1
|
Liu Q, Xu C, Jin J, Li W, Liang J, Zhou S, Weng Z, Zhou Y, Liao X, Gu A. Early-life exposure to lead changes cardiac development and compromises long-term cardiac function. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 904:166667. [PMID: 37652374 DOI: 10.1016/j.scitotenv.2023.166667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/17/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Lead (Pb) is widely used in industrial and daily-use consumer products. Early-life exposure may increase the risk of lead-related heart problems in childhood. However, the effects of early-life lead exposure on fetal heart development and long-term cardiac outcomes are unknown. In this study, pregnant ICR mice were exposed to lead acetate trihydrate (50 mg/kg/d) via oral gavage from gestation day 1.5 until offspring weaning. Thereafter, the second hit model was established, two groups of offspring (4 weeks old) were either administered sterile saline or Angiotensin II (Ang II) for 4 weeks until euthanasia. We investigated lead-induced offspring heart damage from embryonic period to adulthood by echocardiographic analysis, pathological H&E staining, and ultrastructural examination, as well as mitochondrial function detection. The results showed early-life lead exposure predisposed offspring mice to decreased ejection fraction, increased left ventricular volume, accompanied by hypertrophy and dilation, cardiomyocyte sarcomere dysplasia, abnormal mitochondrial structure, mitochondrial dysfunction, and decreased expression of key sarcomeric and mitochondrial genes, rendering them more susceptible to cardiac hypertrophy, vascular wall thickening, cardiac fibrosis, apoptosis, and heart failure induced by Ang II infusion. This study elucidates early-life low dose lead exposure compromises cardiac development and exacerbates second hit-induced cardiac pathological responses in adulthood, which furnishes crucial scientific evidence pertaining to the cardiac toxicity and risk evaluation associated with early-life exposure to lead.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Cheng Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jing Jin
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxiang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jingjia Liang
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Shijie Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Zhenkun Weng
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yong Zhou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Xudong Liao
- College of Life Sciences, Nankai University, Tianjin, China.
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Kim J, Lee C, Han J, Jeong H, Wang S, Choi YH, Jung Y. Targeted Deletion of Thymosin Beta 4 in Hepatic Stellate Cells Ameliorates Liver Fibrosis in a Transgenic Mouse Model. Cells 2023; 12:1658. [PMID: 37371128 DOI: 10.3390/cells12121658] [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: 04/28/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Liver fibrosis is the most common feature of liver disease, and activated hepatic stellate cells (HSCs) are the main contributors to liver fibrosis. Thus, finding key targets that modulate HSC activation is important to prevent liver fibrosis. Previously, we showed that thymosin β4 (Tβ4) influenced HSC activation by interacting with the Hedgehog pathway in vitro. Herein, we generated Tβ4 conditional knockout (Tβ4-flox) mice to investigate in vivo functions of Tβ4 in liver fibrosis. To selectively delete Tβ4 in activated HSCs, double-transgenic (DTG) mice were generated by mating Tβ4-flox mice with α-smooth muscle actin (α-Sma)-Cre-ERT2 mice, and these mice were administered carbon tetrachloride (CCl4) or underwent bile duct ligation to induce liver fibrosis. Tβ4 was selectively suppressed in the activated HSCs of DTG mouse liver, and this reduction attenuated liver injury, including fibrosis, in both fibrotic models by repressing Hedgehog (Hh) signaling. In addition, the re-expression of Tβ4 by an adeno-associated virus reversed the effect of HSC-specific Tβ4 deletion and led to liver fibrosis with Hh activation in CCl4-exposed mice treated with tamoxifen. In conclusion, our results demonstrate that Tβ4 is a crucial regulator of HSC activation, suggesting it as a novel therapeutic target for curing liver fibrosis.
Collapse
Affiliation(s)
- Jieun Kim
- Institute of System Biology, Pusan National University, Pusan 46241, Republic of Korea
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Chanbin Lee
- Institute of System Biology, Pusan National University, Pusan 46241, Republic of Korea
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Jinsol Han
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Hayeong Jeong
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Sihyung Wang
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-Eui University College of Korean Medicine, Pusan 47227, Republic of Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| |
Collapse
|
3
|
Yadunandanan Nair N, Samuel V, Ramesh L, Marib A, David DT, Sundararaman A. Actin cytoskeleton in angiogenesis. Biol Open 2022; 11:bio058899. [PMID: 36444960 PMCID: PMC9729668 DOI: 10.1242/bio.058899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Actin, one of the most abundant intracellular proteins in mammalian cells, is a critical regulator of cell shape and polarity, migration, cell division, and transcriptional response. Angiogenesis, or the formation of new blood vessels in the body is a well-coordinated multi-step process. Endothelial cells lining the blood vessels acquire several new properties such as front-rear polarity, invasiveness, rapid proliferation and motility during angiogenesis. This is achieved by changes in the regulation of the actin cytoskeleton. Actin remodelling underlies the switch between the quiescent and angiogenic state of the endothelium. Actin forms endothelium-specific structures that support uniquely endothelial functions. Actin regulators at endothelial cell-cell junctions maintain the integrity of the blood-tissue barrier while permitting trans-endothelial leukocyte migration. This review focuses on endothelial actin structures and less-recognised actin-mediated endothelial functions. Readers are referred to other recent reviews for the well-recognised roles of actin in endothelial motility, barrier functions and leukocyte transmigration. Actin generates forces that are transmitted to the extracellular matrix resulting in vascular matrix remodelling. In this review, we attempt to synthesize our current understanding of the roles of actin in vascular morphogenesis. We speculate on the vascular bed specific differences in endothelial actin regulation and its role in the vast heterogeneity in endothelial morphology and function across the various tissues of our body.
Collapse
Affiliation(s)
- Nidhi Yadunandanan Nair
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Victor Samuel
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Lariza Ramesh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Areeba Marib
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Deena T. David
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| |
Collapse
|
4
|
Parvin S, Arabfard M, Ghazvini A, Ghanei M, Najafi A. Comparative proteomic analysis of mustard lung as a complicated disease using systems biology approach. BMC Pulm Med 2022; 22:437. [PMID: 36419000 PMCID: PMC9686120 DOI: 10.1186/s12890-022-02240-3] [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: 07/25/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
During Iraq-Iran conflict, chemical weapons, particularly SM gas, were used numerous times, whose aftereffects are still present. This study aimed to compare serum proteome in the chronic ML (n = 10) and HC (n = 10). TMT label-based quantitative proteomics was used to examine serums from two groups. Among total significant proteins, 14 proteins were upregulated (log2 ≥ FC 0.5, p 0.05), and 6 proteins were downregulated (log2 ≤ FC - 0.5, p 0.05). By helping PPI network, and EA, 11 main pathways connected to significantly different protein expression levels were discovered, including inflammatory and cell adhesion signaling pathways. It may be deduced that the wounded organs of exposed individuals experience poor repair cycles of cell degeneration and regeneration because certain repair signals were elevated while other structural and adhesion molecules were downregulated. The systems biology approach can help enhance our basic knowledge of biological processes, and contribute to a deeper understanding of pathophysiological mechanisms, as well as the identification of potential biomarkers of disease.
Collapse
Affiliation(s)
- Shahram Parvin
- grid.420169.80000 0000 9562 2611Education Office, Pasteur Institute of Iran, Tehran, Iran
| | - Masoud Arabfard
- grid.411521.20000 0000 9975 294XChemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ghazvini
- grid.411521.20000 0000 9975 294XChemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- grid.411521.20000 0000 9975 294XChemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Najafi
- grid.411521.20000 0000 9975 294XMolecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Streef TJ, Smits AM. Epicardial Contribution to the Developing and Injured Heart: Exploring the Cellular Composition of the Epicardium. Front Cardiovasc Med 2021; 8:750243. [PMID: 34631842 PMCID: PMC8494983 DOI: 10.3389/fcvm.2021.750243] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The epicardium is an essential cell population during cardiac development. It contributes different cell types to the developing heart through epithelial-to-mesenchymal transition (EMT) and it secretes paracrine factors that support cardiac tissue formation. In the adult heart the epicardium is a quiescent layer of cells which can be reactivated upon ischemic injury, initiating an embryonic-like response in the epicardium that contributes to post-injury repair processes. Therefore, the epicardial layer is considered an interesting target population to stimulate endogenous repair mechanisms. To date it is still not clear whether there are distinct cell populations in the epicardium that contribute to specific lineages or aid in cardiac repair, or that the epicardium functions as a whole. To address this putative heterogeneity, novel techniques such as single cell RNA sequencing (scRNA seq) are being applied. In this review, we summarize the role of the epicardium during development and after injury and provide an overview of the most recent insights into the cellular composition and diversity of the epicardium.
Collapse
Affiliation(s)
| | - Anke M. Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
6
|
Utilizing Developmentally Essential Secreted Peptides Such as Thymosin Beta-4 to Remind the Adult Organs of Their Embryonic State-New Directions in Anti-Aging Regenerative Therapies. Cells 2021; 10:cells10061343. [PMID: 34071596 PMCID: PMC8228050 DOI: 10.3390/cells10061343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/20/2021] [Indexed: 01/10/2023] Open
Abstract
Our dream of defeating the processes of aging has occupied the curious and has challenged scientists globally for hundreds of years. The history is long, and sadly, the solution is still elusive. Our endeavors to reverse the magnitude of damaging cellular and molecular alterations resulted in only a few, yet significant advancements. Furthermore, as our lifespan increases, physicians are facing more mind-bending questions in their routine practice than ever before. Although the ultimate goal is to successfully treat the body as a whole, steps towards regenerating individual organs are even considered significant. As our initial approach to enhance the endogenous restorative capacity by delivering exogenous progenitor cells appears limited, we propose, utilizing small molecules critical during embryonic development may prove to be a powerful tool to increase regeneration and to reverse the processes associated with aging. In this review, we introduce Thymosin beta-4, a 43aa secreted peptide fulfilling our hopes and capable of numerous regenerative achievements via systemic administration in the heart. Observing the broad capacity of this small, secreted peptide, we believe it is not the only molecule which nature conceals to our benefit. Hence, the discovery and postnatal administration of developmentally relevant agents along with other approaches may result in reversing the aging process.
Collapse
|
7
|
Mantri M, Scuderi GJ, Abedini-Nassab R, Wang MFZ, McKellar D, Shi H, Grodner B, Butcher JT, De Vlaminck I. Spatiotemporal single-cell RNA sequencing of developing chicken hearts identifies interplay between cellular differentiation and morphogenesis. Nat Commun 2021; 12:1771. [PMID: 33741943 PMCID: PMC7979764 DOI: 10.1038/s41467-021-21892-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 02/17/2021] [Indexed: 01/01/2023] Open
Abstract
Single-cell RNA sequencing is a powerful tool to study developmental biology but does not preserve spatial information about tissue morphology and cellular interactions. Here, we combine single-cell and spatial transcriptomics with algorithms for data integration to study the development of the chicken heart from the early to late four-chambered heart stage. We create a census of the diverse cellular lineages in developing hearts, their spatial organization, and their interactions during development. Spatial mapping of differentiation transitions in cardiac lineages defines transcriptional differences between epithelial and mesenchymal cells within the epicardial lineage. Using spatially resolved expression analysis, we identify anatomically restricted expression programs, including expression of genes implicated in congenital heart disease. Last, we discover a persistent enrichment of the small, secreted peptide, thymosin beta-4, throughout coronary vascular development. Overall, our study identifies an intricate interplay between cellular differentiation and morphogenesis.
Collapse
Affiliation(s)
- Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Computational Biology Ph.D. Program, Cornell University, Ithaca, NY, USA
| | - Gaetano J Scuderi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Roozbeh Abedini-Nassab
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Engineering, University of Neyshabur, Neyshabur, Iran
| | - Michael F Z Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Computational Biology Ph.D. Program, Cornell University, Ithaca, NY, USA
| | - David McKellar
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Hao Shi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Benjamin Grodner
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jonathan T Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
8
|
Dronkers E, Wauters MMM, Goumans MJ, Smits AM. Epicardial TGFβ and BMP Signaling in Cardiac Regeneration: What Lesson Can We Learn from the Developing Heart? Biomolecules 2020; 10:biom10030404. [PMID: 32150964 PMCID: PMC7175296 DOI: 10.3390/biom10030404] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/31/2022] Open
Abstract
The epicardium, the outer layer of the heart, has been of interest in cardiac research due to its vital role in the developing and diseased heart. During development, epicardial cells are active and supply cells and paracrine cues to the myocardium. In the injured adult heart, the epicardium is re-activated and recapitulates embryonic behavior that is essential for a proper repair response. Two indispensable processes for epicardial contribution to heart tissue formation are epithelial to mesenchymal transition (EMT), and tissue invasion. One of the key groups of cytokines regulating both EMT and invasion is the transforming growth factor β (TGFβ) family, including TGFβ and Bone Morphogenetic Protein (BMP). Abundant research has been performed to understand the role of TGFβ family signaling in the developing epicardium. However, less is known about signaling in the adult epicardium. This review provides an overview of the current knowledge on the role of TGFβ in epicardial behavior both in the development and in the repair of the heart. We aim to describe the presence of involved ligands and receptors to establish if and when signaling can occur. Finally, we discuss potential targets to improve the epicardial contribution to cardiac repair as a starting point for future investigation.
Collapse
|
9
|
Role of complement 3 in the pathogenesis of hypertension. Hypertens Res 2019; 43:255-262. [DOI: 10.1038/s41440-019-0371-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
|
10
|
Lange S, Banerjee I, Carrion K, Serrano R, Habich L, Kameny R, Lengenfelder L, Dalton N, Meili R, Börgeson E, Peterson K, Ricci M, Lincoln J, Ghassemian M, Fineman J, del Álamo JC, Nigam V. miR-486 is modulated by stretch and increases ventricular growth. JCI Insight 2019; 4:125507. [PMID: 31513548 PMCID: PMC6795397 DOI: 10.1172/jci.insight.125507] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Perturbations in biomechanical stimuli during cardiac development contribute to congenital cardiac defects such as hypoplastic left heart syndrome (HLHS). This study sought to identify stretch-responsive pathways involved in cardiac development. miRNA-Seq identified miR-486 as being increased in cardiomyocytes exposed to cyclic stretch in vitro. The right ventricles (RVs) of patients with HLHS experienced increased stretch and had a trend toward higher miR-486 levels. Sheep RVs dilated from excessive pulmonary blood flow had 60% more miR-486 compared with control RVs. The left ventricles of newborn mice treated with miR-486 mimic were 16.9%-24.6% larger and displayed a 2.48-fold increase in cardiomyocyte proliferation. miR-486 treatment decreased FoxO1 and Smad signaling while increasing the protein levels of Stat1. Stat1 associated with Gata-4 and serum response factor (Srf), 2 key cardiac transcription factors with protein levels that increase in response to miR-486. This is the first report to our knowledge of a stretch-responsive miRNA that increases the growth of the ventricle in vivo.
Collapse
Affiliation(s)
- Stephan Lange
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
- Institute of Medicine, Department of Molecular and Clinical Medicine, the Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Indroneal Banerjee
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Katrina Carrion
- Division of Cardiology, Department of Pediatrics, UCSD School of Medicine, San Diego, California, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Louisa Habich
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Rebecca Kameny
- Department of Pediatrics, UCSF School of Medicine, San Francisco, USA
| | - Luisa Lengenfelder
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Nancy Dalton
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Rudolph Meili
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Emma Börgeson
- Institute of Medicine, Department of Molecular and Clinical Medicine, the Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kirk Peterson
- Division of Cardiovascular Medicine, Department of Medicine, UCSD School of Medicine, San Diego, California, USA
| | - Marco Ricci
- Division of Cardiothoracic Surgery and
- Division of Pediatric Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Joy Lincoln
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | | | - Jeffery Fineman
- Department of Pediatrics, UCSF School of Medicine, San Francisco, USA
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, UCSD, San Diego, USA
| | - Vishal Nigam
- Division of Cardiology, Department of Pediatrics, UCSD School of Medicine, San Diego, California, USA
- Division of Cardiology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
11
|
Kassem KM, Vaid S, Peng H, Sarkar S, Rhaleb NE. Tβ4-Ac-SDKP pathway: Any relevance for the cardiovascular system? Can J Physiol Pharmacol 2019; 97:589-599. [PMID: 30854877 PMCID: PMC6824425 DOI: 10.1139/cjpp-2018-0570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The last 20 years witnessed the emergence of the thymosin β4 (Tβ4)-N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) pathway as a new source of future therapeutic tools to treat cardiovascular and renal diseases. In this review article, we attempted to shed light on the numerous experimental findings pertaining to the many promising cardiovascular therapeutic avenues for Tβ4 and (or) its N-terminal derivative, Ac-SDKP. Specifically, Ac-SDKP is endogenously produced from the 43-amino acid Tβ4 by 2 successive enzymes, meprin α and prolyl oligopeptidase. We also discussed the possible mechanisms involved in the Tβ4-Ac-SDKP-associated cardiovascular biological effects. In infarcted myocardium, Tβ4 and Ac-SDKP facilitate cardiac repair after infarction by promoting endothelial cell migration and myocyte survival. Additionally, Tβ4 and Ac-SDKP have antifibrotic and anti-inflammatory properties in the arteries, heart, lungs, and kidneys, and stimulate both in vitro and in vivo angiogenesis. The effects of Tβ4 can be mediated directly through a putative receptor (Ku80) or via its enzymatically released N-terminal derivative Ac-SDKP. Despite the localization and characterization of Ac-SDKP binding sites in myocardium, more studies are needed to fully identify and clone Ac-SDKP receptors. It remains promising that Ac-SDKP or its degradation-resistant analogs could serve as new therapeutic tools to treat cardiac, vascular, and renal injury and dysfunction to be used alone or in combination with the already established pharmacotherapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Kamal M Kassem
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- b Internal Medicine Department, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA
| | - Sonal Vaid
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- c Internal Medicine Department, St. Vincent Indianapolis Hospital, Indianapolis, IN 46260, USA
| | - Hongmei Peng
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Sarah Sarkar
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Nour-Eddine Rhaleb
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- d Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
12
|
Saunders V, Dewing JM, Sanchez-Elsner T, Wilson DI. Expression and localisation of thymosin beta-4 in the developing human early fetal heart. PLoS One 2018; 13:e0207248. [PMID: 30412598 PMCID: PMC6226193 DOI: 10.1371/journal.pone.0207248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/26/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The objective of this study was to investigate the expression and localisation of thymosin β4 (Tβ4) in the developing human heart. Tβ4 is a cardioprotective protein which may have therapeutic potential. While Tβ4 is an endogenously produced protein with known importance during development, its role within the developing human heart is not fully understood. Elucidating the localisation of Tβ4 within the developing heart will help in understanding its role during cardiac development and is crucial for understanding its potential for cardioprotection and repair in the adult heart. METHODS Expression of Tβ4 mRNA in the early fetal human heart was assessed by PCR using both ventricular and atrial tissue. Fluorescence immunohistochemistry was used to assess the localisation of Tβ4 in sections of early fetal human heart. Co-staining with CD31, an endothelial cell marker, and with myosin heavy chain, a cardiomyocyte marker, was used to determine whether Tβ4 is localised to these cell types within the early fetal human heart. RESULTS Tβ4 mRNA was found to be expressed in both the atria and the ventricles of the early fetal human heart. Tβ4 protein was found to be primarily localised to CD31-expressing endothelial cells and the endocardium as well as being present in the epicardium. Tβ4-associated fluorescence was greater in the compact layer of the myocardial wall and the interventricular septum than in the trabecular layer of the myocardium. CONCLUSIONS The data presented illustrates expression of Tβ4 in the developing human heart and demonstrates for the first time that Tβ4 in the human heart is primarily localised to endothelial cells of the cardiac microvasculature and coronary vessels as-well as to the endothelial-like cells of the endocardium and to the epicardium.
Collapse
Affiliation(s)
- Vinay Saunders
- Institute for Developmental Science, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jennifer M. Dewing
- Institute for Developmental Science, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tilman Sanchez-Elsner
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - David I. Wilson
- Institute for Developmental Science, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Dubé KN, Smart N. Thymosin β4 and the vasculature: multiple roles in development, repair and protection against disease. Expert Opin Biol Ther 2018; 18:131-139. [DOI: 10.1080/14712598.2018.1459558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Karina N. Dubé
- BHF Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- BHF Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
Abstract
Death of adult cardiac myocytes and supportive tissues resulting from cardiovascular diseases such as myocardial infarction is the proximal driver of pathological ventricular remodeling that often culminates in heart failure. Unfortunately, no currently available therapeutic barring heart transplantation can directly replenish myocytes lost from the injured heart. For decades, the field has struggled to define the intrinsic capacity and cellular sources for endogenous myocyte turnover in pursuing more innovative therapeutic strategies aimed at regenerating the injured heart. Although controversy persists to this day as to the best therapeutic regenerative strategy to use, a growing consensus has been reached that the very limited capacity for new myocyte formation in the adult mammalian heart is because of proliferation of existing cardiac myocytes but not because of the activity of an endogenous progenitor cell source of some sort. Hence, future therapeutic approaches should take into consideration the fundamental biology of myocyte renewal in designing strategies to potentially replenish these cells in the injured heart.
Collapse
Affiliation(s)
| | - Jeffery D Molkentin
- From the Department of Pediatrics (R.J.V., J.D.M.)
- Howard Hughes Medical Institute (J.D.M.)
| | - Steven R Houser
- Cincinnati Children's Hospital Medical Center, OH; and the Lewis Katz School of Medicine, Cardiovascular Research Center, Temple University, Philadelphia, PA (S.R.H.)
| |
Collapse
|
15
|
Ehler E. Actin-associated proteins and cardiomyopathy-the 'unknown' beyond troponin and tropomyosin. Biophys Rev 2018; 10:1121-1128. [PMID: 29869751 PMCID: PMC6082317 DOI: 10.1007/s12551-018-0428-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
It has been known for several decades that mutations in genes that encode for proteins involved in the control of actomyosin interactions such as the troponin complex, tropomyosin and MYBP-C and thus regulate contraction can lead to hereditary hypertrophic cardiomyopathy. In recent years, it has become apparent that actin-binding proteins not directly involved in the regulation of contraction also can exhibit changed expression levels, show altered subcellular localisation or bear mutations that might lead to hereditary cardiomyopathies. The aim of this review is to look beyond the troponin/tropomyosin mechanism and to give an overview of the different types of actin-associated proteins and their potential roles in cardiomyocytes. It will then discuss recent findings relevant to their involvement in heart disease.
Collapse
Affiliation(s)
- Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences), London, UK. .,School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, Room 3.26A, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
16
|
Weinberger F, Nicol P, Starbatty J, Stubbendorff M, Becher PM, Schrepfer S, Eschenhagen T. No effect of thymosin beta-4 on the expression of the transcription factor Islet-1 in the adult murine heart. Pharmacol Res Perspect 2018; 6:e00407. [PMID: 29864245 PMCID: PMC5986028 DOI: 10.1002/prp2.407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 01/14/2023] Open
Abstract
The transcription factor Islet-1 marks a progenitor cell population of the second heart field during cardiogenesis. In the adult heart Islet-1 expression is limited to the sinoatrial node, the ventricular outflow tract, and parasympathetic ganglia. The regenerative effect in the injured mouse ventricle of thymosin beta-4 (TB4), a 43-aminoacid peptide, was associated with increased Islet-1 immunostaining, suggesting the induction of an Islet-1-positive progenitor state by TB4. Here we aimed to reassess this effect in a genetic model. Mice from the reporter mouse line Isl1-nLacZ were primed with TB4 and subsequently underwent myocardial infarction. Islet-1 expression was assessed 2, 7, and 14 days after infarction. We detected only a single Islet-1+ cell in 8 TB4 treated and infarcted hearts which located outside of the sinoatrial node, the outflow tract or cardiac ganglia (in ~2500 sections). Two cells were identified in 5 control infarcted hearts. TB4 did not induce LacZ positivity in ventricular explants cultures of Isl1-nLacZ mice nor did it affect the density of LacZ+ cells in explant cultures of nLacZ+ regions of the heart. In summary, we found no evidence that TB4 reactivates Islet-1 expression in adult mouse ventricle.
Collapse
Affiliation(s)
- Florian Weinberger
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
| | - Philipp Nicol
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
| | - Jutta Starbatty
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
| | - Mandy Stubbendorff
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
- Department of Cardiovascular Surgery, Transplant and Stem Cell Immunobiology (TSI) LabUniversity Heart Center HamburgHamburgGermany
| | - Peter M. Becher
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
- Department of General and Interventional CardiologyUniversity Heart Center HamburgHamburgGermany
| | - Sonja Schrepfer
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
- Department of Cardiovascular Surgery, Transplant and Stem Cell Immunobiology (TSI) LabUniversity Heart Center HamburgHamburgGermany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK, German Center for Cardiovascular Researchpartner site Hamburg/Kiel/LübeckGermany
| |
Collapse
|
17
|
Abstract
Despite therapeutic advances that have prolonged life, myocardial infarction (MI) remains a leading cause of death worldwide and imparts a significant economic burden. The advancement of treatments to improve cardiac repair post-MI requires the discovery of new targeted treatment strategies. Recent studies have highlighted the importance of the epicardial covering of the heart in both cardiac development and lower vertebrate cardiac regeneration. The epicardium serves as a source of cardiac cells including smooth muscle cells, endothelial cells and cardiac fibroblasts. Mammalian adult epicardial cells are typically quiescent. However, the fetal genetic program is reactivated post-MI, and epicardial epithelial-to-mesenchymal transition (EMT) occurs as an inherent mechanism to support neovascularization and cardiac healing. Unfortunately, endogenous EMT is not enough to encourage sufficient repair. Recent developments in our understanding of the mechanisms supporting the EMT process has led to a number of studies directed at augmenting epicardial EMT post-MI. With a focus on the role of the primary cilium, this review outlines the newly demonstrated mechanisms supporting EMT, the role of epicardial EMT in cardiac development, and promising advances in augmenting epicardial EMT as potential therapeutics to support cardiac repair post-MI.
Collapse
|
18
|
Smart N, Riegler J, Turtle CW, Lygate CA, McAndrew DJ, Gehmlich K, Dubé KN, Price AN, Muthurangu V, Taylor AM, Lythgoe MF, Redwood C, Riley PR. Aberrant developmental titin splicing and dysregulated sarcomere length in Thymosin β4 knockout mice. J Mol Cell Cardiol 2017; 102:94-107. [PMID: 27914791 PMCID: PMC5319848 DOI: 10.1016/j.yjmcc.2016.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 02/07/2023]
Abstract
Sarcomere assembly is a highly orchestrated and dynamic process which adapts, during perinatal development, to accommodate growth of the heart. Sarcomeric components, including titin, undergo an isoform transition to adjust ventricular filling. Many sarcomeric genes have been implicated in congenital cardiomyopathies, such that understanding developmental sarcomere transitions will inform the aetiology and treatment. We sought to determine whether Thymosin β4 (Tβ4), a peptide that regulates the availability of actin monomers for polymerization in non-muscle cells, plays a role in sarcomere assembly during cardiac morphogenesis and influences adult cardiac function. In Tβ4 null mice, immunofluorescence-based sarcomere analyses revealed shortened thin filament, sarcomere and titin spring length in cardiomyocytes, associated with precocious up-regulation of the short titin isoforms during the postnatal splicing transition. By magnetic resonance imaging, this manifested as diminished stroke volume and limited contractile reserve in adult mice. Extrapolating to an in vitro cardiomyocyte model, the altered postnatal splicing was corrected with addition of synthetic Tβ4, whereby normal sarcomere length was restored. Our data suggest that Tβ4 is required for setting correct sarcomere length and for appropriate splicing of titin, not only in the heart but also in skeletal muscle. Distinguishing between thin filament extension and titin splicing as the primary defect is challenging, as these events are intimately linked. The regulation of titin splicing is a previously unrecognised role of Tβ4 and gives preliminary insight into a mechanism by which titin isoforms may be manipulated to correct cardiac dysfunction.
Collapse
Affiliation(s)
- Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Johannes Riegler
- Centre for Advanced Biomedical Imaging, Department of Medicine, University College London (UCL), London, UK
| | - Cameron W Turtle
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Anthony N Price
- Centre for Advanced Biomedical Imaging, Department of Medicine, University College London (UCL), London, UK
| | - Vivek Muthurangu
- Centre for Cardiovascular Imaging, UCL Institute of Cardiovascular Science, London, UK
| | - Andrew M Taylor
- Centre for Cardiovascular Imaging, UCL Institute of Cardiovascular Science, London, UK
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Department of Medicine, University College London (UCL), London, UK
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Thymosin β4 impeded murine stem cell proliferation with an intact cardiovascular differentiation. ACTA ACUST UNITED AC 2016; 36:328-334. [PMID: 27376799 DOI: 10.1007/s11596-016-1587-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Thymosin β4 (Tβ4) is a key factor in cardiac development, growth, disease, epicardial integrity, blood vessel formation and has cardio-protective properties. However, its role in murine embryonic stem cells (mESCs) proliferation and cardiovascular differentiation remains unclear. Thus we aimed to elucidate the influence of Tβ4 on mESCs. Target genes during mESCs proliferation and differentiation were detected by real-time PCR or Western blotting, and patch clamp was applied to characterize the mESCs-derived cardiomyocytes. It was found that Tβ4 decreased mESCs proliferation in a partial dose-dependent manner and the expression of cell cycle regulatory genes c-myc, c-fos and c-jun. However, mESCs self-renewal markers Oct4 and Nanog were elevated, indicating the maintenance of self-renewal ability in these mESCs. Phosphorylation of STAT3 and Akt was inhibited by Tβ4 while the expression of RAS and phosphorylation of ERK were enhanced. No significant difference was found in BMP2/BMP4 or their downstream protein smad. Wnt3 and Wnt11 were remarkably decreased by Tβ4 with upregulation of Tcf3 and constant β-catenin. Under mESCs differentiation, Tβ4 treatment did not change the expression of cardiovascular cell markers α-MHC, PECAM, and α-SMA. Neither the electrophysiological properties of mESCs-derived cardiomyocytes nor the hormonal regulation by Iso/Cch was affected by Tβ4. In conclusion, Tβ4 suppressed mESCs proliferation by affecting the activity of STAT3, Akt, ERK and Wnt pathways. However, Tβ4 did not influence the in vitro cardiovascular differentiation.
Collapse
|
20
|
Marks ED, Kumar A. Thymosin β4: Roles in Development, Repair, and Engineering of the Cardiovascular System. VITAMINS AND HORMONES 2016; 102:227-49. [PMID: 27450737 DOI: 10.1016/bs.vh.2016.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The burden of cardiovascular disease is a growing worldwide issue that demands attention. While many clinical trials are ongoing to test therapies for treating the heart after myocardial infarction (MI) and heart failure, there are few options doctors able to currently give patients to repair the heart. This eventually leads to decreased ventricular contractility and increased systemic disease, including vascular disorders that could result in stroke. Small peptides such as thymosin β4 (Tβ4) are upregulated in the cardiovascular niche during fetal development and after injuries such as MI, providing increased neovasculogenesis and paracrine signals for endogenous stem cell recruitment to aid in wound repair. New research is looking into the effects of in vivo administration of Tβ4 through injections and coatings on implants, as well as its effect on cell differentiation. Results so far demonstrate Tβ4 administration leads to robust increases in angiogenesis and wound healing in the heart after MI and the brain after stroke, and can differentiate adult stem cells toward the cardiac lineage for implantation to the heart to increase contractility and survival. Future work, some of which is currently in clinical trials, will demonstrate the in vivo effect of these therapies on human patients, with the goal of helping the millions of people worldwide affected by cardiovascular disease.
Collapse
Affiliation(s)
- E D Marks
- Nanomedicine Research Laboratory, University of Delaware, Newark, DE, United States
| | - A Kumar
- Nanomedicine Research Laboratory, University of Delaware, Newark, DE, United States.
| |
Collapse
|
21
|
Stark CKJ, Tarkia M, Kentala R, Malmberg M, Vähäsilta T, Savo M, Hynninen VV, Helenius M, Ruohonen S, Jalkanen J, Taimen P, Alastalo TP, Saraste A, Knuuti J, Savunen T, Koskenvuo J. Systemic Dosing of Thymosin Beta 4 before and after Ischemia Does Not Attenuate Global Myocardial Ischemia-Reperfusion Injury in Pigs. Front Pharmacol 2016; 7:115. [PMID: 27199757 PMCID: PMC4853610 DOI: 10.3389/fphar.2016.00115] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
The use of cardiopulmonary bypass (CPB) and aortic cross-clamping causes myocardial ischemia-reperfusion injury (I-RI) and can lead to reduced postoperative cardiac function. We investigated whether this injury could be attenuated by thymosin beta 4 (TB4), a peptide which has showed cardioprotective effects. Pigs received either TB4 or vehicle and underwent CPB and aortic cross-clamping for 60 min with cold intermittent blood-cardioplegia and were then followed for 30 h. Myocardial function and blood flow was studied by cardiac magnetic resonance and PET imaging. Tissue and plasma samples were analyzed to determine the amount of cardiomyocyte necrosis and apoptosis as well as pharmacokinetics of the peptide. In vitro studies were performed to assess its influence on blood coagulation and vasomotor tone. Serum levels of the peptide were increased after administration compared to control samples. TB4 did not decrease the amount of cell death. Cardiac function and global myocardial blood flow was similar between the study groups. At high doses a vasoconstrictor effect on mesentery arteries and a vasodilator effect on coronary arteries was observed and blood clot firmness was reduced when tested in the presence of an antiplatelet agent. Despite promising results in previous trials the cardioprotective effect of TB4 was not demonstrated in this model for global myocardial I-RI.
Collapse
Affiliation(s)
- Christoffer K-J Stark
- Research Center of Applied and Preventive Cardiovascular Medicine, University of TurkuTurku, Finland; Heart Center, Turku University Hospital and University of TurkuTurku, Finland
| | - Miikka Tarkia
- Turku PET Centre, Turku University Hospital and University of Turku Turku, Finland
| | - Rasmus Kentala
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| | - Markus Malmberg
- Heart Center, Turku University Hospital and University of Turku Turku, Finland
| | - Tommi Vähäsilta
- Research Center of Applied and Preventive Cardiovascular Medicine, University of TurkuTurku, Finland; Heart Center, Turku University Hospital and University of TurkuTurku, Finland
| | - Matti Savo
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| | - Ville-Veikko Hynninen
- Department of Anesthesiology, Intensive Care, Emergency Care and Pain Medicine, Turku University Hospital Turku, Finland
| | - Mikko Helenius
- Children's Hospital, Pediatric Cardiology, Helsinki University Hospital Helsinki, Finland
| | - Saku Ruohonen
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| | - Juho Jalkanen
- Department of Vascular Surgery, Turku University Hospital and University of Turku Turku, Finland
| | - Pekka Taimen
- Department of Pathology, Turku University Hospital and University of Turku Turku, Finland
| | - Tero-Pekka Alastalo
- Children's Hospital, Pediatric Cardiology, Helsinki University Hospital Helsinki, Finland
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku Turku, Finland
| | - Timo Savunen
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| | - Juha Koskenvuo
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku Turku, Finland
| |
Collapse
|
22
|
Role of thymosin beta 4 in hair growth. Mol Genet Genomics 2016; 291:1639-46. [DOI: 10.1007/s00438-016-1207-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 04/13/2016] [Indexed: 01/12/2023]
|
23
|
Ariza L, Carmona R, Cañete A, Cano E, Muñoz-Chápuli R. Coelomic epithelium-derived cells in visceral morphogenesis. Dev Dyn 2015; 245:307-22. [DOI: 10.1002/dvdy.24373] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Affiliation(s)
- Laura Ariza
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Rita Carmona
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Ana Cañete
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Elena Cano
- Integrative Vascular Biology Lab, Max Delbrück Center for Molecular Medicine; Robert-Rössle-Str. 10 13092, Berlin Germany
| | - Ramón Muñoz-Chápuli
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| |
Collapse
|
24
|
Lao DH, Esparza MC, Bremner SN, Banerjee I, Zhang J, Veevers J, Bradford WH, Gu Y, Dalton ND, Knowlton KU, Peterson KL, Lieber RL, Chen J. Lmo7 is dispensable for skeletal muscle and cardiac function. Am J Physiol Cell Physiol 2015; 309:C470-9. [PMID: 26157009 DOI: 10.1152/ajpcell.00177.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/06/2015] [Indexed: 11/22/2022]
Abstract
Emery-Dreifuss muscular dystrophy (EDMD) is a degenerative disease primarily affecting skeletal muscles in early childhood as well as cardiac muscle at later stages. EDMD is caused by a number of mutations in genes encoding proteins associated with the nuclear envelope (e.g., Emerin, Lamin A/C, and Nesprin). Recently, a novel protein, Lim-domain only 7 (lmo7) has been reported to play a role in the molecular pathogenesis of EDMD. Prior in vitro and in vivo studies suggested the intriguing possibility that Lmo7 plays a role in skeletal or cardiac muscle pathophysiology. To further understand the in vivo role of Lmo7 in striated muscles, we generated a novel Lmo7-null (lmo7(-/-)) mouse line. Using this mouse line, we examined skeletal and cardiac muscle physiology, as well as the role of Lmo7 in a model of muscular dystrophy and regeneration using the dystrophin-deficient mdx mouse model. Our results demonstrated that lmo7(-/-) mice had no abnormalities in skeletal muscle morphology, physiological function, or regeneration. Cardiac function was also unaffected. Moreover, we found that ablation of lmo7 in mdx mice had no effect on the observed myopathy and muscular regeneration exhibited by mdx mice. Molecular analyses also showed no changes in dystrophin complex factors, MAPK pathway components, and Emerin levels in lmo7 knockout mice. Taken together, we conclude that Lmo7 is dispensable for skeletal muscle and cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dieu Hung Lao
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Mary C Esparza
- University of California San Diego, Department of Orthopedic Surgery, La Jolla, California
| | - Shannon N Bremner
- University of California San Diego, Department of Orthopedic Surgery, La Jolla, California
| | - Indroneal Banerjee
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Jianlin Zhang
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Jennifer Veevers
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - William H Bradford
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Yusu Gu
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Nancy D Dalton
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Kirk U Knowlton
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Kirk L Peterson
- University of California San Diego, Department of Cardiology, La Jolla, California
| | - Richard L Lieber
- University of California San Diego, Department of Orthopedic Surgery, La Jolla, California; Rehabilitation Institute of Chicago, Chicago, Illinois
| | - Ju Chen
- University of California San Diego, Department of Cardiology, La Jolla, California;
| |
Collapse
|
25
|
Gao X, Liang H, Hou F, Zhang Z, Nuo M, Guo X, Liu D. Thymosin Beta-4 Induces Mouse Hair Growth. PLoS One 2015; 10:e0130040. [PMID: 26083021 PMCID: PMC4470810 DOI: 10.1371/journal.pone.0130040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/15/2015] [Indexed: 01/13/2023] Open
Abstract
Thymosin beta-4 (Tβ4) is known to induce hair growth and hair follicle (HF) development; however, its mechanism of action is unknown. We generated mice that overexpressed Tβ4 in the epidermis, as well as Tβ4 global knockout mice, to study the role of Tβ4 in HF development and explore the mechanism of Tβ4 on hair growth. To study Tβ4 function, we depilated control and experimental mice and made tissue sections stained with hematoxylin and eosin (H&E). To explore the effect of Tβ4 on hair growth and HF development, the mRNA and protein levels of Tβ4 and VEGF were detected by real-time PCR and western blotting in control and experimental mice. Protein expression levels and the phosphorylation of P38, ERK and AKT were also examined by western blotting. The results of depilation indicated that hair re-growth was faster in Tβ4-overexpressing mice, but slower in knockout mice. Histological examination revealed that Tβ4-overexpressing mice had a higher number of hair shafts and HFs clustered together to form groups, while the HFs of control mice and knockout mice were separate. Hair shafts in knockout mice were significantly reduced in number compared with control mice. Increased Tβ4 expression at the mRNA and protein levels was confirmed in Tβ4-overexpressing mice, which also had increased VEGF expression. On the other hand, knockout mice had reduced levels of VEGF expression. Mechanistically, Tβ4-overexpressing mice showed increased protein expression levels and phosphorylation of P38, ERK and AKT, whereas knockout mice had decreased levels of both expression and phosphorylation of these proteins. Tβ4 appears to regulate P38/ERK/AKT signaling via its effect on VEGF expression, with a resultant effect on the speed of hair growth, the pattern of HFs and the number of hair shafts.
Collapse
Affiliation(s)
- Xiaoyu Gao
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Hao Liang
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Fang Hou
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Zhipeng Zhang
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Mingtu Nuo
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Xudong Guo
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
- * E-mail: (DL); (XG)
| | - Dongjun Liu
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, China
- * E-mail: (DL); (XG)
| |
Collapse
|
26
|
Abstract
Coronary artery disease causes acute myocardial infarction and heart failure. Identifying coronary vascular progenitors and their developmental program could inspire novel regenerative treatments for cardiac diseases. The developmental origins of the coronary vessels have been shrouded in mystery and debated for several decades. Recent identification of progenitors for coronary vessels within the endocardium, epicardium, and sinus venosus provides new insights into this question. In addition, significant progress has been achieved in elucidating the cellular and molecular programs that orchestrate coronary artery development. Establishing adequate vascular supply will be an essential component of cardiac regenerative strategies, and these findings raise exciting new strategies for therapeutic cardiac revascularization.
Collapse
Affiliation(s)
- Xueying Tian
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - William T Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
27
|
Santhakumar R, Vidyasekar P, Verma RS. Cardiogel: a nano-matrix scaffold with potential application in cardiac regeneration using mesenchymal stem cells. PLoS One 2014; 9:e114697. [PMID: 25521816 PMCID: PMC4270637 DOI: 10.1371/journal.pone.0114697] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 01/05/2023] Open
Abstract
3-Dimensional conditions for the culture of Bone Marrow-derived Stromal/Stem Cells (BMSCs) can be generated with scaffolds of biological origin. Cardiogel, a cardiac fibroblast-derived Extracellular Matrix (ECM) has been previously shown to promote cardiomyogenic differentiation of BMSCs and provide protection against oxidative stress. To determine the matrix composition and identify significant proteins in cardiogel, we investigated the differences in the composition of this nanomatrix and a BMSC-derived ECM scaffold, termed as ‘mesogel’. An optimized protocol was developed that resulted in efficient decellularization while providing the maximum yield of ECM. The proteins were sequentially solubilized using acetic acid, Sodium Dodecyl Sulfate (SDS) and Dithiothreitol (DTT). These proteins were then analyzed using surfactant-assisted in-solution digestion followed by nano-liquid chromatography and tandem mass spectrometry (nLC-MS/MS). The results of these analyses revealed significant differences in their respective compositions and 17 significant ECM/matricellular proteins were differentially identified between cardiogel and mesogel. We observed that cardiogel also promoted cell proliferation, adhesion and migration while enhancing cardiomyogenic differentiation and angiogenesis. In conclusion, we developed a reproducible method for efficient extraction and solubilization of in vitro cultured cell-derived extracellular matrix. We report several important proteins differentially identified between cardiogel and mesogel, which can explain the biological properties of cardiogel. We also demonstrated the cardiomyogenic differentiation and angiogenic potential of cardiogel even in the absence of any external growth factors. The transplantation of Bone Marrow derived Stromal/Stem Cells (BMSCs) cultured on such a nanomatrix has potential applications in regenerative therapy for Myocardial Infarction (MI).
Collapse
Affiliation(s)
- Rajalakshmi Santhakumar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Prasanna Vidyasekar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
28
|
Michelis KC, Boehm M, Kovacic JC. New vessel formation in the context of cardiomyocyte regeneration--the role and importance of an adequate perfusing vasculature. Stem Cell Res 2014; 13:666-82. [PMID: 24841067 PMCID: PMC4213356 DOI: 10.1016/j.scr.2014.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023] Open
Abstract
The history of revascularization for cardiac ischemia dates back to the early 1960's when the first coronary artery bypass graft procedures were performed in humans. With this 50 year history of providing a new vasculature to ischemic and hibernating myocardium, a profound depth of experience has been amassed in clinical cardiovascular medicine as to what does, and does not work in the context of cardiac revascularization, alleviating ischemia and adequacy of myocardial perfusion. These issues are of central relevance to contemporary cell-based cardiac regenerative approaches. While the cardiovascular cell therapy field is surging forward on many exciting fronts, several well accepted clinical axioms related to the cardiac arterial supply appear to be almost overlooked by some of our current basic conceptual and experimental cell therapy paradigms. We present here information drawn from five decades of the clinical revascularization experience, review relevant new data on vascular formation via cell therapy, and put forward the case that for optimal cell-based cardiac regeneration due attention must be paid to providing an adequate vascular supply.
Collapse
Affiliation(s)
- Katherine C Michelis
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manfred Boehm
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Vieira JM, Riley PR. Chemical genetics and its potential in cardiac stem cell therapy. Br J Pharmacol 2014; 169:318-27. [PMID: 22385148 DOI: 10.1111/j.1476-5381.2012.01928.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Over the last decade or so, intensive research in cardiac stem cell biology has led to significant discoveries towards a potential therapy for cardiovascular disease; the main cause of morbidity and mortality in humans. The major goal within the field of cardiovascular regenerative medicine is to replace lost or damaged cardiac muscle and coronaries following ischaemic disease. At present, de novo cardiomyocytes can be generated either in vitro, for cell transplantation or disease modelling using directed differentiation of embryonic stem cells or induced pluripotent stem cells, or in vivo via direct reprogramming of resident adult cardiac fibroblast or ectopic stimulation of resident cardiac stem or progenitor cells. A major bottleneck with all of these approaches is the low efficiency of cardiomyocyte differentiation alongside their relative functional immaturity. Chemical genetics, and the application of phenotypic screening with small molecule libraries, represent a means to enhance understanding of the molecular pathways controlling cardiovascular cell differentiation and, moreover, offer the potential for discovery of new drugs to invoke heart repair and regeneration. Here, we review the potential of chemical genetics in cardiac stem cell therapy, highlighting not only the major contributions to the field so far, but also the future challenges.
Collapse
Affiliation(s)
- Joaquim M Vieira
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | |
Collapse
|
30
|
Chapman MA, Zhang J, Banerjee I, Guo LT, Zhang Z, Shelton GD, Ouyang K, Lieber RL, Chen J. Disruption of both nesprin 1 and desmin results in nuclear anchorage defects and fibrosis in skeletal muscle. Hum Mol Genet 2014; 23:5879-92. [PMID: 24943590 DOI: 10.1093/hmg/ddu310] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Proper localization and anchorage of nuclei within skeletal muscle is critical for cellular function. Alterations in nuclear anchoring proteins modify a number of cellular functions including mechanotransduction, nuclear localization, chromatin positioning/compaction and overall organ function. In skeletal muscle, nesprin 1 and desmin are thought to link the nucleus to the cytoskeletal network. Thus, we hypothesize that both of these factors play a key role in skeletal muscle function. To examine this question, we utilized global ablation murine models of nesprin 1, desmin or both nesprin 1 and desmin. Herein, we have created the nesprin-desmin double-knockout (DKO) mouse, eliminating a major fraction of nuclear-cytoskeletal connections and enabling understanding of the importance of nuclear anchorage in skeletal muscle. Globally, DKO mice are marked by decreased lifespan, body weight and muscle strength. With regard to skeletal muscle, DKO myonuclear anchorage was dramatically decreased compared with wild-type, nesprin 1(-/-) and desmin(-/-) mice. Additionally, nuclear-cytoskeletal strain transmission was decreased in DKO skeletal muscle. Finally, loss of nuclear anchorage in DKO mice coincided with a fibrotic response as indicated by increased collagen and extracellular matrix deposition and increased passive mechanical properties of muscle bundles. Overall, our data demonstrate that nesprin 1 and desmin serve redundant roles in nuclear anchorage and that the loss of nuclear anchorage in skeletal muscle results in a pathological response characterized by increased tissue fibrosis and mechanical stiffness.
Collapse
Affiliation(s)
| | | | | | - Ling T Guo
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Zhiwei Zhang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan 410011, P.R. China and
| | - G Diane Shelton
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Department of Medicine, School of Chemical Biology and Biotechnology, Peking University, Shenzhen 518055, P.R. China
| | - Richard L Lieber
- Department of Bioengineering and Department of Orthopaedic Surgery, University of California San Diego, and Department of Veteran's Affairs, 9500 Gilman Drive, La Jolla, CA 92093-0863, USA
| | | |
Collapse
|
31
|
Moore-Morris T, Guimarães-Camboa N, Banerjee I, Zambon AC, Kisseleva T, Velayoudon A, Stallcup WB, Gu Y, Dalton ND, Cedenilla M, Gomez-Amaro R, Zhou B, Brenner DA, Peterson KL, Chen J, Evans SM. Resident fibroblast lineages mediate pressure overload-induced cardiac fibrosis. J Clin Invest 2014; 124:2921-34. [PMID: 24937432 DOI: 10.1172/jci74783] [Citation(s) in RCA: 492] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 04/24/2014] [Indexed: 01/22/2023] Open
Abstract
Activation and accumulation of cardiac fibroblasts, which result in excessive extracellular matrix deposition and consequent mechanical stiffness, myocyte uncoupling, and ischemia, are key contributors to heart failure progression. Recently, endothelial-to-mesenchymal transition (EndoMT) and the recruitment of circulating hematopoietic progenitors to the heart have been reported to generate substantial numbers of cardiac fibroblasts in response to pressure overload-induced injury; therefore, these processes are widely considered to be promising therapeutic targets. Here, using multiple independent murine Cre lines and a collagen1a1-GFP fusion reporter, which specifically labels fibroblasts, we found that following pressure overload, fibroblasts were not derived from hematopoietic cells, EndoMT, or epicardial epithelial-to-mesenchymal transition. Instead, pressure overload promoted comparable proliferation and activation of two resident fibroblast lineages, including a previously described epicardial population and a population of endothelial origin. Together, these data present a paradigm for the origins of cardiac fibroblasts during development and in fibrosis. Furthermore, these data indicate that therapeutic strategies for reducing pathogenic cardiac fibroblasts should shift from targeting presumptive EndoMT or infiltrating hematopoietically derived fibroblasts, toward common pathways upregulated in two endogenous fibroblast populations.
Collapse
|
32
|
The epicardium signals the way towards heart regeneration. Stem Cell Res 2014; 13:683-92. [PMID: 24933704 PMCID: PMC4241487 DOI: 10.1016/j.scr.2014.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/12/2014] [Accepted: 04/18/2014] [Indexed: 11/23/2022] Open
Abstract
From historical studies of developing chick hearts to recent advances in regenerative injury models, the epicardium has arisen as a key player in heart genesis and repair. The epicardium provides paracrine signals to nurture growth of the developing heart from mid-gestation, and epicardium-derived cells act as progenitors of numerous cardiac cell types. Interference with either process is terminal for heart development and embryogenesis. In adulthood, the dormant epicardium reinstates an embryonic gene programme in response to injury. Furthermore, injury-induced epicardial signalling is essential for heart regeneration in zebrafish. Given these critical roles in development, injury response and heart regeneration, the application of epicardial signals following adult heart injury could offer therapeutic strategies for the treatment of ischaemic heart disease and heart failure. The epicardium is a dynamic signalling centre during heart development and injury. Heart repair in lower vertebrates highlights the importance of epicardial signalling. Epicardial signals may be targeted to regenerate adult mammalian hearts.
Collapse
|
33
|
Banerjee I, Zhang J, Moore-Morris T, Pfeiffer E, Buchholz KS, Liu A, Ouyang K, Stroud MJ, Gerace L, Evans SM, McCulloch A, Chen J. Targeted ablation of nesprin 1 and nesprin 2 from murine myocardium results in cardiomyopathy, altered nuclear morphology and inhibition of the biomechanical gene response. PLoS Genet 2014; 10:e1004114. [PMID: 24586179 PMCID: PMC3930490 DOI: 10.1371/journal.pgen.1004114] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 11/27/2013] [Indexed: 11/17/2022] Open
Abstract
Recent interest has focused on the importance of the nucleus and associated nucleoskeleton in regulating changes in cardiac gene expression in response to biomechanical load. Mutations in genes encoding proteins of the inner nuclear membrane and nucleoskeleton, which cause cardiomyopathy, also disrupt expression of a biomechanically responsive gene program. Furthermore, mutations in the outer nuclear membrane protein Nesprin 1 and 2 have been implicated in cardiomyopathy. Here, we identify for the first time a role for the outer nuclear membrane proteins, Nesprin 1 and Nesprin 2, in regulating gene expression in response to biomechanical load. Ablation of both Nesprin 1 and 2 in cardiomyocytes, but neither alone, resulted in early onset cardiomyopathy. Mutant cardiomyocytes exhibited altered nuclear positioning, shape, and chromatin positioning. Loss of Nesprin 1 or 2, or both, led to impairment of gene expression changes in response to biomechanical stimuli. These data suggest a model whereby biomechanical signals are communicated from proteins of the outer nuclear membrane, to the inner nuclear membrane and nucleoskeleton, to result in changes in gene expression required for adaptation of the cardiomyocyte to changes in biomechanical load, and give insights into etiologies underlying cardiomyopathy consequent to mutations in Nesprin 1 and 2.
Collapse
Affiliation(s)
- Indroneal Banerjee
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America
| | - Jianlin Zhang
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America
| | - Thomas Moore-Morris
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, California, United States of America
| | - Emily Pfeiffer
- Department of Bioengineering, University of California-San Diego, La Jolla, California, United States of America
| | - Kyle S Buchholz
- Department of Bioengineering, University of California-San Diego, La Jolla, California, United States of America
| | - Ao Liu
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America
| | - Kunfu Ouyang
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America ; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Matthew J Stroud
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, California, United States of America
| | - Andrew McCulloch
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America ; Department of Bioengineering, University of California-San Diego, La Jolla, California, United States of America
| | - Ju Chen
- Department of Medicine, University of California-San Diego, La Jolla, California, United States of America
| |
Collapse
|
34
|
Arno AI, Gauglitz GG, Barret JP, Jeschke MG. New molecular medicine-based scar management strategies. Burns 2014; 40:539-51. [PMID: 24438742 DOI: 10.1016/j.burns.2013.11.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/21/2013] [Accepted: 11/18/2013] [Indexed: 02/06/2023]
Abstract
Keloids and hypertrophic scars are prevalent disabling conditions with still suboptimal treatments. Basic science and molecular-based medicine research have contributed to unravel new bench-to-bedside scar therapies and to dissect the complex signalling pathways involved. Peptides such as the transforming growth factor beta (TGF-β) superfamily, with Smads, Ski, SnoN, Fussels, endoglin, DS-Sily, Cav-1p, AZX100, thymosin-β4 and other related molecules may emerge as targets to prevent and treat keloids and hypertrophic scars. The aim of this review is to describe the basic complexity of these new molecular scar management strategies and point out new fibrosis research lines.
Collapse
Affiliation(s)
- Anna I Arno
- Ross Tilley Burn Centre and Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Gerd G Gauglitz
- Department of Dermatology and Allergology, Ludwig Maximilians University, Munich, Germany
| | - Juan P Barret
- Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Marc G Jeschke
- Ross Tilley Burn Centre and Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Rengaraj D, Hwang YS, Liang XH, Deng WB, Yang ZM, Han JY. Comparative expression and regulation of TMSB4X in male reproductive tissues of rats and chickens. ACTA ACUST UNITED AC 2013; 319:584-95. [DOI: 10.1002/jez.1820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 07/29/2013] [Accepted: 08/05/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Deivendran Rengaraj
- World Class University (WCU) Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences; Seoul National University; Seoul South Korea
| | - Young Sun Hwang
- World Class University (WCU) Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences; Seoul National University; Seoul South Korea
| | | | - Wen Bo Deng
- Department of Biology; Shantou University; Shantou China
| | - Zeng Ming Yang
- Department of Biology; Shantou University; Shantou China
| | - Jae Yong Han
- World Class University (WCU) Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences; Seoul National University; Seoul South Korea
| |
Collapse
|
36
|
|
37
|
Microscopy ambient ionization top-down mass spectrometry reveals developmental patterning. Proc Natl Acad Sci U S A 2013; 110:14855-60. [PMID: 23969833 DOI: 10.1073/pnas.1310618110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
There is immense cellular and molecular heterogeneity in biological systems. Here, we demonstrate the utility of integrating an inverted light microscope with an ambient ionization source, nanospray electrospray desorption ionization, attached to a high-resolution mass spectrometer to characterize the molecular composition of mouse spinal cords. We detected a broad range of molecules, including peptides and proteins, as well as metabolites such as lipids, sugars, and other small molecules, including S-adenosyl methionine and glutathione, through top-down MS. Top-down analysis revealed variation in the expression of Hb, including the transition from fetal to adult Hb and heterogeneity in Hb subunits consistent with the genetic diversity of the mouse models. Similarly, temporal changes to actin-sequestering proteins β-thymosins during development were observed. These results demonstrate that interfacing microscopy with ambient ionization provides the means to perform targeted in situ ambient top-down mass spectral analysis to study the pattern of proteins, lipids, and sugars in biologically heterogeneous samples.
Collapse
|
38
|
Li T, Ma SY, Tang XC, Nie LY, Huang H. Production and characterization of highly purified recombinant thymosin beta 4 in Escherichia coli. Protein Expr Purif 2013; 90:90-5. [DOI: 10.1016/j.pep.2013.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/15/2013] [Accepted: 05/17/2013] [Indexed: 12/01/2022]
|
39
|
Circulation Research
Thematic Synopsis: Cardiovascular Development. Circ Res 2013. [DOI: 10.1161/circresaha.113.301305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Banerjee I, Moore Morris T, Evans SM, Chen J. Thymosin β4 is not required for embryonic viability or vascular development. Circ Res 2013; 112:e25-8. [PMID: 23371905 DOI: 10.1161/circresaha.111.300197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Rossdeutsch et al describe a requirement for thymosin β4 (Tβ4) in vascular development. Impaired mural cell migration, differentiation, partial embryonic lethality, and hemorrhaging were observed after analysis of 2 lines of mice, one of which was germline null for Tβ4 and another in which Tβ4 was knocked down by endothelial-specific expression of Tβ4 short hairpin RNA. These data are in direct contrast to our published global and cardiac-specific Tβ4-knockout lines. Thus, the role of Tβ4 needs to be clarified to understand its importance in cardiovascular development. OBJECTIVE To investigate and clarify the role of Tβ4 in vascular smooth muscle cell development and vessel stability. METHODS AND RESULTS Examination of Tβ4 global knockouts did not demonstrate embryonic hemorrhaging, altered mural cell development, or lethality. Endothelial-specific knockouts also did not exhibit any embryonic lethality and were viable to adulthood. CONCLUSIONS Analysis of our Tβ4 global and cardiac- and endothelial-specific knockout models demonstrated that Tβ4 is dispensable for embryonic viability and vascular development.
Collapse
Affiliation(s)
- Indroneal Banerjee
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
41
|
|
42
|
Xu B, Yang M, Li Z, Zhang Y, Jiang Z, Guan S, Jiang D. Thymosin β4 enhances the healing of medial collateral ligament injury in rat. ACTA ACUST UNITED AC 2013; 184:1-5. [PMID: 23523891 DOI: 10.1016/j.regpep.2013.03.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 01/29/2023]
Abstract
The role played by thymosin β4 (Tβ4) in the process of wound healing was reported in several organs. However, there have been no reports that investigated the role of Tβ4 in the repair process after ligament injury. The purpose of this study was to determine whether administration of Tβ4 would improve ligament repair following injury. The medial collateral ligament (MCL) was sharply transected on the day of surgery. Then, the treatment group received 100 μL of fibrin sealant containing 1 μg of Tβ4 placed in the ligament gap. Healing tissues were evaluated by hematoxylin and eosin stain, transmission electron microscopy, and biomechanical test at 4 weeks after surgery. Histologically, healing tissues in Tβ4-treated group exhibited uniform and evenly spaced fiber bundles. However, the collagen fibers were not evenly spaced in control rats. Moreover, diameters of collagen fibrils within granulation tissue from the Tβ4-treated rats were significantly increased. In Tβ4-treated MCLs, the mechanical properties of these healing tissues were significantly higher at 4 weeks after surgery. In terms of the mechanical properties of the healing femur-medial collateral ligament-tibia complexes, the Tβ4-treated group had significantly better biomechanical properties than the control group at 4 weeks after surgery. Local administration of Tβ4 promotes the healing process of MCL, both histologically and mechanically, in a rat model. These findings provide a basis for potential clinical use of Tβ4 in repairing ligaments.
Collapse
Affiliation(s)
- Bo Xu
- Department of Pediatric Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin China
| | | | | | | | | | | | | |
Collapse
|
43
|
Sribenja S, Wongkham S, Wongkham C, Yao Q, Chen C. Roles and Mechanisms of β-Thymosins in Cell Migration and Cancer Metastasis: An Update. Cancer Invest 2013; 31:103-10. [DOI: 10.3109/07357907.2012.756111] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Hinkel R, Trenkwalder T, Kupatt C. Molecular and cellular mechanisms of thymosin β4-mediated cardioprotection. Ann N Y Acad Sci 2013; 1269:102-9. [PMID: 23045977 DOI: 10.1111/j.1749-6632.2012.06693.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Coronary heart disease is still the leading cause of death in industrialized nations. Reduction of infarct size after acute myocardial infarction and, in addition, improvement of myocardial function and perfusion in acute and chronic myocardial ischemia would enhance cardiac survival. Thymosin β4, a 43-amino acid water-soluble peptide with pleiotropic abilities seems to be a promising candidate for the treatment of ischemic heart disease. During cardiac development, thymosin β4 is essential for vascularization of the myocardium, by targeting all three parts of vessel development, that is, vasculogenesis, angiogenesis, and arteriogenesis. In the adult, thymosin β4 is capable of inducing angiogenesis via activation of survival kinases in an actin-dependent and -independent manner. In addition, thymosin β4 has anti-inflammatory properties by reducing NF-κB p65 activation. These protective effects are further enhanced through increased myocyte and endothelial cell survival accompanied by differentiation of epicardial progenitor cells.
Collapse
Affiliation(s)
- Rabea Hinkel
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
| | | | | |
Collapse
|
45
|
Nakajima Y, Imanaka-Yoshida K. New insights into the developmental mechanisms of coronary vessels and epicardium. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 303:263-317. [PMID: 23445813 DOI: 10.1016/b978-0-12-407697-6.00007-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During heart development, the epicardium, which originates from the proepicardial organ (PE), is a source of coronary vessels. The PE develops from the posterior visceral mesoderm of the pericardial coelom after stimulation with a combination of weak bone morphogenetic protein and strong fibroblast growth factor (FGF) signaling. PE-derived cells migrate across the heart surface to form the epicardial sheet, which subsequently seeds multipotent subepicardial mesenchymal cells via epithelial-mesenchymal transition, which is regulated by several signaling pathways including retinoic acid, FGF, sonic hedgehog, Wnt, transforming growth factor-β, and platelet-derived growth factor. Subepicardial endothelial progenitors eventually generate the coronary vascular plexus, which acquires an arterial or venous phenotype, connects with the sinus venosus and aortic sinuses, and then matures through the recruitment of vascular smooth muscle cells under the regulation of complex growth factor signaling pathways. These developmental programs might be activated in the adult heart after injury and play a role in the regeneration/repair of the myocardium.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| | | |
Collapse
|
46
|
Stark C, Taimen P, Tarkia M, Pärkkä J, Saraste A, Alastalo TP, Savunen T, Koskenvuo J. Therapeutic potential of thymosin β4 in myocardial infarct and heart failure. Ann N Y Acad Sci 2012; 1269:117-24. [PMID: 23045979 DOI: 10.1111/j.1749-6632.2012.06695.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Thymosin β4 (Tβ4) is a peptide known for its abilities to protect and facilitate regeneration in a number of tissues following injury. Its cardioprotective effects have been evaluated in different animal models and, currently, a clinical trial is being planned in patients suffering from acute myocardial infarction. This paper focuses on the effects of Tβ4 on cardiac function in animal studies utilizing different imaging modalities for outcome measurements.
Collapse
Affiliation(s)
- Christoffer Stark
- Department of Surgery, Turku University Central Hospital, Turku, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gupta S, Kumar S, Sopko N, Qin Y, Wei C, Kim IK. Thymosin β4 and cardiac protection: implication in inflammation and fibrosis. Ann N Y Acad Sci 2012; 1269:84-91. [DOI: 10.1111/j.1749-6632.2012.06752.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
48
|
von Gise A, Pu WT. Endocardial and epicardial epithelial to mesenchymal transitions in heart development and disease. Circ Res 2012; 110:1628-45. [PMID: 22679138 DOI: 10.1161/circresaha.111.259960] [Citation(s) in RCA: 297] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epithelial to mesenchymal transition (EMT) converts epithelial cells to mobile and developmentally plastic mesenchymal cells. All cells in the heart arise from one or more EMTs. Endocardial and epicardial EMTs produce most of the noncardiomyocyte lineages of the mature heart. Endocardial EMT generates valve progenitor cells and is necessary for formation of the cardiac valves and for complete cardiac septation. Epicardial EMT is required for myocardial growth and coronary vessel formation, and it generates cardiac fibroblasts, vascular smooth muscle cells, a subset of coronary endothelial cells, and possibly a subset of cardiomyocytes. Emerging studies suggest that these developmental mechanisms are redeployed in adult heart valve disease, in cardiac fibrosis, and in myocardial responses to ischemic injury. Redirection and amplification of disease-related EMTs offer potential new therapeutic strategies and approaches for treatment of heart disease. Here, we review the role and molecular regulation of endocardial and epicardial EMT in fetal heart development, and we summarize key literature implicating reactivation of endocardial and epicardial EMT in adult heart disease.
Collapse
Affiliation(s)
- Alexander von Gise
- Department of Cardiology, Children's Hospital Boston, 300 Longwood Ave, Boston, MA 02115, USA
| | | |
Collapse
|
49
|
Rossdeutsch A, Smart N, Dubé KN, Turner M, Riley PR. Essential role for thymosin β4 in regulating vascular smooth muscle cell development and vessel wall stability. Circ Res 2012; 111:e89-102. [PMID: 22723298 DOI: 10.1161/circresaha.111.259846] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RATIONALE Compromised development of blood vessel walls leads to vascular instability that may predispose to aneurysm with risk of rupture and lethal hemorrhage. There is currently a lack of insight into developmental insults that may define the molecular and cellular characteristics of initiating and perpetrating factors in adult aneurismal disease. OBJECTIVE To investigate a role for the actin-binding protein thymosin β4 (Tβ4), previously shown to be proangiogenic, in mural cell development and vascular wall stability. METHODS AND RESULTS Phenotypic analyses of both global and endothelial-specific loss-of-function Tβ4 mouse models revealed a proportion of Tβ4-null embryos with vascular hemorrhage coincident with a reduction in smooth muscle cell coverage of their developing vessels. Mechanistic studies revealed that extracellular Tβ4 can stimulate differentiation of mesodermal progenitor cells to a mature mural cell phenotype through activation of the transforming growth factor-beta (TGFβ) pathway and that reduced TGFβ signaling correlates with the severity of hemorrhagic phenotype in Tβ4-null vasculature. CONCLUSIONS Tβ4 is a novel endothelial secreted trophic factor that functions synergistically with TGFβ to regulate mural cell development and vascular wall stability. These findings have important implications for understanding congenital anomalies that may be causative for adult-onset vascular instability.
Collapse
Affiliation(s)
- Alex Rossdeutsch
- Molecular Medicine Unit, UCL Institute of Child Health, UCL, London, UK
| | | | | | | | | |
Collapse
|
50
|
Kovacic JC, Mercader N, Torres M, Boehm M, Fuster V. Epithelial-to-mesenchymal and endothelial-to-mesenchymal transition: from cardiovascular development to disease. Circulation 2012; 125:1795-808. [PMID: 22492947 DOI: 10.1161/circulationaha.111.040352] [Citation(s) in RCA: 324] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|