di Lorenzo A. Endothelial Nogo-B regulates sphingolipid biosynthesis to promote pathological cardiac hypertrophy during chronic pressure overload. JCI Insight 2016; 1. [PMID: 27158676 DOI: 10.1172/jci.insight.85484] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular function by inhibiting the rate-limiting enzyme, serine palmitoyltransferase (SPT), in de novo sphingolipid biosynthesis. Here, we show that endothelium-derived sphingolipids, particularly sphingosine-1-phosphate (S1P), protect the heart from inflammation, fibrosis, and dysfunction following pressure overload and that Nogo-B regulates this paracrine process. SPT activity is upregulated in banded hearts in vivo as well as in TNF-α-activated endothelium in vitro, and loss of Nogo removes the brake on SPT, increasing local S1P production. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological cardiac hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B-deficient mice to WT levels, whereas SEW2871, an S1P1 receptor agonist, prevents myocardial permeability, inflammation, and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy and proposes a potential therapeutic target for the attenuation or reversal of this clinical condition.
Collapse
Affiliation(s)
- Yi Zhang
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Yan Huang
- Section of Cardiovascular Medicine, Department of Internal Medicine, and Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anna Cantalupo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Paula S Azevedo
- Department of Internal Medicine, Botucatu Medical School, University of Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Mauro Siragusa
- Center for Molecular Medicine, Institute for Vascular Signalling, Goethe University Frankfurt, Frankfurt, Germany
| | - Jacek Bielawski
- Lipidomics Mass Spectrometry Facility, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Frank J Giordano
- Section of Cardiovascular Medicine, Department of Internal Medicine, and Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Annarita Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
8
|
|
9
|
Huhta J, Linask KK. Environmental origins of congenital heart disease: the heart-placenta connection. Semin Fetal Neonatal Med 2013; 18:245-50. [PMID: 23751925 DOI: 10.1016/j.siny.2013.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although the mammalian embryo is well protected in the uterus, environmental chemicals, drugs, and maternal nutritional imbalances can interfere with regulatory pathways directing placental and embryonic development early in gestation. Embryonic cells are most susceptible to environmental influences during cellular specification and differentiation stages. Because biochemical differentiation precedes morphological outcome often by days, the period of susceptibility to environmental chemicals expectedly precedes visible morphogenic effects. The cellular mechanisms by which drugs and other environmental factors disrupt embryonic development and induce cardiac abnormalities have remained undefined.
Collapse
Affiliation(s)
- James Huhta
- All Children's Hospital, Johns Hopkins Medicine, St Petersburg, FL, USA.
| | | |
Collapse
|
10
|
Bao B, Wang Y, Hu H, Yao H, Li Y, Tang S, Zheng L, Xu Y, Liang Z. Karyotypic and molecular genetic changes associated with fetal cardiovascular abnormalities: results of a retrospective 4-year ultrasonic diagnosis study. Int J Biol Sci 2013; 9:463-71. [PMID: 23678296 PMCID: PMC3654495 DOI: 10.7150/ijbs.5404] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/22/2013] [Indexed: 12/04/2022] Open
Abstract
Objective: To investigate the incidence of aneuploidy in fetuses with congenital heart defects (CHDs) and to further identify submicroscopic changes and global DNA methylation levels as potential biomarkers in complex CHD cases. Methods: Fetuses at high risk for birth defects or with obvious sonographic anomalies were recruited at the Prenatal Diagnosis Center and Ultrasonic Diagnosis Center. Elective fetal karyotyping and DNA copy number and promoter methylation analyses were carried out following parental consent. G-banded karyotyping was performed to detect fetal aneuploidy. Copy number variations (CNVs) were detected using the Affymetrix SNP Array 6.0 and validated by real time PCR. Global DNA methylation analyses were conducted using a Roche NimbleGen Human DNA Methylation 3x720K Array, and DNA methylation differences were assayed by a Sequenom MassARRAY EpiTYPER. Results: Conventional karyotyping identified 30 cases with aneuploidy in 179 CHD fetuses. Various CNVs were found in two aneuploid fetuses and in five euploid CHD fetuses. Verified segmental deletion or duplications were not directly associated with cardiovascular malformations except in DAAM1 and GATA6. Verifiable aberrant DNA methylation could not be identified in three complex CHD fetuses. Conclusions: In this study, Trisomy 18, Trisomy 21 and 45,XO were the most common aneuploidies identified in CHD fetuses. In the affected samples, only DAAM1 deletion and GATA6 amplification could be associated with cardiovascular biological processes.
Collapse
Affiliation(s)
- Bihui Bao
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
The heart-placenta axis in the first month of pregnancy: induction and prevention of cardiovascular birth defects. J Pregnancy 2013; 2013:320413. [PMID: 23691322 PMCID: PMC3652177 DOI: 10.1155/2013/320413] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 03/04/2013] [Accepted: 03/13/2013] [Indexed: 01/19/2023] Open
Abstract
Extrapolating from animal studies to human pregnancy, our studies showed that folate (FA) deficiency as well as one-time exposure to environmental factors in the first two to three weeks of human gestation can result in severe congenital heart defects (CHDs). Considering that approximately 49% of pregnancies are unplanned, this period of pregnancy can be considered high-risk for cardiac, as well as for neural, birth defects, as the woman usually is not aware of her pregnancy and may not yet be taking precautionary actions to protect the developing embryo. Using avian and mouse vertebrate models, we demonstrated that FA supplementation prevents CHD induced by alcohol, lithium, or elevation of the metabolite homocysteine, a marker for FA deficiency. All three factors affected the important Wnt signaling pathway by suppressing Wnt-mediated gene expression in the heart fields, resulting in a delay of cardiomyocyte migration, cardiomyogenesis, and CHD. Optimal protection of cardiogenesis was observed to occur with FA supplementation provided upon morning after conception and at higher doses than the presently available in prenatal vitamin supplementation. Our studies demonstrate pathways and cell processes that are involved with protection of one-carbon metabolism during heart development.
Collapse
|
12
|
Gould RA, Aboulmouna LM, Varner JD, Butcher JT. Hierarchical approaches for systems modeling in cardiac development. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 5:289-305. [PMID: 23463736 DOI: 10.1002/wsbm.1217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ordered cardiac morphogenesis and function are essential for all vertebrate life. The heart begins as a simple contractile tube, but quickly grows and morphs into a multichambered pumping organ complete with valves, while maintaining regulation of blood flow and nutrient distribution. Though not identical, cardiac morphogenesis shares many molecular and morphological processes across vertebrate species. Quantitative data across multiple time and length scales have been gathered through decades of reductionist single variable analyses. These range from detailed molecular signaling pathways at the cellular levels to cardiac function at the tissue/organ levels. However, none of these components act in true isolation from others, and each, in turn, exhibits short- and long-range effects in both time and space. With the absence of a gene, entire signaling cascades and genetic profiles may be shifted, resulting in complex feedback mechanisms. Also taking into account local microenvironmental changes throughout development, it is apparent that a systems level approach is an essential resource to accelerate information generation concerning the functional relationships across multiple length scales (molecular data vs physiological function) and structural development. In this review, we discuss relevant in vivo and in vitro experimental approaches, compare different computational frameworks for systems modeling, and the latest information about systems modeling of cardiac development. Finally, we conclude with some important future directions for cardiac systems modeling.
Collapse
Affiliation(s)
- Russell A Gould
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | | | | |
Collapse
|
13
|
Dawson K, Aflaki M, Nattel S. Role of the Wnt-Frizzled system in cardiac pathophysiology: a rapidly developing, poorly understood area with enormous potential. J Physiol 2012. [PMID: 23207593 DOI: 10.1113/jphysiol.2012.235382] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract The Wnt-Frizzled (Fzd) G-protein-coupled receptor system, involving 19 distinct Wnt ligands and 10 Fzd receptors, plays key roles in the development and functioning of many organ systems. There is increasing evidence that Wnt-Fzd signalling is important in regulating cardiac function. Wnt-Fzd signalling primarily involves a canonical pathway, with dishevelled-1-dependent nuclear translocation of β-catenin that derepresses Wnt-sensitive gene transcription, but can also include non-canonical pathways via phospholipase-C/Ca(2+) mobilization and dishevelled-protein activation of small GTPases. Wnt-Fzd effects vary with specific ligand/receptor interactions and associated downstream pathways. This paper reviews the biochemistry and physiology of the Wnt-Fzd complex, and presents current knowledge of Wnt signalling in cardiac remodelling processes such as hypertrophy and fibrosis, as well as disease states such as myocardial infarction (MI), heart failure and arrhythmias. Wnt signalling is activated during hypertrophy; inhibiting Wnt signalling by activating glycogen synthase kinase attenuates the hypertrophic response. Wnt signalling has complex and time-dependent actions post-MI, so that either beneficial or harmful effects might result from Wnt-directed interventions. Stem cell biology, a promising area for therapeutic intervention, is highly regulated by Wnt signalling. The Wnt system regulates fibroblast function, and is prominently altered in arrhythmogenic ventricular cardiomyopathy, a familial disease involving excess deposition of fibroadipose tissue. Wnt signalling controls connexin43 expression, thereby contributing to the regulation of cardiac electrical stability and arrhythmia generation. Although much has been learned about Wnt-Fzd signalling in hypertrophy and infarction, its role is poorly understood for a broad range of other heart disorders. Much more needs to be learned for its contributions to be fully appreciated, and to permit more effective exploitation of its enormous potential in therapeutic development.
Collapse
Affiliation(s)
- Kristin Dawson
- S. Nattel: 5000 Belanger St. E, Montreal, Quebec, Canada H1T 1C8.
| | | | | |
Collapse
|
14
|
Bao B, Zhang L, Hu H, Yin S, Liang Z. Deletion of a single-copy DAAM1 gene in congenital heart defect: a case report. BMC MEDICAL GENETICS 2012; 13:63. [PMID: 22857009 PMCID: PMC3482563 DOI: 10.1186/1471-2350-13-63] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 07/24/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND With an increasing incidence of congenital heart defects (CHDs) in recent years, genotype-phenotype correlation and array-based methods have contributed to the genome-wide analysis and understanding of genetic variations in the CHD population. Here, we report a copy number deletion of chromosomal 14q23.1 in a female fetus with complex congenital heart defects. This is the first description of DAAM1 gene deletion associated with congenital heart anomalies. CASE PRESENTATION Compared with the control population, one CHD fetus showed a unique copy number deletion of 14q23.1, a region that harbored DAAM1 and KIAA0666 genes. CONCLUSIONS Results suggest that the copy number deletion on chromosome 14q23.1 may be critical for cardiogenesis. However, the exact relationship and mechanism of how DAAM1 and KIAA0666 deletion contributes to the onset of CHD is yet to be determined.
Collapse
Affiliation(s)
- Bihui Bao
- Department of Gynecology and Obstetrics, South-West Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
15
|
Sgarra L, Addabbo F, Potenza MA, Montagnani M. Determinants of evolving metabolic and cardiovascular benefit/risk profiles of rosiglitazone therapy during the natural history of diabetes: molecular mechanisms in the context of integrated pathophysiology. Am J Physiol Endocrinol Metab 2012; 302:E1171-82. [PMID: 22374753 DOI: 10.1152/ajpendo.00038.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Rosiglitazone is a thiazolidinedione, a synthetic PPARγ receptor agonist with insulin-sensitizing properties that is used as an antidiabetic drug. In addition to improving glycemic control through actions in metabolic target tissues, rosiglitazone has numerous biological actions that impact on cardiovascular homeostasis. Some of these actions are helpful (e.g., improving endothelial function), whereas others are potentially harmful (e.g., promoting fluid retention). Since cardiovascular morbidity and mortality are major endpoints for diabetes, it is essential to understand how the natural history of diabetes alters the net benefits and risks of rosiglitazone therapy. This complex issue is an important determinant of optimal use of rosiglitazone and is critical for understanding cardiovascular safety issues. We give special attention to the effects of rosiglitazone in diabetic patients with stable coronary artery disease and the impact of rosiglitazone actions on atherosclerosis and plaque instability. This provides a rational conceptual framework for predicting evolving benefit/risk profiles that inform optimal use of rosiglitazone in the clinical setting and help explain the results of recent large clinical intervention trials where rosiglitazone had disappointing cardiovascular outcomes. Thus, in this perspective, we describe what is known about the molecular mechanisms of action of rosiglitazone on cardiovascular targets in the context of the evolving pathophysiology of diabetes over its natural history.
Collapse
Affiliation(s)
- Luca Sgarra
- Department of Biomedical Sciences and Human Oncology, Medical School, University of Bari, Bari, Italy
| | | | | | | |
Collapse
|
16
|
Gude N, Sussman M. Notch signaling and cardiac repair. J Mol Cell Cardiol 2012; 52:1226-32. [PMID: 22465038 DOI: 10.1016/j.yjmcc.2012.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/12/2012] [Accepted: 03/13/2012] [Indexed: 02/04/2023]
Abstract
Notch signaling is critical for proper heart development and recently has been reported to participate in adult cardiac repair. Notch resides at the cell surface as a single pass transmembrane receptor, transits through the cytoplasm following activation, and acts as a transcription factor upon entering the nucleus. This dynamic and widespread cellular distribution allows for potential interactions with many signaling and binding partners. Notch displays temporal as well as spatial versatility, acting as a strong developmental signal, controlling cell fate determination and lineage commitment, and playing a pivotal role in embryonic and adult stem cell proliferation and differentiation. This review serves as an update of recent literature addressing Notch signaling in the heart, with attention to findings from noncardiac research that provide clues for further interpretation of how the Notch pathway influences cardiac biology. Specific areas of focus include Notch signaling in adult myocardium following pathologic injury, the role of Notch in cardiac progenitor cells with respect to differentiation and cardiac repair, crosstalk between Notch and other cardiac signaling pathways, and emerging aspects of noncanonical Notch signaling in heart.
Collapse
Affiliation(s)
- Natalie Gude
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | | |
Collapse
|
17
|
Li H, Chen Y, Zhou B, Peng Y, Bai W, Rao L. RNT4 3'-UTR insertion/deletion polymorphisms are not associated with atrial septal defect in Chinese Han population: a brief communication. DNA Cell Biol 2012; 31:1121-4. [PMID: 22313113 DOI: 10.1089/dna.2011.1386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Atrial septal defect (ASD) is a common type of congenital heart disease, which is defined as any communication through atrial septum. Several studies have revealed that genetic factors may influence the susceptibility of ASD. Recent studies have shown that reticulon 4 (RTN4) gene might be involved in some processes relevant to heart development, such as regulation of cell migration and vascular remodeling. This study aimed to evaluate RTN4 gene polymorphisms of CAA and TATC insertion/deletion in relation to the risk of ASD in Chinese Han population. A total of 175 ASD patients and 308 unrelated healthy controls were successfully investigated. The polymorphisms of patients were determined by polymerase chain reaction-polyacrylamide gel electrophoresis. There was no significant difference in the allele frequencies of CAA and TATC insertion/deletion in RNT4 gene between ASD patients and controls. The same results were seen in their genotypes. The present study suggests that CAA and TATC insertion/deletion polymorphisms of RNT4 gene may not be a useful marker to predict the susceptibility of ASD in Chinese Han population.
Collapse
Affiliation(s)
- Hui Li
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | | | | | | | | | | |
Collapse
|
18
|
Amano M, Hashimoto R, Nishimura SI. Effects of single genetic damage in carbohydrate-recognizing proteins in mouse serum N-glycan profile revealed by simple glycotyping analysis. Chembiochem 2012; 13:451-64. [PMID: 22271523 DOI: 10.1002/cbic.201100595] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Indexed: 12/29/2022]
Abstract
Gene knock-out of C-type lectin receptors expressed in dendritic cells induced significant alteration of serum N-glycans compared with that of gender-matched controls. Glycotyping analysis suggested that putative-core fucosylation is strongly influenced by differences in the dominant mechanisms after carbohydrate recognition by pattern-recognition receptors, endocytosis of ligands, or induction of cytokines/chemokines. However, the loss of galectin-9, a ligand for T-helper type 1-specific cell-surface molecule, did not affect most N-glycan profiles. Interestingly, lack of the Chst3 gene (chondroitin 6-sulfotransferase) appeared to influence markedly the expression of most N-glycans, especially highly modified glycoforms bearing multiple Neu5Gc, Fuc, and LacNAc units. In contrast, genetic mutations in B4galnt1 and B4galnt2 (GalNAc transferase, responsible for the synthesis of many gangliosides) induced no discernable alteration. These results indicate that the biosynthesis of N-glycans of serum glycoproteins can be affected not only by direct genetic mutations in the glycosyltransferases but also by changes in metabolite availability in sugar nucleotide synthesis and Golgi N-glycosylation pathways caused concertedly in whole cells, tissues, and organs by milder deficiencies in immune cell-surface lectins. Many common chronic conditions, such as autoimmunity, metabolic syndrome, and aging/dementia result.
Collapse
Affiliation(s)
- Maho Amano
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Graduate School of Life Sciences, Hokkaido University, Sapporo 001-0021, Japan.
| | | | | |
Collapse
|
19
|
Jiang LH, Gamper N, Beech DJ. Properties and therapeutic potential of transient receptor potential channels with putative roles in adversity: focus on TRPC5, TRPM2 and TRPA1. Curr Drug Targets 2011; 12:724-36. [PMID: 21291387 PMCID: PMC3267159 DOI: 10.2174/138945011795378568] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 06/28/2010] [Indexed: 01/13/2023]
Abstract
Mammals contain 28 genes encoding Transient Receptor Potential (TRP) proteins. The proteins assemble into cationic channels, often with calcium permeability. Important roles in physiology and disease have emerged and so there is interest in whether the channels might be suitable therapeutic drug targets. Here we review selected members of three subfamilies of mammalian TRP channel (TRPC5, TRPM2 and TRPA1) that show relevance to sensing of adversity by cells and biological systems. Summarized are the cellular and tissue distributions, general properties, endogenous modulators, protein partners, cellular and tissue functions, therapeutic potential, and pharmacology. TRPC5 is stimulated by receptor agonists and other factors that include lipids and metal ions; it heteromultimerises with other TRPC proteins and is involved in cell movement and anxiety control. TRPM2 is activated by hydrogen peroxide; it is implicated in stress-related inflammatory, vascular and neurodegenerative conditions. TRPA1 is stimulated by a wide range of irritants including mustard oil and nicotine but also, controversially, noxious cold and mechanical pressure; it is implicated in pain and inflammatory responses, including in the airways. The channels have in common that they show polymodal stimulation, have activities that are enhanced by redox factors, are permeable to calcium, and are facilitated by elevations of intracellular calcium. Developing inhibitors of the channels could lead to new agents for a variety of conditions: for example, suppressing unwanted tissue remodeling, inflammation, pain and anxiety, and addressing problems relating to asthma and stroke.
Collapse
Affiliation(s)
- L H Jiang
- Institute of Membrane & Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | | |
Collapse
|
20
|
AL-Shawaf E, Tumova S, Naylor J, Majeed Y, Li J, Beech DJ. GVI phospholipase A2 role in the stimulatory effect of sphingosine-1-phosphate on TRPC5 cationic channels. Cell Calcium 2011; 50:343-50. [PMID: 21742378 PMCID: PMC3195672 DOI: 10.1016/j.ceca.2011.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/08/2011] [Accepted: 06/09/2011] [Indexed: 11/16/2022]
Abstract
The Transient Receptor Potential Canonical 5 (TRPC5) protein forms calcium-permeable cationic channels that are stimulated by G protein-coupled receptor agonists. The signaling pathways of such agonist effects are poorly understood. Here we investigated the potential for involvement of lysophosphatidylcholine (LPC) and arachidonic acid generated by group 6 (GVI) phospholipase A2 (PLA2) enzymes, focusing on stimulation of TRPC5 by sphingosine-1-phosphate (S1P) which acts via a pertussis toxin-sensitive (Gi/o protein) pathway without Ca2+-release. Experiments were on HEK 293 cells containing conditional expression of human TRPC5. Channel activity was recorded using an intracellular calcium indicator or whole-cell patch-clamp and PLA2 activity was detected using 3H-arachidonic acid. S1P stimulated PLA2 and TRPC5 activities. Both effects were suppressed by the GVI PLA2 inhibitor bromoenol lactone. Knock-down of GVI PLA2 by RNA interference suppressed channel activity evoked by S1P whereas activity evoked by the direct channel stimulator LPC was unaffected. Arachidonic acid did not stimulate the channels. Prior exposure of channels to LPC but not arachidonic acid suppressed channel activity evoked by S1P but not gadolinium, a putative direct stimulator of the channels. The data suggest roles of LPC and GVI PLA2 in S1P-evoked TRPC5 activity.
Collapse
Affiliation(s)
- Eman AL-Shawaf
- Multidisciplinary Cardiovascular Research Centre and the Institute of Membrane & Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | |
Collapse
|
21
|
Bower DV, Sato Y, Lansford R. Dynamic lineage analysis of embryonic morphogenesis using transgenic quail and 4D multispectral imaging. Genesis 2011; 49:619-43. [PMID: 21509927 DOI: 10.1002/dvg.20754] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 03/27/2011] [Accepted: 03/28/2011] [Indexed: 12/17/2022]
Abstract
We describe the development of transgenic quail that express various fluorescent proteins in targeted manners and their use as a model system that integrates advanced imaging approaches with conventional and emerging molecular genetics technologies. We also review the progression and complications of past fate mapping techniques that led us to generate transgenic quail, which permit dynamic imaging of amniote embryogenesis with unprecedented subcellular resolution.
Collapse
Affiliation(s)
- Danielle V Bower
- Department of Biology and the Biological Imaging Center, California Institute of Technology, Pasadena, California 91125, USA
| | | | | |
Collapse
|
22
|
Majeed Y, Bahnasi Y, Seymour VAL, Wilson LA, Milligan CJ, Agarwal AK, Sukumar P, Naylor J, Beech DJ. Rapid and contrasting effects of rosiglitazone on transient receptor potential TRPM3 and TRPC5 channels. Mol Pharmacol 2011; 79:1023-30. [PMID: 21406603 PMCID: PMC3102547 DOI: 10.1124/mol.110.069922] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 03/15/2011] [Indexed: 01/16/2023] Open
Abstract
The aim of this study was to generate new insight into chemical regulation of transient receptor potential (TRP) channels with relevance to glucose homeostasis and the metabolic syndrome. Human TRP melastatin 2 (TRPM2), TRPM3, and TRP canonical 5 (TRPC5) were conditionally overexpressed in human embryonic kidney 293 cells and studied by using calcium-measurement and patch-clamp techniques. Rosiglitazone and other peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists were investigated. TRPM2 was unaffected by rosiglitazone at concentrations up to 10 μM but was inhibited completely at higher concentrations (IC(50), ∼22.5 μM). TRPM3 was more potently inhibited, with effects occurring in a biphasic concentration-dependent manner such that there was approximately 20% inhibition at low concentrations (0.1-1 μM) and full inhibition at higher concentrations (IC(50), 5-10 μM). PPAR-γ antagonism by 2-chloro-5-nitrobenzanilide (GW9662) did not prevent inhibition of TRPM3 by rosiglitazone. TRPC5 was strongly stimulated by rosiglitazone at concentrations of ≥10 μM (EC(50), ∼30 μM). Effects on TRPM3 and TRPC5 occurred rapidly and reversibly. Troglitazone and pioglitazone inhibited TRPM3 (IC(50), 12 μM) but lacked effect on TRPC5, suggesting no relevance of PPAR-γ or the thiazolidinedione moiety to rosiglitazone stimulation of TRPC5. A rosiglitazone-related but nonthiazolidinedione PPAR-γ agonist, N-(2-benzoylphenyl)-O-[2-(methyl-2-pyridinylamino)ethyl]-l-tyrosine (GW1929), was a weak stimulator of TRPM3 and TRPC5. The natural PPAR-γ agonist 15-deoxy prostaglandin J(2), had no effect on TRPM3 or TRPC5. The data suggest that rosiglitazone contains chemical moieties that rapidly, strongly, and differentially modulate TRP channels independently of PPAR-γ, potentially contributing to biological consequences of the agent and providing the basis for novel TRP channel pharmacology.
Collapse
Affiliation(s)
- Yasser Majeed
- Multidisciplinary Cardiovascular Research Centre and Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sperling SR. Systems biology approaches to heart development and congenital heart disease. Cardiovasc Res 2011; 91:269-78. [DOI: 10.1093/cvr/cvr126] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
24
|
Sarkey JP, Chu M, McShane M, Bovo E, Ait Mou Y, Zima AV, de Tombe PP, Kartje GL, Martin JL. Nogo-A knockdown inhibits hypoxia/reoxygenation-induced activation of mitochondrial-dependent apoptosis in cardiomyocytes. J Mol Cell Cardiol 2011; 50:1044-55. [PMID: 21420413 DOI: 10.1016/j.yjmcc.2011.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 03/11/2011] [Accepted: 03/11/2011] [Indexed: 01/15/2023]
Abstract
Programmed cell death of cardiomyocytes following myocardial ischemia increases biomechanical stress on the remaining myocardium, leading to myocardial dysfunction that may result in congestive heart failure or sudden death. Nogo-A is well characterized as a potent inhibitor of axonal regeneration and plasticity in the central nervous system, however, the role of Nogo-A in non-nervous tissues is essentially unknown. In this study, Nogo-A expression was shown to be significantly increased in cardiac tissue from patients with dilated cardiomyopathy and from patients who have experienced an ischemic event. Nogo-A expression was clearly associated with cardiomyocytes in culture and was localized predominantly in the endoplasmic reticulum. In agreement with the findings from human tissue, Nogo-A expression was significantly increased in cultured neonatal rat cardiomyocytes subjected to hypoxia/reoxygenation. Knockdown of Nogo-A in cardiomyocytes markedly attenuated hypoxia/reoxygenation-induced apoptosis, as indicated by the significant reduction of DNA fragmentation, phosphatidylserine translocation, and caspase-3 cleavage, by a mechanism involving the preservation of mitochondrial membrane potential, the inhibition of ROS accumulation, and the improvement of intracellular calcium regulation. Together, these data demonstrate that knockdown of Nogo-A may serve as a novel therapeutic strategy to prevent the loss of cardiomyocytes following ischemic/hypoxic injury.
Collapse
Affiliation(s)
- J P Sarkey
- Department of Cell and Molecular Physiology, Loyola University Medical Center, Maywood, IL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen Y, Zhou B, Li H, Peng Y, Wang Y, Rao L. Analysis of RTN4 3'UTR insertion/deletion polymorphisms in ventricular septal defect in a Chinese Han population. DNA Cell Biol 2010; 30:323-7. [PMID: 21166502 DOI: 10.1089/dna.2010.1116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Congenital heart disease is the most common type of birth defect and the leading cause of infant mortality in the first year of life. Ventricular septal defect (VSD) is one of the most general congenital heart defects and is a defect in the wall between the right and left ventricles of the heart. The pathogenesis of VSD has been extensively investigated for many years, but it remains uncertain. To determine whether reticulon 4 gene (RTN4) 3'UTR insertion/deletion polymorphisms are associated with VSD, we genotyped the TATC and CAA insertion/deletion polymorphisms of RTN4 by polymerase chain reaction-polyacrylamide gel electrophoresis in 151 VSD patients and 308 unrelated healthy subjects in a Chinese Han population. No significant differences in 3'UTR TATC and CAA insertion/deletion polymorphisms genotype and allele frequencies were observed between the VSD and controls. These data indicate that, for the first time, RTN4 3'UTR insertion/deletion polymorphisms may not appear to play a role in the susceptibility of VSD in Chinese Han population.
Collapse
Affiliation(s)
- Yu Chen
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, PR China
| | | | | | | | | | | |
Collapse
|
26
|
Naylor J, Al-Shawaf E, McKeown L, Manna PT, Porter KE, O'Regan D, Muraki K, Beech DJ. TRPC5 channel sensitivities to antioxidants and hydroxylated stilbenes. J Biol Chem 2010; 286:5078-86. [PMID: 21127073 PMCID: PMC3037619 DOI: 10.1074/jbc.m110.196956] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Transient receptor potential canonical 5 (TRPC5) forms cationic channels that are polymodal sensors of factors including oxidized phospholipids, hydrogen peroxide, and reduced thioredoxin. The aim of this study was to expand knowledge of the chemical-sensing capabilities of TRPC5 by investigating dietary antioxidants. Human TRPC5 channels were expressed in HEK 293 cells and studied by patch clamp and intracellular Ca(2+) recording. GFP- and HA-tagged channels were used to quantify plasma membrane localization. Gallic acid and vitamin C suppressed TRPC5 activity if it was evoked by exogenous hydrogen peroxide or lanthanide ions but not by lysophosphatidylcholine or carbachol. Catalase mimicked the effects, suggesting that lanthanide-evoked activity depended on endogenous hydrogen peroxide. Trans-resveratrol, by contrast, inhibited all modes of TRPC5, and its effect was additive with that of vitamin C, suggesting antioxidant-independent action. The IC(50) was ∼10 μM. Diethylstilbestrol, a related hydroxylated stilbene, inhibited TRPC5 with a similar IC(50), but its action contrasted sharply with that of resveratrol in outside-out membrane patches where diethylstilbestrol caused strong and reversible inhibition and resveratrol had no effect, suggesting indirect modulation by resveratrol. Resveratrol did not affect channel surface density, but its effect was calcium-sensitive, indicating an action via a calcium-dependent intermediate. The data suggest previously unrecognized chemical-sensing properties of TRPC5 through multiple mechanisms: (i) inhibition by scavengers of reactive oxygen species because a mode of TRPC5 activity depends on endogenous hydrogen peroxide; (ii) direct channel blockade by diethylstilbestrol; and (iii) indirect, antioxidant-independent inhibition by resveratrol.
Collapse
Affiliation(s)
- Jacqueline Naylor
- Multidisciplinary Cardiovascular Research Centre, Institute of Membrane and Systems Biology, Faculty of Biological Sciences, and Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Bon E, Steegers R, Steegers EAP, Ursem N, Charif H, Burgers PC, Luider TM, Dekker LJM. Proteomic analyses of the developing chicken cardiovascular system. J Proteome Res 2010; 9:268-74. [PMID: 19874049 DOI: 10.1021/pr900614w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Up until today, no proteomics approaches have been described for heart muscle development. We describe a proteomics method to study the proteome of different heart structures at three stages of chicken embryonic development. For this purpose, a combination of gel separation, nanoLC separation and mass spectrometry was used. With this method, we identified in total 267 proteins in different tissue structures of chicken heart. We observed differences in protein abundance for a number of proteins between the different tissue structures and time points of development using spectral counting as a semiquantitative measure of protein abundance. For myosin-heavy chain 6, myosin-heavy chain 7, titin, connectin, collagen alpha-1, and xin, differences in protein levels for the different stages and structures (great arteries, outflow tract and ventricles) have been observed. A pathway analysis is performed in which the identified proteins are related to theoretical protein networks. Most prominent was the 'cardiovascular system development and function' network with the abundantly present proteins myosin 6 and myosin 7. We showed that myosin 6 is highly regulated in a stage and heart tissue specific manner. In conclusion, this method can be used to study changes in protein levels of chicken heart tissue in a spatiotemporal manner.
Collapse
Affiliation(s)
- Els Bon
- Laboratories of Neuro-Oncology & Clinical and Cancer Proteomics, Department of Neurology, Erasmus MC, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Stimulation of TRPC5 cationic channels by low micromolar concentrations of lead ions (Pb2+). Biochem Biophys Res Commun 2010; 393:50-4. [PMID: 20100462 PMCID: PMC2877782 DOI: 10.1016/j.bbrc.2010.01.074] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 01/17/2010] [Indexed: 11/22/2022]
Abstract
Lead toxicity is long-recognised but continues to be a major public health problem. Its effects are wide-ranging and include induction of hyper-anxiety states. In general it is thought to act by interfering with Ca(2+) signalling but specific targets are not clearly identified. Transient receptor potential canonical 5 (TRPC5) is a Ca(2+)-permeable ion channel that is linked positively to innate fear responses and unusual amongst ion channels in being stimulated by trivalent lanthanides, which include gadolinium. Here we show investigation of the effect of lead, which is a divalent ion (Pb(2+)). Intracellular Ca(2+) and whole-cell patch-clamp recordings were performed on HEK 293 cells conditionally over-expressing TRPC5 or other TRP channels. Extracellular application of Pb(2+) stimulated TRPC5 at concentrations greater than 1 microM. Control cells without TRPC5 showed little or no response to Pb(2+) and expression of other TRP channels (TRPM2 or TRPM3) revealed partial inhibition by 10 microM Pb(2+). The stimulatory effect on TRPC5 depended on an extracellular residue (E543) near the ion pore: similar to gadolinium action, E543Q TRPC5 was resistant to Pb(2+) but showed normal stimulation by the receptor agonist sphingosine-1-phosphate. The study shows that Pb(2+) is a relatively potent stimulator of the TRPC5 channel, generating the hypothesis that a function of the channel is to sense metal ion poisoning.
Collapse
|
29
|
Industrialized MS-based proteomics in the search for circulating biomarkers. Bioanalysis 2009; 1:1149-63. [DOI: 10.4155/bio.09.105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Proteomics is the study of the expression, structure and function of proteins under a range of cellular conditions. A rapidly evolving component of this field is clinical proteomics, which focuses on proteins involved in human disease and how they are affected by therapeutic intervention. MS is the main analytical technology for identifying and quantifying proteins whose expression is modulated across the normal to disease continuum. Applying this technology to clinical samples, however, is particularly challenging due to high biological variability in the population, a variety of disease stages, nonuniform response to therapy, multiple concomitant treatments and special requirements for handling samples from clinical trials. Given these challenges, an ‘industrialized’ approach is best suited to clinical biomarker development, with its standard operating procedures, process control and ‘chain of custody’. This review will focus, therefore, on MS-based industrialized proteomics for the discovery and verification of circulating candidate clinical protein biomarkers.
Collapse
|